1
|
Kates SL, Owen JR, Xie C, Ren Y, Muthukrishnan G, Schwarz EM. Vaccines: Do they have a role in orthopedic trauma? Injury 2024; 55 Suppl 6:111631. [PMID: 39482036 DOI: 10.1016/j.injury.2024.111631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 05/20/2024] [Accepted: 05/24/2024] [Indexed: 11/03/2024]
Abstract
Although vaccines have been hailed as one of the greatest advances in medicine based on their unparalleled cost-effectiveness in eradicating life-threatening infectious diseases, their role in orthopedic trauma-related infections is unclear. This is largely because vaccines are primarily made against pathogens that cause communicable diseases rather than opportunistic infections secondary to trauma, and most successful vaccines are against viruses rather than biofilm forming bacteria. Nonetheless, the tremendous costs to patients and healthcare systems warrant orthopedic trauma vaccine research, which has been a focal topic in recent international consensus meetings on musculoskeletal infection. This subject was also covered at the 2023 Osteosynthesis and Trauma Care Foundation (OTCF) meeting in Rome, Italy, and the purpose of this supplement article is to (1) highlight the osteoimmunology, animal models, translational research and clinical pilots that were discussed, (2) the proposed future directions that could lead to diagnostics and prognostics that are critically needed for evidence-based decision making, and (3) vaccines and passive-immunization strategies that could potentially be utilized to treat patients with orthopedic infections.
Collapse
Affiliation(s)
- Stephen L Kates
- Department of Orthopaedic Surgery, Virginia Commonwealth University, Richmond, VA, USA
| | - John R Owen
- Department of Orthopaedic Surgery, Virginia Commonwealth University, Richmond, VA, USA
| | - Chao Xie
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - Youliang Ren
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | | | - Edward M Schwarz
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
2
|
Wu W, Pang CNI, Mediati DG, Tree JJ. The functional small RNA interactome reveals targets for the vancomycin-responsive sRNA RsaOI in vancomycin-tolerant Staphylococcus aureus. mSystems 2024; 9:e0097123. [PMID: 38534138 PMCID: PMC11019875 DOI: 10.1128/msystems.00971-23] [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: 09/11/2023] [Accepted: 03/11/2024] [Indexed: 03/28/2024] Open
Abstract
Small RNAs have been found to control a broad range of bacterial phenotypes including tolerance to antibiotics. Vancomycin tolerance in multidrug resistance Staphylococcus aureus is correlated with dysregulation of small RNAs although their contribution to antibiotic tolerance is poorly understood. RNA-RNA interactome profiling techniques are expanding our understanding of sRNA-mRNA interactions in bacteria; however, determining the function of these interactions for hundreds of sRNA-mRNA pairs is a major challenge. At steady-state, protein and mRNA abundances are often highly correlated and lower than expected protein abundance may indicate translational repression of an mRNA. To identify sRNA-mRNA interactions that regulate mRNA translation, we examined the correlation between gene transcript abundance, ribosome occupancy, and protein levels. We used the machine learning technique self-organizing maps (SOMs) to cluster genes with similar transcription and translation patterns and identified a cluster of mRNAs that appeared to be post-transcriptionally repressed. By integrating our clustering with sRNA-mRNA interactome data generated in vancomycin-tolerant S. aureus by RNase III-CLASH, we identified sRNAs that may be mediating translational repression. We have confirmed sRNA-dependant post-transcriptional repression of several mRNAs in this cluster. Two of these interactions are mediated by RsaOI, a sRNA that is highly upregulated by vancomycin. We demonstrate the regulation of HPr and the cell-wall autolysin Atl. These findings suggest that RsaOI coordinates carbon metabolism and cell wall turnover during vancomycin treatment. IMPORTANCE The emergence of multidrug-resistant Staphylococcus aureus (MRSA) is a major public health concern. Current treatment is dependent on the efficacy of last-line antibiotics like vancomycin. The most common cause of vancomycin treatment failure is strains with intermediate resistance or tolerance that arise through the acqusition of a diverse repertoire of point mutations. These strains have been shown to altered small RNA (sRNA) expression in response to antibiotic treatment. Here, we have used a technique termed RNase III-CLASH to capture sRNA interactions with their target mRNAs. To understand the function of these interactions, we have looked at RNA and protein abundance for mRNAs targeted by sRNAs. Messenger RNA and protein levels are generally well correlated and we use deviations from this correlation to infer post-transcriptional regulation and the function of individual sRNA-mRNA interactions. Using this approach we identify mRNA targets of the vancomycin-induced sRNA, RsaOI, that are repressed at the translational level. We find that RsaOI represses the cell wall autolysis Atl and carbon transporter HPr suggestion a link between vancomycin treatment and suppression of cell wall turnover and carbon metabolism.
Collapse
Affiliation(s)
- Winton Wu
- School of Biotechnology and Biomolecular Sciences, Sydney, New South Wales, Australia
| | | | - Daniel G. Mediati
- School of Biotechnology and Biomolecular Sciences, Sydney, New South Wales, Australia
| | - Jai Justin Tree
- School of Biotechnology and Biomolecular Sciences, Sydney, New South Wales, Australia
| |
Collapse
|
3
|
Leonard AC, Goncheva MI, Gilbert SE, Shareefdeen H, Petrie LE, Thompson LK, Khursigara CM, Heinrichs DE, Cox G. Autolysin-mediated peptidoglycan hydrolysis is required for the surface display of Staphylococcus aureus cell wall-anchored proteins. Proc Natl Acad Sci U S A 2023; 120:e2301414120. [PMID: 36920922 PMCID: PMC10041135 DOI: 10.1073/pnas.2301414120] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 02/14/2023] [Indexed: 03/16/2023] Open
Abstract
Peptidoglycan hydrolases, or autolysins, play a critical role in cell wall remodeling and degradation, facilitating bacterial growth, cell division, and cell separation. In Staphylococcus aureus, the so-called "major" autolysin, Atl, has long been associated with host adhesion; however, the molecular basis underlying this phenomenon remains understudied. To investigate, we used the type V glycopeptide antibiotic complestatin, which binds to peptidoglycan and blocks the activity of autolysins, as a chemical probe of autolysin function. We also generated a chromosomally encoded, catalytically inactive variant of the Atl enzyme. Autolysin-mediated peptidoglycan hydrolysis, in particular Atl-mediated daughter cell separation, was shown to be critical for maintaining optimal surface levels of S. aureus cell wall-anchored proteins, including the fibronectin-binding proteins (FnBPs) and protein A (Spa). As such, disrupting autolysin function reduced the affinity of S. aureus for host cell ligands, and negatively impacted early stages of bacterial colonization in a systemic model of S. aureus infection. Phenotypic studies revealed that Spa was sequestered at the septum of complestatin-treated cells, highlighting that autolysins are required to liberate Spa during cell division. In summary, we reveal the hydrolytic activities of autolysins are associated with the surface display of S. aureus cell wall-anchored proteins. We demonstrate that by blocking autolysin function, type V glycopeptide antibiotics are promising antivirulence agents for the development of strategies to control S. aureus infections.
Collapse
Affiliation(s)
- Allison C. Leonard
- Department of Molecular and Cellular Biology, College of Biological Sciences, University of Guelph, GuelphONN1G 2W1, Canada
| | - Mariya I. Goncheva
- Department of Microbiology and Immunology, University of Western Ontario, LondonONN6A 5C1, Canada
| | - Stephanie E. Gilbert
- Department of Molecular and Cellular Biology, College of Biological Sciences, University of Guelph, GuelphONN1G 2W1, Canada
| | - Hiba Shareefdeen
- Department of Molecular and Cellular Biology, College of Biological Sciences, University of Guelph, GuelphONN1G 2W1, Canada
| | - Laurenne E. Petrie
- Department of Molecular and Cellular Biology, College of Biological Sciences, University of Guelph, GuelphONN1G 2W1, Canada
| | - Laura K. Thompson
- Department of Molecular and Cellular Biology, College of Biological Sciences, University of Guelph, GuelphONN1G 2W1, Canada
| | - Cezar M. Khursigara
- Department of Molecular and Cellular Biology, College of Biological Sciences, University of Guelph, GuelphONN1G 2W1, Canada
| | - David E. Heinrichs
- Department of Microbiology and Immunology, University of Western Ontario, LondonONN6A 5C1, Canada
| | - Georgina Cox
- Department of Molecular and Cellular Biology, College of Biological Sciences, University of Guelph, GuelphONN1G 2W1, Canada
| |
Collapse
|
4
|
Kutsuno S, Hayashi I, Yu L, Yamada S, Hisatsune J, Sugai M. Non-deacetylated poly- N-acetylglucosamine-hyperproducing Staphylococcus aureus undergoes immediate autoaggregation upon vortexing. Front Microbiol 2023; 13:1101545. [PMID: 36699608 PMCID: PMC9868172 DOI: 10.3389/fmicb.2022.1101545] [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: 11/18/2022] [Accepted: 12/14/2022] [Indexed: 01/11/2023] Open
Abstract
Biofilms are microbial communities of cells embedded in a matrix of extracellular polymeric substances generated and adhering to each other or to a surface. Cell aggregates formed in the absence of a surface and floating pellicles that form biofilms at the air-liquid interface are also considered to be a type of biofilm. Staphylococcus aureus is a well-known cause of biofilm infections and high-molecular-weight polysaccharides, poly-N-acetylglucosamine (PNAG) is a main constituent of the biofilm. An icaADBC operon comprises major machinery to synthesize and extracellularly secrete PNAG. Extracellular PNAG is partially deacetylated by IcaB deacetylase, and the positively charged PNAG hence interacts with negatively charged cell surface to form the major component of biofilm. We previously reported a new regulator of biofilm (Rob) and demonstrated that Rob binds to a unique 5-bp motif, TATTT, present in intergenic region between icaADBC operon and its repressor gene icaR in Yu et al. The deletion of the 5-bp motif induces excessive adherent biofilm formation. The real function of the 5-bp motif is still unknown. In an attempt to isolate the 5-bp motif deletion mutant, we isolated several non-adherent mutants. They grew normally in turbid broth shaking culture but immediately auto-aggregated upon weak vortexing and sedimented as a lump resulting in a clear supernatant. Whole genome sequencing of the mutants identified they all carried mutations in icaB in addition to deletion of the 5-bp motif. Purification and molecular characterization of auto-aggregating factor in the culture supernatant of the mutant identified that the factor was a massively produced non-deacetylated PNAG. Therefore, we created a double deficient strain of biofilm inhibitory factors (5-bp motif, icaR, rob) and icaB to confirm the aggregation phenomenon. This peculiar phenomenon was only observed in Δ5bpΔicaB double mutant but not in ΔicaR ΔicaB or ΔrobΔicaB mutant. This study explains large amount of extracellularly produced non-deacetylated PNAG by Δ5bpΔicaB double mutation induced rapid auto-aggregation of S. aureus cells by vortexing. This phenomenon indicated that Staphylococcus aureus may form biofilms that do not adhere to solid surfaces and we propose this as a new mechanism of non-adherent biofilm formation of S. aureus.
Collapse
Affiliation(s)
- Shoko Kutsuno
- Antimicrobial Resistance Research Center, National Institute of Infectious Diseases, Tokyo, Japan,Department of Antimicrobial Resistance, Hiroshima University Graduate School of Biomedical & Health Sciences, Hiroshima, Japan
| | - Ikue Hayashi
- Research Facility, Hiroshima University Faculty of Dentistry, Hiroshima, Japan
| | - Liansheng Yu
- Antimicrobial Resistance Research Center, National Institute of Infectious Diseases, Tokyo, Japan,Department of Antimicrobial Resistance, Hiroshima University Graduate School of Biomedical & Health Sciences, Hiroshima, Japan
| | - Sakuo Yamada
- Department of Medical Technology, Faculty of Health Sciences & Technology, Kawasaki University of Medical Welfare, Okayama, Japan
| | - Junzo Hisatsune
- Antimicrobial Resistance Research Center, National Institute of Infectious Diseases, Tokyo, Japan,Department of Antimicrobial Resistance, Hiroshima University Graduate School of Biomedical & Health Sciences, Hiroshima, Japan
| | - Motoyuki Sugai
- Antimicrobial Resistance Research Center, National Institute of Infectious Diseases, Tokyo, Japan,Department of Antimicrobial Resistance, Hiroshima University Graduate School of Biomedical & Health Sciences, Hiroshima, Japan,*Correspondence: Motoyuki Sugai,
| |
Collapse
|
5
|
Hemmadi V, Biswas M, Mohsin M, Bano R. Biochemical and biophysical analysis of the interaction of a recombinant form of Staphylococcus aureus enolase with plasminogen. Future Microbiol 2022; 17:1455-1473. [DOI: 10.2217/fmb-2022-0169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Aim: Pathogenic invasion of Staphylococcus aureus is critically dependent on host plasminogen activation. Materials & methods: The pathophysiological implications of the interactions between S. aureus recombinant enolase and host plasminogen were investigated. The effects of mutation and small synthetic peptide inhibitors on interactions were assessed. Results: In vitro, the S. aureus recombinant enolase exists as a catalytically active fragile octamer and a robust dimer. The dimer interacts with the host plasminogen on the S. aureus surface. Conclusion: The interaction of host plasminogen and S. aureus enolase might mediate bacterial adherence to the host, activate the plasminogen with the help of plasminogen activators and prevent α2-antiplasmin-mediated inhibition of plasmin. Incorporating mutant and synthetic peptides inhibited the interactions and their associated pathophysiological consequences.
Collapse
Affiliation(s)
- Vijay Hemmadi
- Department of Biological Sciences, Birla Institute of Technology & Science, Pilani, K. K. Birla Goa Campus, NH17B, Zuarinagar, Goa, 403726, India
| | - Malabika Biswas
- Department of Biological Sciences, Birla Institute of Technology & Science, Pilani, K. K. Birla Goa Campus, NH17B, Zuarinagar, Goa, 403726, India
| | - Mohd Mohsin
- Department of Biosciences, Metabolic Engineering Lab, Jamia Millia Islamia, New Delhi, 110025, India
| | - Reshma Bano
- Department of Biosciences, Metabolic Engineering Lab, Jamia Millia Islamia, New Delhi, 110025, India
| |
Collapse
|
6
|
The Discovery of the Role of Outer Membrane Vesicles against Bacteria. Biomedicines 2022; 10:biomedicines10102399. [PMID: 36289660 PMCID: PMC9598313 DOI: 10.3390/biomedicines10102399] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 09/02/2022] [Accepted: 09/22/2022] [Indexed: 11/23/2022] Open
Abstract
Gram-negative bacteria are intrinsically resistant to many commercialized antibiotics. The outer membrane (OM) of Gram-negative bacteria prevents the entry of such antibiotics. Outer membrane vesicles (OMV) are naturally released from the OM of Gram-negative bacteria for a range of purposes, including competition with other bacteria. OMV may carry, as part of the membrane or lumen, molecules with antibacterial activity. Such OMV can be exposed to and can fuse with the cell surface of different bacterial species. In this review we consider how OMV can be used as tools to deliver antimicrobial agents. This includes the characteristics of OMV production and how this process can be used to create the desired antibacterial activity of OMV.
Collapse
|
7
|
Sherchand SP, Adhikari RP, Muthukrishnan G, Kanipakala T, Owen JR, Xie C, Aman MJ, Proctor RA, Schwarz EM, Kates SL. Evidence of Neutralizing and Non-Neutralizing Anti-Glucosaminidase Antibodies in Patients With S. Aureus Osteomyelitis and Their Association With Clinical Outcome Following Surgery in a Clinical Pilot. Front Cell Infect Microbiol 2022; 12:876898. [PMID: 35923804 PMCID: PMC9339635 DOI: 10.3389/fcimb.2022.876898] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 06/20/2022] [Indexed: 01/07/2023] Open
Abstract
Staphylococcus aureus osteomyelitis remains a very challenging condition; recent clinical studies have shown infection control rates following surgery/antibiotics to be ~60%. Additionally, prior efforts to produce an effective S. aureus vaccine have failed, in part due to lack of knowledge of protective immunity. Previously, we demonstrated that anti-glucosaminidase (Gmd) antibodies are protective in animal models but found that only 6.7% of culture-confirmed S. aureus osteomyelitis patients in the AO Clinical Priority Program (AO-CPP) Registry had basal serum levels (>10 ng/ml) of anti-Gmd at the time of surgery (baseline). We identified a small subset of patients with high levels of anti-Gmd antibodies and adverse outcomes following surgery, not explained by Ig class switching to non-functional isotypes. Here, we aimed to test the hypothesis that clinical cure following surgery is associated with anti-Gmd neutralizing antibodies in serum. Therefore, we first optimized an in vitro assay that quantifies recombinant Gmd lysis of the M. luteus cell wall and used it to demonstrate the 50% neutralizing concentration (NC50) of a humanized anti-Gmd mAb (TPH-101) to be ~15.6 μg/ml. We also demonstrated that human serum deficient in anti-Gmd antibodies can be complemented by TPH-101 to achieve the same dose-dependent Gmd neutralizing activity as purified TPH-101. Finally, we assessed the anti-Gmd physical titer and neutralizing activity in sera from 11 patients in the AO-CPP Registry, who were characterized into four groups post-hoc. Group 1 patients (n=3) had high anti-Gmd physical and neutralizing titers at baseline that decreased with clinical cure of the infection over time. Group 2 patients (n=3) had undetectable anti-Gmd antibodies throughout the study and adverse outcomes. Group 3 (n=3) had high titers +/- neutralizing anti-Gmd at baseline with adverse outcomes. Group 4 (n=2) had low titers of non-neutralizing anti-Gmd at baseline with delayed high titers and adverse outcomes. Collectively, these findings demonstrate that both neutralizing and non-neutralizing anti-Gmd antibodies exist in S. aureus osteomyelitis patients and that screening for these antibodies could have a value for identifying patients in need of passive immunization prior to surgery. Future prospective studies to test the prognostic value of anti-Gmd antibodies to assess the potential of passive immunization with TPH-101 are warranted.
Collapse
Affiliation(s)
| | | | - Gowrishankar Muthukrishnan
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States
| | | | - John R. Owen
- Department of Orthopaedic Surgery, Virginia Commonwealth University, Richmond, VA, United States
| | - Chao Xie
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States
| | - M. Javad Aman
- Integrated BioTherapeutics, Inc., Rockville, MD, United States
| | - Richard A. Proctor
- Departments of Medical Microbiology/Immunology and Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Edward M. Schwarz
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, United States
| | - Stephen L. Kates
- Department of Orthopaedic Surgery, Virginia Commonwealth University, Richmond, VA, United States
| |
Collapse
|
8
|
Wang M, Buist G, van Dijl JM. Staphylococcus aureus cell wall maintenance - the multifaceted roles of peptidoglycan hydrolases in bacterial growth, fitness, and virulence. FEMS Microbiol Rev 2022; 46:6604383. [PMID: 35675307 PMCID: PMC9616470 DOI: 10.1093/femsre/fuac025] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 04/22/2022] [Accepted: 05/25/2022] [Indexed: 01/07/2023] Open
Abstract
Staphylococcus aureus is an important human and livestock pathogen that is well-protected against environmental insults by a thick cell wall. Accordingly, the wall is a major target of present-day antimicrobial therapy. Unfortunately, S. aureus has mastered the art of antimicrobial resistance, as underscored by the global spread of methicillin-resistant S. aureus (MRSA). The major cell wall component is peptidoglycan. Importantly, the peptidoglycan network is not only vital for cell wall function, but it also represents a bacterial Achilles' heel. In particular, this network is continuously opened by no less than 18 different peptidoglycan hydrolases (PGHs) encoded by the S. aureus core genome, which facilitate bacterial growth and division. This focuses attention on the specific functions executed by these enzymes, their subcellular localization, their control at the transcriptional and post-transcriptional levels, their contributions to staphylococcal virulence and their overall importance in bacterial homeostasis. As highlighted in the present review, our understanding of the different aspects of PGH function in S. aureus has been substantially increased over recent years. This is important because it opens up new possibilities to exploit PGHs as innovative targets for next-generation antimicrobials, passive or active immunization strategies, or even to engineer them into effective antimicrobial agents.
Collapse
Affiliation(s)
- Min Wang
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, PO Box 30001, 9700 RB Groningen, the Netherlands
| | | | - Jan Maarten van Dijl
- Corresponding author: Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, P.O. box 30001, HPC EB80, 9700 RB Groningen, the Netherlands, Tel. +31-50-3615187; Fax. +31-50-3619105; E-mail:
| |
Collapse
|
9
|
Haubrich BA, Nayyab S, Gallati M, Hernandez J, Williams C, Whitman A, Zimmerman T, Li Q, Chen Y, Zhou CZ, Basu A, Reid CW. Inhibition of Streptococcus pneumoniae growth by masarimycin. MICROBIOLOGY (READING, ENGLAND) 2022; 168. [PMID: 35467499 DOI: 10.1099/mic.0.001182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Despite renewed interest, development of chemical biology methods to study peptidoglycan metabolism has lagged in comparison to the glycobiology field in general. To address this, a panel of diamides were screened against the Gram-positive bacterium Streptococcus pneumoniae to identify inhibitors of bacterial growth. The screen identified the diamide masarimycin as a bacteriostatic inhibitor of S. pneumoniae growth with an MIC of 8 µM. The diamide inhibited detergent-induced autolysis in a concentration-dependent manner, indicating perturbation of peptidoglycan degradation as the mode-of-action. Cell based screening of masarimycin against a panel of autolysin mutants, identified a higher MIC against a ΔlytB strain lacking an endo-N-acetylglucosaminidase involved in cell division. Subsequent biochemical and phenotypic analyses suggested that the higher MIC was due to an indirect interaction with LytB. Further analysis of changes to the cell surface in masarimycin treated cells identified the overexpression of several moonlighting proteins, including elongation factor Tu which is implicated in regulating cell shape. Checkerboard assays using masarimycin in concert with additional antibiotics identified an antagonistic relationship with the cell wall targeting antibiotic fosfomycin, which further supports a cell wall mode-of-action.
Collapse
Affiliation(s)
- Brad A Haubrich
- Center for Health and Behavioral Sciences, Department of Science and Technology, Bryant University, 1150 Douglas Pike, Smithfield, RI 02917, USA.,Department of Basic Sciences, Touro University Nevada, College of Osteopathic Medicine, Henderson, NV 89014, USA
| | - Saman Nayyab
- Center for Health and Behavioral Sciences, Department of Science and Technology, Bryant University, 1150 Douglas Pike, Smithfield, RI 02917, USA.,Amherst Department of Molecular and Cellular Biology, University of Massachusetts, 230 Stockbridge Rd Amherst, MA, USA
| | - Mika Gallati
- Center for Health and Behavioral Sciences, Department of Science and Technology, Bryant University, 1150 Douglas Pike, Smithfield, RI 02917, USA
| | - Jazmeen Hernandez
- Center for Health and Behavioral Sciences, Department of Science and Technology, Bryant University, 1150 Douglas Pike, Smithfield, RI 02917, USA
| | - Caroline Williams
- Center for Health and Behavioral Sciences, Department of Science and Technology, Bryant University, 1150 Douglas Pike, Smithfield, RI 02917, USA
| | - Andrew Whitman
- Center for Health and Behavioral Sciences, Department of Science and Technology, Bryant University, 1150 Douglas Pike, Smithfield, RI 02917, USA
| | - Tahl Zimmerman
- Department of Family and Consumer Sciences, North Carolina A&T State University, Greensboro, NC, USA
| | - Qiong Li
- School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, 230027, PR China
| | - Yuxing Chen
- School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, 230027, PR China
| | - Cong-Zhao Zhou
- School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, 230027, PR China
| | - Amit Basu
- Department of Chemistry, Brown University, Providence, RI, USA
| | - Christopher W Reid
- Center for Health and Behavioral Sciences, Department of Science and Technology, Bryant University, 1150 Douglas Pike, Smithfield, RI 02917, USA
| |
Collapse
|
10
|
Dörr T. Understanding tolerance to cell wall-active antibiotics. Ann N Y Acad Sci 2021; 1496:35-58. [PMID: 33274447 PMCID: PMC8359209 DOI: 10.1111/nyas.14541] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 11/02/2020] [Accepted: 11/04/2020] [Indexed: 12/19/2022]
Abstract
Antibiotic tolerance-the ability of bacteria to survive for an extended time in the presence of bactericidal antibiotics-is an understudied contributor to antibiotic treatment failure. Herein, I review the manifestations, mechanisms, and clinical relevance of tolerance to cell wall-active (CWA) antibiotics, one of the most important groups of antibiotics at the forefront of clinical use. I discuss definitions of tolerance and assays for tolerance detection, comprehensively discuss the mechanism of action of β-lactams and other CWA antibiotics, and then provide an overview of how cells mitigate the potentially lethal effects of CWA antibiotic-induced cell damage to become tolerant. Lastly, I discuss evidence for a role of CWA antibiotic tolerance in clinical antibiotic treatment failure.
Collapse
Affiliation(s)
- Tobias Dörr
- Weill Institute for Cell and Molecular Biology, Department of Microbiology, and Cornell Institute of Host–Pathogen Interactions and DiseaseCornell UniversityIthacaNew York
| |
Collapse
|
11
|
Scaffidi SJ, Shebes MA, Yu W. Tracking the Subcellular Localization of Surface Proteins in Staphylococcus aureus by Immunofluorescence Microscopy. Bio Protoc 2021; 11:e4038. [PMID: 34150940 DOI: 10.21769/bioprotoc.4038] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 03/03/2021] [Accepted: 04/27/2021] [Indexed: 11/02/2022] Open
Abstract
Surface proteins of Staphylococcus aureus and other Gram-positive bacteria play essential roles in bacterial colonization and host-microbe interactions. Surface protein precursors containing a YSIRK/GXXS signal peptide are translocated across the septal membrane at mid-cell, anchored to the cell wall peptidoglycan at the cross-wall compartment, and presented on the new hemispheres of the daughter cells following cell division. After several generations of cell division, these surface proteins will eventually cover the entire cell surface. To understand how these proteins travel from the bacterial cytoplasm to the cell surface, we describe a series of immunofluorescence microscopy protocols designed to detect the stepwise subcellular localization of the surface protein precursors: surface display (protocol A), cross-wall localization (protocol B), and cytoplasmic/septal membrane localization (protocol C). Staphylococcal protein A (SpA) is the model protein used in this work. The protocols described here are readily adapted to study the localization of other surface proteins as well as other cytoplasmic or membrane proteins in S. aureus in general. Furthermore, the protocols can be modified and adapted for use in other Gram-positive bacteria. Graphic abstract: Tracking the subcellular localization of surface proteins in S. aureus.
Collapse
Affiliation(s)
- Salvatore J Scaffidi
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, Tampa, FL, USA
| | - Mac A Shebes
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, Tampa, FL, USA
| | - Wenqi Yu
- Department of Cell Biology, Microbiology and Molecular Biology, University of South Florida, Tampa, FL, USA
| |
Collapse
|
12
|
Somarathne RP, Chappell ER, Perera YR, Yadav R, Park JY, Fitzkee NC. Understanding How Staphylococcal Autolysin Domains Interact With Polystyrene Surfaces. Front Microbiol 2021; 12:658373. [PMID: 34093472 PMCID: PMC8170090 DOI: 10.3389/fmicb.2021.658373] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 04/19/2021] [Indexed: 01/04/2023] Open
Abstract
Biofilms, when formed on medical devices, can cause malfunctions and reduce the efficiency of these devices, thus complicating treatments and serving as a source of infection. The autolysin protein of Staphylococcus epidermidis contributes to its biofilm forming ability, especially on polystyrene surfaces. R2ab and amidase are autolysin protein domains thought to have high affinity to polystyrene surfaces, and they are involved in initial bacterial attachment in S. epidermidis biofilm formation. However, the structural details of R2ab and amidase binding to surfaces are poorly understood. In this study, we have investigated how R2ab and amidase influence biofilm formation on polystyrene surfaces. We have also studied how these proteins interact with polystyrene nanoparticles (PSNPs) using biophysical techniques. Pretreating polystyrene plates with R2ab and amidase domains inhibits biofilm growth relative to a control protein, indicating that these domains bind tightly to polystyrene surfaces and can block bacterial attachment. Correspondingly, we find that both domains interact strongly with anionic, carboxylate-functionalized as well as neutral, non-functionalized PSNPs, suggesting a similar binding interaction for nanoparticles and macroscopic surfaces. Both anionic and neutral PSNPs induce changes to the secondary structure of both R2ab and amidase as monitored by circular dichroism (CD) spectroscopy. These changes are very similar, though not identical, for both types of PSNPs, suggesting that carboxylate functionalization is only a small perturbation for R2ab and amidase binding. This structural change is also seen in limited proteolysis experiments, which exhibit substantial differences for both proteins when in the presence of carboxylate PSNPs. Overall, our results demonstrate that the R2ab and amidase domains strongly favor adsorption to polystyrene surfaces, and that surface adsorption destabilizes the secondary structure of these domains. Bacterial attachment to polystyrene surfaces during the initial phases of biofilm formation, therefore, may be mediated by aromatic residues, since these residues are known to drive adsorption to PSNPs. Together, these experiments can be used to develop new strategies for biofilm eradication, ensuring the proper long-lived functioning of medical devices.
Collapse
Affiliation(s)
- Radha P. Somarathne
- Department of Chemistry, Mississippi State University, Mississippi State, MS, United States
| | - Emily R. Chappell
- Department of Chemistry, Mississippi State University, Mississippi State, MS, United States
| | - Y. Randika Perera
- Department of Biochemistry, Chemistry, and Center for Structural Biology, Vanderbilt University, Nashville, TN, United States
| | - Rahul Yadav
- Department of Chemistry, Mississippi State University, Mississippi State, MS, United States
| | - Joo Youn Park
- Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Mississippi State, MS, United States
| | - Nicholas C. Fitzkee
- Department of Chemistry, Mississippi State University, Mississippi State, MS, United States
| |
Collapse
|
13
|
Buchad H, Nair M. The small RNA SprX regulates the autolysin regulator WalR in Staphylococcus aureus. Microbiol Res 2021; 250:126785. [PMID: 34000511 DOI: 10.1016/j.micres.2021.126785] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 04/29/2021] [Accepted: 04/30/2021] [Indexed: 10/21/2022]
Abstract
Pathogenesis of Staphylococcus aureus is attributed to its remarkable adaptation to changes in the environment, mediated by the arsenal of virulence factors, which are regulated by intricate mechanisms that include small RNAs (sRNAs) as important regulatory molecules. The sRNA SprX was previously described to be involved in the regulation of S. aureus pathogenicity, by modifying the expression of surface-associated clumping factor B and the secreted delta haemolysin. This study describes the regulation by SprX, of expression of multiple autolysins, which play an essential role in cell wall metabolism and function as important virulence factors that facilitate adhesion, internalization, and immune evasion during S. aureus colonization and pathogenesis. SprX acts by positively regulating the expression of autolysin regulator WalR. Overexpression of SprX resulted in differential regulation of autolysins IsaA, and LytM, while WalR levels were unchanged. SprX knockdown strain exhibited down-regulation of multiple autolytic bands corresponding to the major autolysin AtlA and its process intermediates in cell wall degradation zymography, and 0.2 to 0.1 fold reduction of lytM, atlA, isaA, and walR transcripts in qRT-PCRs. Down-regulation of SprX resulted in altered phenotype with high cell aggregation as analyzed by SEM, decrease in biofilm formation and higher resistance to Triton X-100-induced lysis, all of which indicate that SprX is essential for expression of autolysins. A putative RNA-RNA interaction was indicated in silico between SprX and walR mRNA and further confirmed by in vitro RNA-RNA interaction in electrophoretic mobility shift assays. These findings elucidate a new mechanism in which SprX modulates the S. aureus pathogenicity by regulating the regulator of autolysins in cell wall metabolism and as virulence factors.
Collapse
Affiliation(s)
- Hasmatbanu Buchad
- Department of Microbiology and Biotechnology Centre, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, 390002, India.
| | - Mrinalini Nair
- Department of Microbiology and Biotechnology Centre, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, 390002, India.
| |
Collapse
|
14
|
Shi M, Willing SE, Kim HK, Schneewind O, Missiakas D. Peptidoglycan Contribution to the B Cell Superantigen Activity of Staphylococcal Protein A. mBio 2021; 12:e00039-21. [PMID: 33879590 PMCID: PMC8092194 DOI: 10.1128/mbio.00039-21] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Accepted: 03/17/2021] [Indexed: 11/20/2022] Open
Abstract
Staphylococcus aureus causes reiterative and chronic persistent infections. This can be explained by the formidable ability of this pathogen to escape immune surveillance mechanisms. Cells of S. aureus display the abundant staphylococcal protein A (SpA). SpA binds to immunoglobulin (Ig) molecules and coats the bacterial surface to prevent phagocytic uptake. SpA also binds and cross-links variable heavy 3 (VH3) idiotype (IgM) B cell receptors, promoting B cell expansion and the secretion of nonspecific VH3-IgM via a mechanism requiring CD4+ T cell help. SpA binding to antibodies is mediated by the N-terminal Ig-binding domains (IgBDs). The so-called region X, uncharacterized LysM domain, and C-terminal LPXTG sorting signal for peptidoglycan attachment complete the linear structure of the protein. Here, we report that both the LysM domain and the LPXTG motif sorting signal are required for the B cell superantigen activity of SpA in a mouse model of infection. SpA molecules purified from staphylococcal cultures are sufficient to exert B cell superantigen activity and promote immunoglobulin secretion as long as they carry intact LysM and LPXTG motif domains with bound peptidoglycan fragments. The LysM domain binds the glycan chains of peptidoglycan fragments, whereas the LPXTG motif is covalently linked to wall peptides lacking glycan. These findings emphasize the complexity of SpA interactions with B cell receptors.IMPORTANCE The LysM domain is found in all kingdoms of life. While their function in mammals is not known, LysM domains of bacteria and their phage parasites are associated with enzymes that cleave or remodel peptidoglycan. Plants recognize microbe-associated molecular patterns such as chitin via receptors endowed with LysM-containing ectodomains. In plants, such receptors play equally important roles in defense and symbiosis signaling. SpA of S. aureus carries a LysM domain that binds glycan strands of peptidoglycan to influence defined B cell responses that divert pathogen-specific adaptive immune responses.
Collapse
Affiliation(s)
- Miaomiao Shi
- Howard Taylor Ricketts Laboratory, Argonne National Laboratory, Lemont, Illinois, USA
| | | | - Hwan Keun Kim
- Department of Microbiology, University of Chicago, Chicago, Illinois, USA
| | - Olaf Schneewind
- Department of Microbiology, University of Chicago, Chicago, Illinois, USA
| | - Dominique Missiakas
- Howard Taylor Ricketts Laboratory, Argonne National Laboratory, Lemont, Illinois, USA
- Department of Microbiology, University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
15
|
Fisher JF, Mobashery S. β-Lactams against the Fortress of the Gram-Positive Staphylococcus aureus Bacterium. Chem Rev 2021; 121:3412-3463. [PMID: 33373523 PMCID: PMC8653850 DOI: 10.1021/acs.chemrev.0c01010] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The biological diversity of the unicellular bacteria-whether assessed by shape, food, metabolism, or ecological niche-surely rivals (if not exceeds) that of the multicellular eukaryotes. The relationship between bacteria whose ecological niche is the eukaryote, and the eukaryote, is often symbiosis or stasis. Some bacteria, however, seek advantage in this relationship. One of the most successful-to the disadvantage of the eukaryote-is the small (less than 1 μm diameter) and nearly spherical Staphylococcus aureus bacterium. For decades, successful clinical control of its infection has been accomplished using β-lactam antibiotics such as the penicillins and the cephalosporins. Over these same decades S. aureus has perfected resistance mechanisms against these antibiotics, which are then countered by new generations of β-lactam structure. This review addresses the current breadth of biochemical and microbiological efforts to preserve the future of the β-lactam antibiotics through a better understanding of how S. aureus protects the enzyme targets of the β-lactams, the penicillin-binding proteins. The penicillin-binding proteins are essential enzyme catalysts for the biosynthesis of the cell wall, and understanding how this cell wall is integrated into the protective cell envelope of the bacterium may identify new antibacterials and new adjuvants that preserve the efficacy of the β-lactams.
Collapse
Affiliation(s)
- Jed F Fisher
- Department of Chemistry and Biochemistry, McCourtney Hall, University of Notre Dame, Notre Dame Indiana 46556, United States
| | - Shahriar Mobashery
- Department of Chemistry and Biochemistry, McCourtney Hall, University of Notre Dame, Notre Dame Indiana 46556, United States
| |
Collapse
|
16
|
New insights in the coordinated amidase and glucosaminidase activity of the major autolysin (Atl) in Staphylococcus aureus. Commun Biol 2020; 3:695. [PMID: 33219282 PMCID: PMC7679415 DOI: 10.1038/s42003-020-01405-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 10/15/2020] [Indexed: 11/17/2022] Open
Abstract
After bacterial cell division, the daughter cells are still covalently interlinked by the peptidoglycan network which is resolved by specific hydrolases (autolysins) to release the daughter cells. In staphylococci, the major autolysin (Atl) with its two domain enzymes, N-acetylmuramyl-L-alanine amidase (AmiA) and β-N-acetylglucosaminidase (GlcA), resolves the peptidoglycan to release the daughter cells. Internal deletions in each of the enzyme domains revealed defined morphological alterations such as cell cluster formation in ΔamiA, ΔglcA and Δatl, and asymmetric cell division in the ΔglcA. A most important finding was that GlcA activity requires the prior removal of the stem peptide by AmiA for its activity thus the naked glycan strand is its substrate. Furthermore, GlcA is not an endo-β-N-acetylglucosaminidase but an exo-enzyme that cuts the glycan backbone to disaccharides independent of its O-acetylation modification. Our results shed new light into the sequential peptidoglycan hydrolysis by AmiA and GlcA during cell division in staphylococci. Nega et al. shed light on the interplay of the two domain enzymes of the major autolysin, AmiA and GlcA, in S. aureus for peptidoglycan hydrolysis during bacterial cell division. They show that GlcA requires the prior removal of the stem peptide by AmiA for its activity and that GlcA is not an endo-enzyme as previously thought, but an exo-enzyme that chops down the glycan backbone to disaccharides independent of its O-acetylation modification.
Collapse
|
17
|
Kates SL, Owen JR, Beck CA, Xie C, Muthukrishnan G, Daiss JL, Schwarz EM. Lack of Humoral Immunity Against Glucosaminidase Is Associated with Postoperative Complications in Staphylococcus aureus Osteomyelitis. J Bone Joint Surg Am 2020; 102:1842-1848. [PMID: 32858560 PMCID: PMC9018051 DOI: 10.2106/jbjs.20.00029] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND Glucosaminidase (Gmd) is known to be a protective antigen in animal models of Staphylococcus aureus osteomyelitis. We compared the endogenous anti-Gmd antibody levels in sera of patients with culture-confirmed S. aureus bone infections to their sera at 1 year after operative treatment of the infection. METHODS A novel global biospecimen registry of 297 patients with deep-wound culture-confirmed S. aureus osteomyelitis was analyzed to assess relationships between baseline anti-Gmd serum titers (via custom Luminex assay), known host risk factors for infection, and 1-year postoperative clinical outcomes (e.g., infection control, inconclusive, refracture, persistent infection, septic nonunion, amputation, and septic death). RESULTS All patients had measurable humoral immunity against some S. aureus antigens, but only 20 patients (6.7%; p < 0.0001) had high levels of anti-Gmd antibodies (>10 ng/mL) in serum at baseline. A subset of 194 patients (65.3%) who completed 1 year of follow-up was divided into groups based on anti-Gmd level: low (<1 ng/mL, 54 patients; 27.8%), intermediate (<10 ng/mL, 122 patients; 62.9%), and high (>10 ng/mL, 18 patients; 9.3%), and infection control rates were 40.7%, 50.0%, and 66.7%, respectively. The incidence of adverse outcomes in these groups was 33.3%, 16.4%, and 11.1%, respectively. Assessing anti-Gmd level as a continuous variable showed a 60% reduction in adverse-event odds (p = 0.04) for every tenfold increase in concentration. No differences in patient demographics, body mass index of >40 kg/m, diabetes status, age of ≥70 years, male sex, Charlson Comorbidity Index of >1, or Cierny-Mader host type were observed between groups, and these risk factors were not associated with adverse events. Patients with low anti-Gmd titer demonstrated a significant 2.68-fold increased odds of adverse outcomes (p = 0.008). CONCLUSIONS Deficiency in circulating anti-Gmd antibodies was associated serious adverse outcomes following operative treatment of S. aureus osteomyelitis. At 1 year, high levels of anti-Gmd antibodies were associated with a nearly 3-fold increase in infection-control odds. Additional prospective studies clarifying Gmd immunization for osteomyelitis are needed. LEVEL OF EVIDENCE Prognostic Level IV. See Instructions for Authors for a complete description of levels of evidence.
Collapse
Affiliation(s)
- Stephen L. Kates
- Department of Orthopaedic Surgery, Virginia Commonwealth University, Richmond, Virginia
| | - John R. Owen
- Department of Orthopaedic Surgery, Virginia Commonwealth University, Richmond, Virginia
| | - Christopher A. Beck
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York
| | - Chao Xie
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York
| | | | - John L. Daiss
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York
| | - Edward M. Schwarz
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York
| |
Collapse
|
18
|
Hemmadi V, Biswas M. An overview of moonlighting proteins in Staphylococcus aureus infection. Arch Microbiol 2020; 203:481-498. [PMID: 33048189 PMCID: PMC7551524 DOI: 10.1007/s00203-020-02071-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 09/29/2020] [Accepted: 10/01/2020] [Indexed: 01/01/2023]
Abstract
Staphylococcus aureus is responsible for numerous instances of superficial, toxin-mediated, and invasive infections. The emergence of methicillin-resistant (MRSA), as well as vancomycin-resistant (VRSA) strains of S. aureus, poses a massive threat to human health. The tenacity of S. aureus to acquire resistance against numerous antibiotics in a very short duration makes the effort towards developing new antibiotics almost futile. S. aureus owes its destructive pathogenicity to the plethora of virulent factors it produces among which a majority of them are moonlighting proteins. Moonlighting proteins are the multifunctional proteins in which a single protein, with different oligomeric conformations, perform multiple independent functions in different cell compartments. Peculiarly, proteins involved in key ancestral functions and metabolic pathways typically exhibit moonlighting functions. Pathogens mainly employ those proteins as virulent factors which exhibit high structural conservation towards their host counterparts. Consequentially, the host immune system counteracts these invading bacterial virulent factors with minimal protective action. Additionally, many moonlighting proteins also play multiple roles in various stages of pathogenicity while augmenting the virulence of the bacterium. This has necessitated elaborative studies to be conducted on moonlighting proteins of S. aureus that can serve as drug targets. This review is a small effort towards understanding the role of various moonlighting proteins in the pathogenicity of S. aureus.
Collapse
Affiliation(s)
- Vijay Hemmadi
- Department of Biological Sciences, Birla Institute of Technology and Science, BITS-Pilani, K. K. Birla Goa Campus, NH17B, Zuarinagar, Goa, 403726, India
| | - Malabika Biswas
- Department of Biological Sciences, Birla Institute of Technology and Science, BITS-Pilani, K. K. Birla Goa Campus, NH17B, Zuarinagar, Goa, 403726, India.
| |
Collapse
|
19
|
Kamohara A, Hirata H, Xu X, Shiraki M, Yamada S, Zhang JQ, Kukita T, Toyonaga K, Hara H, Urano Y, Yamashita Y, Miyamoto H, Kukita A. IgG immune complexes with Staphylococcus aureus protein A enhance osteoclast differentiation and bone resorption by stimulating Fc receptors and TLR2. Int Immunol 2020; 32:89-104. [PMID: 31713625 DOI: 10.1093/intimm/dxz063] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 09/26/2019] [Indexed: 01/18/2023] Open
Abstract
Staphylococcus aureus is a main pathogen of osteomyelitis and protein A is a virulence factor with high affinity for IgG. In this study, we investigated whether S. aureus affects the differentiation and bone resorption of osteoclasts through the IgG-binding capacity of protein A. Staphylococcus aureus pre-treated with serum or IgG showed marked enhancement in osteoclastogenesis and bone resorption compared to non-treated S. aureus or a protein A-deficient mutant. Blocking of the Fc receptor and deletion of the Fcγ receptor gene in osteoclast precursor cells showed that enhanced osteoclastogenesis stimulated by S. aureus IgG immune complexes (ICs) was mediated by the Fc receptor on osteoclast precursor cells. In addition, osteoclastogenesis stimulated by S. aureus ICs but not the protein A-deficient mutant was markedly reduced in osteoclast precursor cells of Myd88-knockout mice. Moreover, NFATc1, Syk and NF-κB signals were necessary for osteoclastogenesis stimulated by S. aureus ICs. The results suggest the contribution of a of Toll-like receptor 2 (TLR2)-Myd88 signal to the activity of S. aureus ICs. We further examined the expression of pro-inflammatory cytokines that is known to be enhanced by FcγR-TLR cross-talk. Osteoclasts induced by S. aureus ICs showed higher expression of TNF-α and IL-1β, and marked stimulation of proton secretion of osteoclasts activated by pro-inflammatory cytokines. Finally, injection of S. aureus, but not the protein A-deficient mutant, exacerbated bone loss in implantation and intra-peritoneal administration mouse models. Our results provide a novel mechanistic aspect of bone loss induced by S. aureus in which ICs and both Fc receptors and TLR pathways are involved.
Collapse
Affiliation(s)
- Asana Kamohara
- Department of Pathology and Microbiology, Saga, Japan.,Department of Oral & Maxillofacial Surgery, Saga, Japan
| | - Hirohito Hirata
- Department of Pathology and Microbiology, Saga, Japan.,Department of Orthopedic Surgery, Faculty of Medicine, Saga University, Saga, Japan
| | - Xianghe Xu
- Department of Pathology and Microbiology, Saga, Japan.,Department of Molecular Cell Biology & Oral Anatomy, Faculty of Dentistry, Kyushu University, Fukuoka, Japan
| | - Makoto Shiraki
- Department of Pathology and Microbiology, Saga, Japan.,Department of Orthopedic Surgery, Faculty of Medicine, Saga University, Saga, Japan
| | - Sakuo Yamada
- Department of Medical Technology, Department of Clinical Nutrition, Faculty of Health Science & Technology, Kawasaki University of Medical Welfare, Kurashiki, Okayama, Japan
| | - Jing-Qi Zhang
- Department of Orthopedic Surgery, Faculty of Medicine, Saga University, Saga, Japan
| | - Toshio Kukita
- Department of Molecular Cell Biology & Oral Anatomy, Faculty of Dentistry, Kyushu University, Fukuoka, Japan
| | - Kenji Toyonaga
- Department of Immunology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Hiromitsu Hara
- Department of Immunology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Yasuteru Urano
- Department of Chemical Biology & Molecular Imaging, Graduate School of Medicine , Hongo, Tokyo, Japan.,Department of Chemistry & Biology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Hongo, Tokyo, Japan
| | | | | | - Akiko Kukita
- Department of Pathology and Microbiology, Saga, Japan
| |
Collapse
|
20
|
Reassessment of the distinctive geometry of Staphylococcus aureus cell division. Nat Commun 2020; 11:4097. [PMID: 32796861 PMCID: PMC7427965 DOI: 10.1038/s41467-020-17940-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 07/24/2020] [Indexed: 01/03/2023] Open
Abstract
Staphylococcus aureus is generally thought to divide in three alternating orthogonal planes over three consecutive division cycles. Although this mode of division was proposed over four decades ago, the molecular mechanism that ensures this geometry of division has remained elusive. Here we show, for three different strains, that S. aureus cells do not regularly divide in three alternating perpendicular planes as previously thought. Imaging of the divisome shows that a plane of division is always perpendicular to the previous one, avoiding bisection of the nucleoid, which segregates along an axis parallel to the closing septum. However, one out of the multiple planes perpendicular to the septum which divide the cell in two identical halves can be used in daughter cells, irrespective of its orientation in relation to the penultimate division plane. Therefore, division in three orthogonal planes is not the rule in S. aureus.
Collapse
|
21
|
Domain sliding of two Staphylococcus aureus N-acetylglucosaminidases enables their substrate-binding prior to its catalysis. Commun Biol 2020; 3:178. [PMID: 32313083 PMCID: PMC7170848 DOI: 10.1038/s42003-020-0911-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 03/23/2020] [Indexed: 12/22/2022] Open
Abstract
To achieve productive binding, enzymes and substrates must align their geometries to complement each other along an entire substrate binding site, which may require enzyme flexibility. In pursuit of novel drug targets for the human pathogen S. aureus, we studied peptidoglycan N-acetylglucosaminidases, whose structures are composed of two domains forming a V-shaped active site cleft. Combined insights from crystal structures supported by site-directed mutagenesis, modeling, and molecular dynamics enabled us to elucidate the substrate binding mechanism of SagB and AtlA-gl. This mechanism requires domain sliding from the open form observed in their crystal structures, leading to polysaccharide substrate binding in the closed form, which can enzymatically process the bound substrate. We suggest that these two hydrolases must exhibit unusual extents of flexibility to cleave the rigid structure of a bacterial cell wall.
Collapse
|
22
|
Lee CC, Southgate R, Jiao C, Gersz E, Owen JR, Kates SL, Beck CA, Xie C, Daiss JL, Post V, Moriarty TF, Zeiter S, Schwarz EM, Muthukrishnan G. Deriving a dose and regimen for anti-glucosaminidase antibody passive-immunisation for patients with Staphylococcus aureus osteomyelitis. Eur Cell Mater 2020; 39:96-107. [PMID: 32003439 PMCID: PMC7236896 DOI: 10.22203/ecm.v039a06] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Staphylococcus aureus (S. aureus) osteomyelitis remains a major clinical problem. Anti-glucosaminidase (Gmd) antibodies (1C11) are efficacious in prophylactic and therapeutic murine models. Feasibility, safety and pharmacokinetics of 1C11 passive immunisation in sheep and endogenous anti-Gmd levels were quantified in osteomyelitis patients. 3 sheep received a 500 mg intravenous (i.v.) bolus of 1C11 and its levels in sera were determined by enzyme-linked immunosorbent assay (ELISA) over 52 d. A humanised anti-Gmd monoclonal antibody, made by grafting the antigen-binding fragment (Fab) portion of 1C11 onto the fragment crystallisable region (Fc) of human IgG1, was used to make a standard curve of mean fluorescent intensity versus concentration of anti-Gmd. Anti-Gmd serum levels were determined in 297 patients with culture-confirmed S. aureus osteomyelitis and 40 healthy controls. No complications or adverse events were associated with the sheep 1C11 i.v. infusion and the estimated circulating half-life of 1C11 was 23.7 d. Endogenous anti-Gmd antibody levels in sera of osteomyelitis patients ranged from < 1 ng/mL to 300 µg/mL, with a mean concentration of 21.7 µg/mL. The estimated circulating half-life of endogenous anti-Gmd antibodies in sera of 12 patients with cured osteomyelitis was 120.4 d. A clinically relevant administration of anti-Gmd (500 mg i.v. = 7 mg/kg/70 kg human) was safe in sheep. This dose was 8 times more than the endogenous anti-Gmd levels observed in osteomyelitis patients and was predicted to have a half-life of > 3 weeks. Anti-Gmd passive immunisation has potential to prevent and treat S. aureus osteomyelitis. Further clinical development is warranted.
Collapse
Affiliation(s)
- Charles C. Lee
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - Richard Southgate
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - Cindy Jiao
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - Elaine Gersz
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - John R. Owen
- Department of Orthopaedic Surgery, Virginia Commonwealth University, Richmond, VA, USA
| | - Stephen L. Kates
- Department of Orthopaedic Surgery, Virginia Commonwealth University, Richmond, VA, USA
| | - Christopher A. Beck
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - Chao Xie
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - John L. Daiss
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | | | | | | | - Edward M. Schwarz
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA,Corresponding Author: Edward M. Schwarz, Ph.D., Burton Professor of Orthopaedics, Director of Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Avenue, Box 665, Rochester, NY 14642, Phone: (585) 275-3063, FAX: (585) 276-2177,
| | | |
Collapse
|
23
|
Jensen C, Bæk KT, Gallay C, Thalsø-Madsen I, Xu L, Jousselin A, Ruiz Torrubia F, Paulander W, Pereira AR, Veening JW, Pinho MG, Frees D. The ClpX chaperone controls autolytic splitting of Staphylococcus aureus daughter cells, but is bypassed by β-lactam antibiotics or inhibitors of WTA biosynthesis. PLoS Pathog 2019; 15:e1008044. [PMID: 31518377 PMCID: PMC6760813 DOI: 10.1371/journal.ppat.1008044] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 09/25/2019] [Accepted: 08/26/2019] [Indexed: 12/02/2022] Open
Abstract
β-lactam antibiotics interfere with cross-linking of the bacterial cell wall, but the killing mechanism of this important class of antibiotics is not fully understood. Serendipitously we found that sub-lethal doses of β-lactams rescue growth and prevent spontaneous lysis of Staphylococcus aureus mutants lacking the widely conserved chaperone ClpX, and we reasoned that a better understanding of the clpX phenotypes could provide novel insights into the downstream effects of β-lactam binding to the PBP targets. Super-resolution imaging revealed that clpX cells display aberrant septum synthesis, and initiate daughter cell separation prior to septum completion at 30°C, but not at 37°C, demonstrating that ClpX becomes critical for coordinating the S. aureus cell cycle as the temperature decreases. FtsZ localization and dynamics were not affected in the absence of ClpX, suggesting that ClpX affects septum formation and autolytic activation downstream of Z-ring formation. Interestingly, oxacillin antagonized the septum progression defects of clpX cells and prevented lysis of prematurely splitting clpX cells. Strikingly, inhibitors of wall teichoic acid (WTA) biosynthesis that work synergistically with β-lactams to kill MRSA synthesis also rescued growth of the clpX mutant, as did genetic inactivation of the gene encoding the septal autolysin, Sle1. Taken together, our data support a model in which Sle1 causes premature splitting and lysis of clpX daughter cells unless Sle1-dependent lysis is antagonized by β-lactams or by inhibiting an early step in WTA biosynthesis. The finding that β-lactams and inhibitors of WTA biosynthesis specifically prevent lysis of a mutant with dysregulated autolytic activity lends support to the idea that PBPs and WTA biosynthesis play an important role in coordinating cell division with autolytic splitting of daughter cells, and that β-lactams do not kill S. aureus simply by weakening the cell wall. The bacterium Staphylococcus aureus is a major cause of human disease, and the rapid spread of S. aureus strains that are resistant to almost all β-lactam antibiotics has made treatment increasingly difficult. β-lactams interfere with cross-linking of the bacterial cell wall but the killing mechanism of this important class of antibiotics is not fully understood. Here we provide novel insight into this topic by examining a defined S. aureus mutant that has the unusual property of growing markedly better in the presence of β-lactams. Without β-lactams this mutant dies spontaneously at a high frequency due to premature separation of daughter cells during cell division. Cell death of the mutant can, however, be prevented either by exposure to β-lactam antibiotics or by inhibiting synthesis of wall teichoic acid, a major component of the cell wall in Gram-positive bacteria with a conserved role in activation of autolytic splitting of daughter cells. The finding that β-lactam antibiotics can prevent lysis of a mutant with deregulated activity of autolytic enzymes involved in daughter cell splitting, emphasizes the idea that β-lactams interfere with the coordination between cell division and daughter cell splitting, and do not kill S. aureus simply by weakening the cell wall.
Collapse
Affiliation(s)
- Camilla Jensen
- Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kristoffer T. Bæk
- Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Clement Gallay
- Department of Fundamental Microbiology, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Ida Thalsø-Madsen
- Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lijuan Xu
- Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ambre Jousselin
- Bacterial Cell Biology, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Fernando Ruiz Torrubia
- Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Wilhelm Paulander
- Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ana R. Pereira
- Bacterial Cell Biology, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Jan-Willem Veening
- Department of Fundamental Microbiology, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Mariana G. Pinho
- Bacterial Cell Biology, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Dorte Frees
- Department of Veterinary and Animal Sciences, University of Copenhagen, Copenhagen, Denmark
- * E-mail:
| |
Collapse
|
24
|
Mayer C, Kluj RM, Mühleck M, Walter A, Unsleber S, Hottmann I, Borisova M. Bacteria's different ways to recycle their own cell wall. Int J Med Microbiol 2019; 309:151326. [PMID: 31296364 DOI: 10.1016/j.ijmm.2019.06.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Revised: 05/28/2019] [Accepted: 06/30/2019] [Indexed: 01/05/2023] Open
Abstract
The ability to recover components of their own cell wall is a common feature of bacteria. This was initially recognized in the Gram-negative bacterium Escherichia coli, which recycles about half of the peptidoglycan of its cell wall during one cell doubling. Moreover, E. coli was shown to grow on peptidoglycan components provided as nutrients. A distinguished recycling enzyme of E. coli required for both, recovery of the cell wall sugar N-acetylmuramic acid (MurNAc) of the own cell wall and for growth on external MurNAc, is the MurNAc 6-phosphate (MurNAc 6P) lactyl ether hydrolase MurQ. We revealed however, that most Gram-negative bacteria lack a murQ ortholog and instead harbor a pathway, absent in E. coli, that channels MurNAc directly to peptidoglycan biosynthesis. This "anabolic recycling pathway" bypasses the initial steps of peptidoglycan de novo synthesis, including the target of the antibiotic fosfomycin, thus providing intrinsic resistance to the antibiotic. The Gram-negative oral pathogen Tannerella forsythia is auxotrophic for MurNAc and apparently depends on the anabolic recycling pathway to synthesize its own cell wall by scavenging cell wall debris of other bacteria. In contrast, Gram-positive bacteria lack the anabolic recycling genes, but mostly contain one or two murQ orthologs. Quantification of MurNAc 6P accumulation in murQ mutant cells by mass spectrometry allowed us to demonstrate for the first time that Gram-positive bacteria do recycle their own peptidoglycan. This had been questioned earlier, since peptidoglycan turnover products accumulate in the spent media of Gram-positives. We showed, that these fragments are recovered during nutrient limitation, which prolongs starvation survival of Bacillus subtilis and Staphylococcus aureus. Peptidoglycan recycling in these bacteria however differs, as the cell wall is either cleaved exhaustively and monosaccharide building blocks are taken up (B. subtilis) or disaccharides are released and recycled involving a novel phosphomuramidase (MupG; S.aureus). In B. subtilis also the teichoic acids, covalently bound to the peptidoglycan (wall teichoic acids; WTAs), are recycled. During phosphate limitation, the sn-glycerol-3-phosphate phosphodiesterase GlpQ specifically degrades WTAs of B. subtilis. In S. aureus, in contrast, GlpQ is used to scavenge external teichoic acid sources. Thus, although bacteria generally recover their own cell wall, they apparently apply distinct strategies for breakdown and reutilization of cell wall fragments. This review summarizes our work on this topic funded between 2011 and 2019 by the DFG within the collaborative research center SFB766.
Collapse
Affiliation(s)
- Christoph Mayer
- Mikrobiologie/Biotechnologie, Interfakultäres Institut für Mikrobiologie und Infektionsmedizin Tübingen (IMIT), Eberhard Karls Universität Tübingen, Auf der Morgenstelle 28, 72076, Tübingen, Germany.
| | - Robert Maria Kluj
- Mikrobiologie/Biotechnologie, Interfakultäres Institut für Mikrobiologie und Infektionsmedizin Tübingen (IMIT), Eberhard Karls Universität Tübingen, Auf der Morgenstelle 28, 72076, Tübingen, Germany
| | - Maraike Mühleck
- Mikrobiologie/Biotechnologie, Interfakultäres Institut für Mikrobiologie und Infektionsmedizin Tübingen (IMIT), Eberhard Karls Universität Tübingen, Auf der Morgenstelle 28, 72076, Tübingen, Germany
| | - Axel Walter
- Mikrobiologie/Biotechnologie, Interfakultäres Institut für Mikrobiologie und Infektionsmedizin Tübingen (IMIT), Eberhard Karls Universität Tübingen, Auf der Morgenstelle 28, 72076, Tübingen, Germany
| | - Sandra Unsleber
- Mikrobiologie/Biotechnologie, Interfakultäres Institut für Mikrobiologie und Infektionsmedizin Tübingen (IMIT), Eberhard Karls Universität Tübingen, Auf der Morgenstelle 28, 72076, Tübingen, Germany
| | - Isabel Hottmann
- Mikrobiologie/Biotechnologie, Interfakultäres Institut für Mikrobiologie und Infektionsmedizin Tübingen (IMIT), Eberhard Karls Universität Tübingen, Auf der Morgenstelle 28, 72076, Tübingen, Germany
| | - Marina Borisova
- Mikrobiologie/Biotechnologie, Interfakultäres Institut für Mikrobiologie und Infektionsmedizin Tübingen (IMIT), Eberhard Karls Universität Tübingen, Auf der Morgenstelle 28, 72076, Tübingen, Germany
| |
Collapse
|
25
|
Abstract
Dating back to the 1960s, initial studies on the staphylococcal cell wall were driven by the need to clarify the mode of action of the first antibiotics and the resistance mechanisms developed by the bacteria. During the following decades, the elucidation of the biosynthetic path and primary composition of staphylococcal cell walls was propelled by advances in microbial cell biology, specifically, the introduction of high-resolution analytical techniques and molecular genetic approaches. The field of staphylococcal cell wall gradually gained its own significance as the complexity of its chemical structure and involvement in numerous cellular processes became evident, namely its versatile role in host interactions, coordination of cell division and environmental stress signaling.This chapter includes an updated description of the anatomy of staphylococcal cell walls, paying particular attention to information from the last decade, under four headings: high-resolution analysis of the Staphylococcus aureus peptidoglycan; variations in peptidoglycan composition; genetic determinants and enzymes in cell wall synthesis; and complex functions of cell walls. The latest contributions to a more precise picture of the staphylococcal cell envelope were possible due to recently developed state-of-the-art microscopy and spectroscopy techniques and to a wide combination of -omics approaches, that are allowing to obtain a more integrative view of this highly dynamic structure.
Collapse
Affiliation(s)
- Rita Sobral
- UCIBIO-REQUIMTE, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Caparica, Portugal
| | | |
Collapse
|
26
|
Flores-Kim J, Dobihal GS, Fenton A, Rudner DZ, Bernhardt TG. A switch in surface polymer biogenesis triggers growth-phase-dependent and antibiotic-induced bacteriolysis. eLife 2019; 8:44912. [PMID: 30964003 PMCID: PMC6456293 DOI: 10.7554/elife.44912] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Accepted: 03/11/2019] [Indexed: 01/21/2023] Open
Abstract
Penicillin and related antibiotics disrupt cell wall synthesis to induce bacteriolysis. Lysis in response to these drugs requires the activity of cell wall hydrolases called autolysins, but how penicillins misactivate these deadly enzymes has long remained unclear. Here, we show that alterations in surface polymers called teichoic acids (TAs) play a key role in penicillin-induced lysis of the Gram-positive pathogen Streptococcus pneumoniae (Sp). We find that during exponential growth, Sp cells primarily produce lipid-anchored TAs called lipoteichoic acids (LTAs) that bind and sequester the major autolysin LytA. However, penicillin-treatment or prolonged stationary phase growth triggers the degradation of a key LTA synthase, causing a switch to the production of wall-anchored TAs (WTAs). This change allows LytA to associate with and degrade its cell wall substrate, thus promoting osmotic lysis. Similar changes in surface polymer assembly may underlie the mechanism of antibiotic- and/or growth phase-induced lysis for other important Gram-positive pathogens.
Collapse
Affiliation(s)
- Josué Flores-Kim
- Department of Microbiology, Harvard Medical School, Boston, United States
| | | | - Andrew Fenton
- Department of Microbiology, Harvard Medical School, Boston, United States.,The Florey Institute, Molecular Biology Biotechnology, University of Sheffield, Sheffield, United Kingdom
| | - David Z Rudner
- Department of Microbiology, Harvard Medical School, Boston, United States
| | - Thomas G Bernhardt
- Department of Microbiology, Harvard Medical School, Boston, United States.,Howard Hughes Medical Institute, Boston, United States
| |
Collapse
|
27
|
Vermassen A, Leroy S, Talon R, Provot C, Popowska M, Desvaux M. Cell Wall Hydrolases in Bacteria: Insight on the Diversity of Cell Wall Amidases, Glycosidases and Peptidases Toward Peptidoglycan. Front Microbiol 2019; 10:331. [PMID: 30873139 PMCID: PMC6403190 DOI: 10.3389/fmicb.2019.00331] [Citation(s) in RCA: 189] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 02/08/2019] [Indexed: 11/13/2022] Open
Abstract
The cell wall (CW) of bacteria is an intricate arrangement of macromolecules, at least constituted of peptidoglycan (PG) but also of (lipo)teichoic acids, various polysaccharides, polyglutamate and/or proteins. During bacterial growth and division, there is a constant balance between CW degradation and biosynthesis. The CW is remodeled by bacterial hydrolases, whose activities are carefully regulated to maintain cell integrity or lead to bacterial death. Each cell wall hydrolase (CWH) has a specific role regarding the PG: (i) cell wall amidase (CWA) cleaves the amide bond between N-acetylmuramic acid and L-alanine residue at the N-terminal of the stem peptide, (ii) cell wall glycosidase (CWG) catalyses the hydrolysis of the glycosidic linkages, whereas (iii) cell wall peptidase (CWP) cleaves amide bonds between amino acids within the PG chain. After an exhaustive overview of all known conserved catalytic domains responsible for CWA, CWG, and CWP activities, this review stresses that the CWHs frequently display a modular architecture combining multiple and/or different catalytic domains, including some lytic transglycosylases as well as CW binding domains. From there, direct physiological and collateral roles of CWHs in bacterial cells are further discussed.
Collapse
Affiliation(s)
- Aurore Vermassen
- Université Clermont Auvergne, INRA, MEDiS, Clermont-Ferrand, France
| | - Sabine Leroy
- Université Clermont Auvergne, INRA, MEDiS, Clermont-Ferrand, France
| | - Régine Talon
- Université Clermont Auvergne, INRA, MEDiS, Clermont-Ferrand, France
| | | | - Magdalena Popowska
- Department of Applied Microbiology, Faculty of Biology, Institute of Microbiology, University of Warsaw, Warsaw, Poland
| | - Mickaël Desvaux
- Université Clermont Auvergne, INRA, MEDiS, Clermont-Ferrand, France
| |
Collapse
|
28
|
Kalali Y, Haghighat S, Mahdavi M. Passive immunotherapy with specific IgG fraction against autolysin: Analogous protectivity in the MRSA infection with antibiotic therapy. Immunol Lett 2018; 212:125-131. [PMID: 30496765 DOI: 10.1016/j.imlet.2018.11.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 11/20/2018] [Accepted: 11/25/2018] [Indexed: 11/16/2022]
Abstract
Staphylococcus aureus is a leading infectious cause of life-threatening diseases in human beings, with no effective vaccine available to date against this bacterium. Treatment of methicillin-resistant S. aureus (MRSA) infections has become increasingly difficult because of the emergence of multidrug-resistant isolates. Immunotherapy represents a potential approach to prevent S. aureus-related infections. Autolysin is one of the virulence factors, which controls the growth, cell lysis, daughter-cell separation, and biofilm formation. Our study focused on passive immunization against MRSA infection. Herein, rabbit polyclonal IgG was produced following the preparation of r-autolysin. Specificity of IgG against r-autolysin was investigated by ELISA and western blotting assays. IgG fraction was prepared using sulfate ammonium precipitation, and the ability of antiserum to promote phagocytosis of bacteria was assessed by opsonophagocytosis assay. Then, passive immunization of mice was carried out with polyclonal IgG fraction and, mice were sacrificed three days after challenge and their kidneys, liver, and spleen were collected. Results exhibited that the passive immunization with rabbit polyclonal anti-IgG fraction tremendously improved survival rates of mice challenged by S. aureus as well as vancomycin treatment compared with the negative control groups. In addition, a remarkable decrease in bacterial numbers was observed in mice treated with rabbit polyclonal anti-IgG. Importantly, our findings demonstrated that passive immunotherapy and antibiotic therapy lead to decreased histopathological damage in mice infected by S. aureus as compared with control groups. Our results suggested that the passive immunization may result in the introduction of excellent strategies to control infections caused by MRSA, like antibiotic therapy.
Collapse
Affiliation(s)
- Yasamin Kalali
- Department of Biotechnology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Setareh Haghighat
- Department of Microbiology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Mehdi Mahdavi
- Recombinant Vaccine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
29
|
Kluj RM, Ebner P, Adamek M, Ziemert N, Mayer C, Borisova M. Recovery of the Peptidoglycan Turnover Product Released by the Autolysin Atl in Staphylococcus aureus Involves the Phosphotransferase System Transporter MurP and the Novel 6-phospho- N-acetylmuramidase MupG. Front Microbiol 2018; 9:2725. [PMID: 30524387 PMCID: PMC6262408 DOI: 10.3389/fmicb.2018.02725] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 10/24/2018] [Indexed: 11/29/2022] Open
Abstract
The peptidoglycan of the bacterial cell wall undergoes a permanent turnover during cell growth and differentiation. In the Gram-positive pathogen Staphylococcus aureus, the major peptidoglycan hydrolase Atl is required for accurate cell division, daughter cell separation and autolysis. Atl is a bifunctional N-acetylmuramoyl-L-alanine amidase/endo-β-N-acetylglucosaminidase that releases peptides and the disaccharide N-acetylmuramic acid-β-1,4-N-acetylglucosamine (MurNAc-GlcNAc) from the peptido-glycan. Here we revealed the recycling pathway of the cell wall turnover product MurNAc-GlcNAc in S. aureus. The latter disaccharide is internalized and concomitantly phosphorylated by the phosphotransferase system (PTS) transporter MurP, which had been implicated previously in the uptake and phosphorylation of MurNAc. Since MurP mutant cells accumulate MurNAc-GlcNAc and not MurNAc in the culture medium during growth, the disaccharide represents the physiological substrate of the PTS transporter. We further identified and characterized a novel 6-phospho-N-acetylmuramidase, named MupG, which intracellularly hydrolyses MurNAc 6-phosphate-GlcNAc, the product of MurP-uptake and phosphorylation, yielding MurNAc 6-phosphate and GlcNAc. MupG is the first characterized representative of a novel family of glycosidases containing domain of unknown function 871 (DUF871). The corresponding gene mupG (SAUSA300_0192) of S. aureus strain USA300 is the first gene within a putative operon that also includes genes encoding the MurNAc 6-phosphate etherase MurQ, MurP, and the putative transcriptional regulator MurR. Using mass spectrometry, we observed cytoplasmic accumulation of MurNAc 6-phosphate-GlcNAc in ΔmupG and ΔmupGmurQ markerless non-polar deletion mutants, but not in the wild type or in the complemented ΔmupG strain. MurNAc 6-phosphate-GlcNAc levels in the mutants increased during stationary phase, in accordance with previous observations regarding peptidoglycan recycling in S. aureus.
Collapse
Affiliation(s)
- Robert Maria Kluj
- Microbiology/Biotechnology, Department of Biology, Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany
| | - Patrick Ebner
- Microbial Genetics, Department of Biology, Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany
| | - Martina Adamek
- Microbiology/Biotechnology, Department of Biology, Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany
| | - Nadine Ziemert
- Microbiology/Biotechnology, Department of Biology, Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany
| | - Christoph Mayer
- Microbiology/Biotechnology, Department of Biology, Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany
| | - Marina Borisova
- Microbiology/Biotechnology, Department of Biology, Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany
| |
Collapse
|
30
|
Carvalho F, Sousa S, Cabanes D. l-Rhamnosylation of wall teichoic acids promotes efficient surface association of Listeria monocytogenes virulence factors InlB and Ami through interaction with GW domains. Environ Microbiol 2018; 20:3941-3951. [PMID: 29984543 DOI: 10.1111/1462-2920.14351] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 06/18/2018] [Accepted: 07/02/2018] [Indexed: 11/29/2022]
Abstract
Wall teichoic acids (WTAs) are important surface glycopolymers involved in various physiological processes occurring in the Gram-positive cell envelope. We previously showed that the decoration of Listeria monocytogenes (Lm) WTAs with l-rhamnose conferred resistance against antimicrobial peptides. Here, we show that WTA l-rhamnosylation also contributes to physiological levels of autolysis in Lm through a mechanism that requires efficient association of Ami, a virulence-promoting autolysin belonging to the GW protein family, to the bacterial cell surface. Importantly, WTA l-rhamnosylation also controls the surface association of another GW protein, the invasin internalin B (InlB), that promotes Lm invasion of host cells. Whereas WTA N-acetylglucosaminylation is not a prerequisite for GW protein surface association, lipoteichoic acids appear to also play a role in the surface anchoring of InlB. Strikingly, while the GW domains of Ami, InlB and Auto (another autolysin contributing to cell invasion and virulence) are sufficient to mediate surface association, this is not the case for the GW domains of the remaining six uncharacterized Lm GW proteins. Overall, we reveal WTA l-rhamnosylation as a bacterial surface modification mechanism that contributes to Lm physiology and pathogenesis by controlling the surface association of GW proteins involved in autolysis and infection.
Collapse
Affiliation(s)
- Filipe Carvalho
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Group of Molecular Microbiology, IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Sandra Sousa
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Group of Molecular Microbiology, IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Didier Cabanes
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Group of Molecular Microbiology, IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| |
Collapse
|
31
|
Yokogawa N, Ishikawa M, Nishitani K, Beck CA, Tsuchiya H, Mesfin A, Kates SL, Daiss JL, Xie C, Schwarz EM. Immunotherapy synergizes with debridement and antibiotic therapy in a murine 1-stage exchange model of MRSA implant-associated osteomyelitis. J Orthop Res 2018; 36:1590-1598. [PMID: 29405452 PMCID: PMC6541030 DOI: 10.1002/jor.23801] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 10/31/2017] [Indexed: 02/04/2023]
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) reinfection following revision surgery remains a major orthopaedic problem. Toward the development of immunotherapy with anti-glucosaminidase monoclonal antibodies (anti-Gmd), we aimed to: (i) develop a murine 1-stage exchange model of bioluminescent MRSA (USA300LAC::lux) contaminated femoral implants; and (ii) utilize this model to demonstrate the synergistic effects of combination vancomycin and anti-Gmd therapy on reinfection and bone healing. Following an infection surgery, the original plate and two screws were removed on day 7, and exchanged with sterile implants. Mice were randomized to five groups: (i) no infection control; (ii) infected placebo; (iii) anti-Gmd; (iv) vancomycin; and (v) combination therapy. Bioluminescent imaging (BLI) was performed on days 0, 1, 3, 5, 7, 8, 10, 12, and 14. Mice were euthanized on day 14 (day 7 post-revision), and efficacy was assessed via colony forming units (CFU) on explanted hardware, micro-CT, and histology. As monotherapies, anti-Gmd inhibited Staphylococcus abscess communities, and vancomycin reduced CFU on the implants. However, only combination therapy prevented increased BLI post-revision surgery, with a significant 6.5-fold reduction on day 10 (p < 0.05 vs. placebo), and achieved sterile implant levels by day 12. Synergistic effects were also apparent from reduced osteolysis and increased new bone formation around the screws only observed following combination therapy. Taken together, we find that: (i) this murine femoral plate 1-stage revision model can efficiently evaluate therapies to prevent reinfection; and (ii) immunotherapy plays a distinct role from antibiotics to reduce reinfection following revision surgery, such that synergy to achieve osseointegration is possible. © 2018 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:1590-1598, 2018.
Collapse
Affiliation(s)
- Noriaki Yokogawa
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY,Department of Orthopaedic Surgery, Kanazawa University, Kanazawa, Japan
| | - Masahiro Ishikawa
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY,Department of Orthopaedics Surgery, Kyoto University, Kyoto, Japan
| | - Kohei Nishitani
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY,Department of Orthopaedics Surgery, Kyoto University, Kyoto, Japan
| | - Christopher A. Beck
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY,Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, NY,Department of Orthopedics, University of Rochester Medical Center, Rochester, NY
| | - Hiroyuki Tsuchiya
- Department of Orthopaedic Surgery, Kanazawa University, Kanazawa, Japan
| | - Addisu Mesfin
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY,Department of Orthopedics, University of Rochester Medical Center, Rochester, NY
| | - Stephen L. Kates
- Department of Orthopedic Surgery, Virginia Commonwealth University, Richmond, VA
| | - John L. Daiss
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY,Department of Orthopedics, University of Rochester Medical Center, Rochester, NY
| | - Chao Xie
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY,Department of Orthopedics, University of Rochester Medical Center, Rochester, NY
| | - Edward M. Schwarz
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY,Department of Orthopedics, University of Rochester Medical Center, Rochester, NY
| |
Collapse
|
32
|
Release of Staphylococcus aureus extracellular vesicles and their application as a vaccine platform. Nat Commun 2018; 9:1379. [PMID: 29643357 PMCID: PMC5895597 DOI: 10.1038/s41467-018-03847-z] [Citation(s) in RCA: 189] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 03/14/2018] [Indexed: 01/08/2023] Open
Abstract
Secretion of extracellular vesicles (EVs), a process common to eukaryotes, archae, and bacteria, represents a secretory pathway that allows cell-free intercellular communication. Microbial EVs package diverse proteins and influence the host-pathogen interaction, but the mechanisms underlying EV production in Gram-positive bacteria are poorly understood. Here we show that EVs purified from community-associated methicillin-resistant Staphylococcus aureus package cytosolic, surface, and secreted proteins, including cytolysins. Staphylococcal alpha-type phenol-soluble modulins promote EV biogenesis by disrupting the cytoplasmic membrane; whereas, peptidoglycan cross-linking and autolysin activity modulate EV production by altering the permeability of the cell wall. We demonstrate that EVs purified from a S. aureus mutant that is genetically engineered to express detoxified cytolysins are immunogenic in mice, elicit cytolysin-neutralizing antibodies, and protect the animals in a lethal sepsis model. Our study reveals mechanisms underlying S. aureus EV production and highlights the usefulness of EVs as a S. aureus vaccine platform. Extracellular vesicles (EVs) influence host-pathogen interactions, but EV biogenesis in gram-positive bacteria remains elusive. Here authors characterize EVs from Staphylococcus aureus and show that phenol-soluble modulins and autolysins promote EV biogenesis by disrupting the membrane and cell wall.
Collapse
|
33
|
Bornikoel J, Staiger J, Madlung J, Forchhammer K, Maldener I. LytM factor Alr3353 affects filament morphology and cell-cell communication in the multicellular cyanobacteriumAnabaenasp. PCC 7120. Mol Microbiol 2018; 108:187-203. [DOI: 10.1111/mmi.13929] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/09/2018] [Indexed: 01/16/2023]
Affiliation(s)
- Jan Bornikoel
- Interfaculty Institute of Microbiology and Infection Medicine Tübingen, Organismic Interactions; University of Tübingen, Auf der Morgenstelle 28; 72076 Tübingen Germany
| | - Julia Staiger
- Interfaculty Institute of Microbiology and Infection Medicine Tübingen, Organismic Interactions; University of Tübingen, Auf der Morgenstelle 28; 72076 Tübingen Germany
| | - Johannes Madlung
- Proteome Center Tübingen; University of Tübingen, Auf der Morgenstelle 15; 72076 Tübingen Germany
| | - Karl Forchhammer
- Interfaculty Institute of Microbiology and Infection Medicine Tübingen, Organismic Interactions; University of Tübingen, Auf der Morgenstelle 28; 72076 Tübingen Germany
| | - Iris Maldener
- Interfaculty Institute of Microbiology and Infection Medicine Tübingen, Organismic Interactions; University of Tübingen, Auf der Morgenstelle 28; 72076 Tübingen Germany
| |
Collapse
|
34
|
Proteomics of Staphylococcus aureus biofilm matrix in a rat model of orthopedic implant-associated infection. PLoS One 2017; 12:e0187981. [PMID: 29121106 PMCID: PMC5679556 DOI: 10.1371/journal.pone.0187981] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 10/30/2017] [Indexed: 01/09/2023] Open
Abstract
The matrix proteins of Staphylococcus aureus biofilm have not been well defined. Previous efforts to identify these proteins were performed using in vitro systems. Here we use a proteomic approach to identify biofilm matrix proteins directly from infected bone implants using a rat model of orthopedic implant-associated S. aureus infection. Despite heavy presence of host proteins, a total of 28 and 105 S. aureus proteins were identified during acute infection and chronic infection, respectively. Our results show that biofilm matrix contains mostly intracellular cytoplasmic proteins and, to a much less extent, extracellular and cell surface-associated proteins. Significantly, leukocidins were identified in the biofilm matrix during chronic infection, suggesting S. aureus is actively attacking the host immune system even though they are protected within the biofilm. The presence of two surface-associated proteins, Ebh and SasF, in the infected bone tissue during acute infection was confirmed by immunohistochemistry. In addition, a large number of host proteins were found differentially expressed in response to S. aureus biofilm formed on bone implants.
Collapse
|
35
|
A novel recombinant vaccine candidate comprising PBP2a and autolysin against Methicillin Resistant Staphylococcus aureus confers protection in the experimental mice. Mol Immunol 2017; 91:1-7. [DOI: 10.1016/j.molimm.2017.08.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 08/01/2017] [Accepted: 08/14/2017] [Indexed: 11/18/2022]
|
36
|
Vargas S, Millán-Chiu BE, Arvizu-Medrano SM, Loske AM, Rodríguez R. Dynamic light scattering: A fast and reliable method to analyze bacterial growth during the lag phase. J Microbiol Methods 2017; 137:34-39. [DOI: 10.1016/j.mimet.2017.04.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 04/08/2017] [Accepted: 04/08/2017] [Indexed: 10/19/2022]
|
37
|
Ujimine S, Tone S, Saito M, Yamada S. Intracellular morphological changes in Staphylococcus aureus induced by treatment with sodium hypochlorite. Med Mol Morphol 2017; 50:178-184. [PMID: 28516285 DOI: 10.1007/s00795-017-0159-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 05/08/2017] [Indexed: 12/15/2022]
Abstract
Sodium hypochlorite (NaOCl) is commonly used as a disinfectant; however, its bactericidal mechanism has not yet been clarified. In the present study, the bactericidal mechanism of NaOCl was examined using microscopy and gel electrophoresis techniques with Staphylococcus aureus strain 209P. S. aureus cells treated with 500 and 1000 ppm NaOCl for 5 and 15 min were observed by SEM and TEM. SEM images of the bacterial cells treated with NaOCl showed an irregular surface, with cells being partially invaginated. TEM images of the bacterial cells showed cytoplasmic alterations, accompanied by a partially irregular cellular surface. Under a fluorescence microscope, we clearly observed fluorescence quenching in the 1000 ppm NaOCl-treated cells. Based on these observations, which indicated that NaOCl damaged chromosomal DNA, we next extracted chromosomal DNA from bacterial cells treated with NaOCl and performed agarose gel electrophoresis. Chromosomal DNA was absent in the DNA sample from the bacterial cells treated with 500 ppm NaOCl. From these biochemical results, it was strongly suggested that NaOCl degrades the chromosomal DNA of S. aureus. We consider that the morphological changes in the cytoplasm induced by NaOCl may be related to NaOCl-induced degradation of S. aureus chromosomal DNA.
Collapse
Affiliation(s)
- Shiori Ujimine
- Department of Health Science, Kawasaki University of Medical Welfare, 288 Matsushima, Kurashiki, Okayama, 701-0193, Japan.
| | - Shigenobu Tone
- Graduate School of Science and Engineering, Tokyo Denki University, Hatoyama, Saitama, 350-0394, Japan
| | - Mineki Saito
- Department of Microbiology, Kawasaki Medical School, 577 Matsushima, Kurashiki, Okayama, 701-0192, Japan
| | - Sakuo Yamada
- Department of Health Science, Kawasaki University of Medical Welfare, 288 Matsushima, Kurashiki, Okayama, 701-0193, Japan
- Department of Microbiology, Kawasaki Medical School, 577 Matsushima, Kurashiki, Okayama, 701-0192, Japan
| |
Collapse
|
38
|
Lysibodies are IgG Fc fusions with lysin binding domains targeting Staphylococcus aureus wall carbohydrates for effective phagocytosis. Proc Natl Acad Sci U S A 2017; 114:4781-4786. [PMID: 28428342 DOI: 10.1073/pnas.1619249114] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The cell wall of Gram-positive bacteria contains abundant surface-exposed carbohydrate molecules that are highly conserved within and often across species. The potential therapeutic usefulness of high-affinity antibodies to cell wall carbohydrates is unquestioned, however obtaining such antibodies is challenging due to the poor overall immunogenicity of these bacterial targets. Autolysins and phage lysins are peptidoglycan hydrolases, enzymes that have evolved over a billion years to degrade bacterial cell wall. Such wall hydrolases are modular enzymes, composed of discrete domains for high-affinity binding to cell wall carbohydrates and cleavage activity. In this study, we demonstrate that binding domains from autolysins and lysins can be fused to the Fc region of human IgG, creating a fully functional homodimer (or "lysibody") with high-affinity binding and specificity for carbohydrate determinants on the bacterial surface. Furthermore, we demonstrate that this process is reproducible with three different binding domains specific to methicillin-resistant Staphylococcus aureus (MRSA). Cell-bound lysibodies induced the fixation of complement on the bacterial surface, promoted phagocytosis by macrophages and neutrophils, and protected mice from MRSA infection in two model systems. The lysibody approach could be used to target a range of difficult-to-treat pathogenic bacteria, given that cell wall hydrolases are ubiquitous in nature.
Collapse
|
39
|
Haghighat S, Siadat SD, Sorkhabadi SMR, Sepahi AA, Mahdavi M. Cloning, expression and purification of autolysin from methicillin-resistant Staphylococcus aureus: potency and challenge study in Balb/c mice. Mol Immunol 2016; 82:10-18. [PMID: 28006655 DOI: 10.1016/j.molimm.2016.12.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 12/09/2016] [Accepted: 12/12/2016] [Indexed: 10/20/2022]
Abstract
Staphylococcus aureus (MRSA) is an opportunistic pathogen which causes a variety of clinical diseases and leads to high rates of morbidity and mortality. Development of an effective vaccine appears to be a useful strategy to control the infection. Here, the internal region of atl was cloned into the pET24a plasmid and expressed in E. coli BL21 (DE3). Cloning of atl was confirmed by colony-PCR, enzymatic digestion and sequencing. Protein expressed in E coli, BL21 DE3 and was confirmed with SDS-PAGE and western blot analysis. Subsequently, BALB/c mice were injected subcutaneously three times with 20μg of the recombinant autolysin. After Bleeding, autolysin-specific total IgG antibodies and isotypes were evaluated using ELISA. Opsonophagocytic killing assay was performed and experimental challenge was done by intraperitoneal injection with sub lethal doses of MRSA in mice and also survival rate was regularly monitored. Results showed that vaccinated mice could exhibit higher levels of autolysin-specific antibodies (P<0.0001) with a predominant IgG1 response versus control group. Results from in vitro experiments indicated that S. aureus opsonized with immunized-mice sera displayed significantly increased phagocytic uptake and effective intracellular killing versus non-immunized mice. The number of viable bacteria in the kidney of immunized mice showed 1000 times less than the control mice; additionally, an increased survival rate was found after immunization with the candidate vaccine versus control group. Results from this study demonstrated that the autolysin is a valuable target for the development of immunotherapeutic strategies against S. aureus and candidate vaccines.
Collapse
Affiliation(s)
- Setareh Haghighat
- Department of Microbiology, Faculty of Advanced Sciences and Technology, Pharmaceutical Sciences Branch, Islamic Azad University (IAUPS), Tehran, Iran.
| | - Seyed Davar Siadat
- Department of Mycobacteriology & Pulmonary Research, Microbiology Research center, Pasteur Institute of Iran, Tehran, Iran.
| | - Seyed Mehdi Rezayat Sorkhabadi
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Pharmaceutical Sciences Branch, Islamic Azad University (IAUPS), Tehran, Iran
| | - Abbas Akhavan Sepahi
- Department of Microbiology, Faculty of Basic Sciences, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Mehdi Mahdavi
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
40
|
Excreted Cytoplasmic Proteins Contribute to Pathogenicity in Staphylococcus aureus. Infect Immun 2016; 84:1672-81. [PMID: 27001537 DOI: 10.1128/iai.00138-16] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2016] [Accepted: 03/13/2016] [Indexed: 11/20/2022] Open
Abstract
Excretion of cytoplasmic proteins in pro- and eukaryotes, also referred to as "nonclassical protein export," is a well-known phenomenon. However, comparatively little is known about the role of the excreted proteins in relation to pathogenicity. Here, the impact of two excreted glycolytic enzymes, aldolase (FbaA) and glyceraldehyde-3-phosphate dehydrogenase (GAPDH), on pathogenicity was investigated in Staphylococcus aureus Both enzymes bound to certain host matrix proteins and enhanced adherence of the bacterial cells to host cells but caused a decrease in host cell invasion. FbaA and GAPDH also bound to the cell surfaces of staphylococcal cells by interaction with the major autolysin, Atl, that is involved in host cell internalization. Surprisingly, FbaA showed high cytotoxicity to both MonoMac 6 (MM6) and HaCaT cells, while GAPDH was cytotoxic only for MM6 cells. Finally, the contribution of external FbaA and GAPDH to S. aureus pathogenicity was confirmed in an insect infection model.
Collapse
|
41
|
Effects of Low-Dose Amoxicillin on Staphylococcus aureus USA300 Biofilms. Antimicrob Agents Chemother 2016; 60:2639-51. [PMID: 26856828 DOI: 10.1128/aac.02070-15] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 01/31/2016] [Indexed: 01/01/2023] Open
Abstract
Previous studies showed that sub-MIC levels of β-lactam antibiotics stimulate biofilm formation in most methicillin-resistant Staphylococcus aureus (MRSA) strains. Here, we investigated this process by measuring the effects of sub-MIC amoxicillin on biofilm formation by the epidemic community-associated MRSA strain USA300. We found that sub-MIC amoxicillin increased the ability of USA300 cells to attach to surfaces and form biofilms under both static and flow conditions. We also found that USA300 biofilms cultured in sub-MIC amoxicillin were thicker, contained more pillar and channel structures, and were less porous than biofilms cultured without antibiotic. Biofilm formation in sub-MIC amoxicillin correlated with the production of extracellular DNA (eDNA). However, eDNA released by amoxicillin-induced cell lysis alone was evidently not sufficient to stimulate biofilm. Sub-MIC levels of two other cell wall-active agents with different mechanisms of action-d-cycloserine and fosfomycin-also stimulated eDNA-dependent biofilm, suggesting that biofilm formation may be a mechanistic adaptation to cell wall stress. Screening a USA300 mariner transposon library for mutants deficient in biofilm formation in sub-MIC amoxicillin identified numerous known mediators of S. aureus β-lactam resistance and biofilm formation, as well as novel genes not previously associated with these phenotypes. Our results link cell wall stress and biofilm formation in MRSA and suggest that eDNA-dependent biofilm formation by strain USA300 in low-dose amoxicillin is an inducible phenotype that can be used to identify novel genes impacting MRSA β-lactam resistance and biofilm formation.
Collapse
|
42
|
Moriarty TF, Kuehl R, Coenye T, Metsemakers WJ, Morgenstern M, Schwarz EM, Riool M, Zaat SA, Khana N, Kates SL, Richards RG. Orthopaedic device-related infection: current and future interventions for improved prevention and treatment. EFORT Open Rev 2016; 1:89-99. [PMID: 28461934 PMCID: PMC5367564 DOI: 10.1302/2058-5241.1.000037] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Orthopaedic and trauma device-related infection (ODRI) remains one of the major complications in modern trauma and orthopaedic surgery.Despite best practice in medical and surgical management, neither prophylaxis nor treatment of ODRI is effective in all cases, leading to infections that negatively impact clinical outcome and significantly increase healthcare expenditure.The following review summarises the microbiological profile of modern ODRI, the impact antibiotic resistance has on treatment outcomes, and some of the principles and weaknesses of the current systemic and local antibiotic delivery strategies.The emerging novel strategies aimed at preventing or treating ODRI will be reviewed. Particular attention will be paid to the potential for clinical impact in the coming decades, when such interventions are likely to be critically important.The review focuses on this problem from an interdisciplinary perspective, including basic science innovations and best practice in infectious disease. Cite this article: Moriarty TF, Kuehl R, Coenye T, et al. Orthopaedic device related infection: current and future interventions for improved prevention and treatment. EFORT Open Rev 2016;1:89-99. DOI: 10.1302/2058-5241.1.000037.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Nina Khana
- University Hospital of Basel, Switzerland
| | | | | |
Collapse
|
43
|
Wilmes M, Stockem M, Bierbaum G, Schlag M, Götz F, Tran DQ, Schaal JB, Ouellette AJ, Selsted ME, Sahl HG. Killing of staphylococci by θ-defensins involves membrane impairment and activation of autolytic enzymes. Antibiotics (Basel) 2016; 3:617-31. [PMID: 25632351 PMCID: PMC4306331 DOI: 10.3390/antibiotics3040617] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
θ-Defensins are cyclic antimicrobial peptides expressed in leukocytes of Old world monkeys. To get insight into their antibacterial mode of action, we studied the activity of RTDs (rhesus macaque θ-defensins) against staphylococci. We found that in contrast to other defensins, RTDs do not interfere with peptidoglycan biosynthesis, but rather induce bacterial lysis in staphylococci by interaction with the bacterial membrane and/or release of cell wall lytic enzymes. Potassium efflux experiments and membrane potential measurements revealed that the membrane impairment by RTDs strongly depends on the energization of the membrane. In addition, RTD treatment caused the release of Atl-derived cell wall lytic enzymes probably by interaction with membrane-bound lipoteichoic acid. Thus, the premature and uncontrolled activity of these enzymes contributes strongly to the overall killing by θ-defensins. Interestingly, a similar mode of action has been described for Pep5, an antimicrobial peptide of bacterial origin.
Collapse
Affiliation(s)
- Miriam Wilmes
- Institute of Medical Microbiology, Immunology and Parasitology, University of Bonn, 53105 Bonn, Germany; E-Mails: (M.S.); (G.B.); (H.-G.S.)
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +49-228-28711397
| | - Marina Stockem
- Institute of Medical Microbiology, Immunology and Parasitology, University of Bonn, 53105 Bonn, Germany; E-Mails: (M.S.); (G.B.); (H.-G.S.)
| | - Gabriele Bierbaum
- Institute of Medical Microbiology, Immunology and Parasitology, University of Bonn, 53105 Bonn, Germany; E-Mails: (M.S.); (G.B.); (H.-G.S.)
| | - Martin Schlag
- Interfaculty Institute of Microbiology and Infection Medicine, Microbial Genetics, University of Tübingen, 72076 Tübingen, Germany; E-Mails: (M.S.); (F.G.)
| | - Friedrich Götz
- Interfaculty Institute of Microbiology and Infection Medicine, Microbial Genetics, University of Tübingen, 72076 Tübingen, Germany; E-Mails: (M.S.); (F.G.)
| | - Dat Q. Tran
- Department of Pathology and Laboratory Medicine, USC Norris Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089-9601, USA; E-Mails: (D.Q.T.); (J.B.S.); (A.J.O.); (M.E.S.)
| | - Justin B. Schaal
- Department of Pathology and Laboratory Medicine, USC Norris Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089-9601, USA; E-Mails: (D.Q.T.); (J.B.S.); (A.J.O.); (M.E.S.)
| | - André J. Ouellette
- Department of Pathology and Laboratory Medicine, USC Norris Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089-9601, USA; E-Mails: (D.Q.T.); (J.B.S.); (A.J.O.); (M.E.S.)
| | - Michael E. Selsted
- Department of Pathology and Laboratory Medicine, USC Norris Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089-9601, USA; E-Mails: (D.Q.T.); (J.B.S.); (A.J.O.); (M.E.S.)
| | - Hans-Georg Sahl
- Institute of Medical Microbiology, Immunology and Parasitology, University of Bonn, 53105 Bonn, Germany; E-Mails: (M.S.); (G.B.); (H.-G.S.)
| |
Collapse
|
44
|
Chan YGY, Frankel MB, Missiakas D, Schneewind O. SagB Glucosaminidase Is a Determinant of Staphylococcus aureus Glycan Chain Length, Antibiotic Susceptibility, and Protein Secretion. J Bacteriol 2016; 198:1123-36. [PMID: 26811319 PMCID: PMC4800868 DOI: 10.1128/jb.00983-15] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 01/20/2016] [Indexed: 01/18/2023] Open
Abstract
UNLABELLED The envelope of Staphylococcus aureus is comprised of peptidoglycan and its attached secondary polymers, teichoic acid, capsular polysaccharide, and protein. Peptidoglycan synthesis involves polymerization of lipid II precursors into glycan strands that are cross-linked at wall peptides. It is not clear whether peptidoglycan structure is principally determined during polymerization or whether processive enzymes affect cell wall structure and function, for example, by generating conduits for protein secretion. We show here that S. aureus lacking SagB, a membrane-associated N-acetylglucosaminidase, displays growth and cell-morphological defects caused by the exaggerated length of peptidoglycan strands. SagB cleaves polymerized glycan strands to their physiological length and modulates antibiotic resistance in methicillin-resistant S. aureus (MRSA). Deletion of sagB perturbs protein trafficking into and across the envelope, conferring defects in cell wall anchoring and secretion, as well as aberrant excretion of cytoplasmic proteins. IMPORTANCE Staphylococcus aureus is thought to secrete proteins across the plasma membrane via the Sec pathway; however, protein transport across the cell wall envelope has heretofore not been studied. We report that S. aureus sagB mutants generate elongated peptidoglycan strands and display defects in protein secretion as well as aberrant excretion of cytoplasmic proteins. These results suggest that the thick peptidoglycan layer of staphylococci presents a barrier for protein secretion and that SagB appears to extend the Sec pathway across the cell wall envelope.
Collapse
Affiliation(s)
- Yvonne G Y Chan
- Department of Microbiology, University of Chicago, Chicago, Illinois, USA
| | - Matthew B Frankel
- Department of Microbiology, University of Chicago, Chicago, Illinois, USA
| | - Dominique Missiakas
- Department of Microbiology, University of Chicago, Chicago, Illinois, USA Howard Taylor Ricketts Laboratory, Argonne National Laboratory, Argonne, Illinois, USA
| | - Olaf Schneewind
- Department of Microbiology, University of Chicago, Chicago, Illinois, USA Howard Taylor Ricketts Laboratory, Argonne National Laboratory, Argonne, Illinois, USA
| |
Collapse
|
45
|
Antibacterial Activity and Antibiotic-Enhancing Effects of Honeybee Venom against Methicillin-Resistant Staphylococcus aureus. Molecules 2016; 21:79. [PMID: 26771592 PMCID: PMC6273778 DOI: 10.3390/molecules21010079] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2015] [Revised: 01/04/2016] [Accepted: 01/08/2016] [Indexed: 01/23/2023] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA), along with other antibiotic resistant bacteria, has become a significant social and clinical problem. There is thus an urgent need to develop naturally bioactive compounds as alternatives to the few antibiotics that remain effective. Here we assessed the in vitro activities of bee venom (BV), alone or in combination with ampicillin, penicillin, gentamicin or vancomycin, on growth of MRSA strains. The antimicrobial activity of BV against MRSA strains was investigated using minimum inhibitory concentrations (MIC), minimum bactericidal concentrations (MBC) and a time-kill assay. Expression of atl which encodes murein hydrolase, a peptidoglycan-degrading enzyme involved in cell separation, was measured by reverse transcription-polymerase chain reaction. The MICs of BV were 0.085 µg/mL and 0.11 µg/mL against MRSA CCARM 3366 and MRSA CCARM 3708, respectively. The MBC of BV against MRSA 3366 was 0.106 µg/mL and that against MRSA 3708 was 0.14 µg/mL. The bactericidal activity of BV corresponded to a decrease of at least 3 log CFU/g cells. The combination of BV with ampicillin or penicillin yielded an inhibitory concentration index ranging from 0.631 to 1.002, indicating a partial and indifferent synergistic effect. Compared to ampicillin or penicillin, both MRSA strains were more susceptible to the combination of BV with gentamicin or vancomycin. The expression of atl gene was increased in MRSA 3366 treated with BV. These results suggest that BV exhibited antibacterial activity and antibiotic-enhancing effects against MRSA strains. The atl gene was increased in MRSA exposed to BV, suggesting that cell division was interrupted. BV warrants further investigation as a natural antimicrobial agent and synergist of antibiotic activity.
Collapse
|
46
|
Monteiro JM, Fernandes PB, Vaz F, Pereira AR, Tavares AC, Ferreira MT, Pereira PM, Veiga H, Kuru E, VanNieuwenhze MS, Brun YV, Filipe SR, Pinho MG. Cell shape dynamics during the staphylococcal cell cycle. Nat Commun 2015; 6:8055. [PMID: 26278781 PMCID: PMC4557339 DOI: 10.1038/ncomms9055] [Citation(s) in RCA: 152] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Accepted: 07/13/2015] [Indexed: 12/04/2022] Open
Abstract
Staphylococcus aureus is an aggressive pathogen and a model organism to study cell division in sequential orthogonal planes in spherical bacteria. However, the small size of staphylococcal cells has impaired analysis of changes in morphology during the cell cycle. Here we use super-resolution microscopy and determine that S. aureus cells are not spherical throughout the cell cycle, but elongate during specific time windows, through peptidoglycan synthesis and remodelling. Both peptidoglycan hydrolysis and turgor pressure are required during division for reshaping the flat division septum into a curved surface. In this process, the septum generates less than one hemisphere of each daughter cell, a trait we show is common to other cocci. Therefore, cell surface scars of previous divisions do not divide the cells in quadrants, generating asymmetry in the daughter cells. Our results introduce a need to reassess the models for division plane selection in cocci. Staphylococci are spherical bacteria that divide in sequential orthogonal planes. Here, the authors use super-resolution microscopy to show that staphylococcal cells elongate before dividing, and that the division septum generates less than one hemisphere of each daughter cell, generating asymmetry.
Collapse
Affiliation(s)
- João M Monteiro
- Laboratory of Bacterial Cell Biology, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, 2780-157 Oeiras, Portugal
| | - Pedro B Fernandes
- Laboratory of Bacterial Cell Biology, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, 2780-157 Oeiras, Portugal
| | - Filipa Vaz
- Laboratory of Bacterial Cell Surfaces and Pathogenesis, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, 2780-157 Oeiras, Portugal
| | - Ana R Pereira
- Laboratory of Bacterial Cell Biology, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, 2780-157 Oeiras, Portugal
| | - Andreia C Tavares
- Laboratory of Bacterial Cell Biology, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, 2780-157 Oeiras, Portugal
| | - Maria T Ferreira
- Laboratory of Bacterial Cell Biology, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, 2780-157 Oeiras, Portugal
| | - Pedro M Pereira
- Laboratory of Bacterial Cell Biology, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, 2780-157 Oeiras, Portugal
| | - Helena Veiga
- Laboratory of Bacterial Cell Biology, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, 2780-157 Oeiras, Portugal
| | - Erkin Kuru
- 1] Department of Chemistry, Indiana University Bloomington, Bloomington, Indiana 47405, USA [2] Department of Biology, Indiana University Bloomington, Bloomington, Indiana 47405, USA
| | | | - Yves V Brun
- Department of Biology, Indiana University Bloomington, Bloomington, Indiana 47405, USA
| | - Sérgio R Filipe
- Laboratory of Bacterial Cell Surfaces and Pathogenesis, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, 2780-157 Oeiras, Portugal
| | - Mariana G Pinho
- Laboratory of Bacterial Cell Biology, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, 2780-157 Oeiras, Portugal
| |
Collapse
|
47
|
Zheng L, Yan M, Fan F, Ji Y. The Essential WalK Histidine Kinase and WalR Regulator Differentially Mediate Autolysis of Staphylococcus aureus RN4220. JOURNAL OF NATURE AND SCIENCE 2015; 1:e111. [PMID: 26052549 PMCID: PMC4457336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
The two-component regulatory system, WalR/WalK is necessary for growth of different gram-positive bacteria, including Staphylococcus aureus. In present study, we confirmed the essentiality of both the histidine kinase protein WalK and the response regulator WalR for growth using S. aureus RN4220 strain and demonstrated that the histidine kinase protein WalK and the response regulator WalR function differently in regulation of staphylococcal autolysis. The down-regulation of walR expression effectively inhibited Triton X-100-induced lysis and had a weak impact on bacterial tolerance to penicillin induced cell lysis. In contrast, the down-regulation of walK expression had no influence on either Triton X-100- or penicillin-caused autolysis. Moreover, we determined the effect of WalR and WalK on bacterial hydrolase activity using a zymogram analysis. The results showed that the cell lysate of down-regulated walR expression mutant displayed several bands of decreased cell wall hydrolytic activities; however, the down-regulation of WalK had no dramatic impact on the hydrolytic activities. Furthermore, we examined the impact of WalR on the transcription of cidA associated with staphylococcal autolysis, and the results showed that the down-regulation of WalR led to decreased transcription of cidA in the log phase of growth. Taken together, the above results suggest that the essential WalR response regulator and the essential WalK histidine kinase might differently control bacterial lysis in RN4220 strain.
Collapse
Affiliation(s)
- Li Zheng
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota. 1971 Commonwealth Ave. St. Paul. MN 55108, USA
| | - Meiying Yan
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota. 1971 Commonwealth Ave. St. Paul. MN 55108, USA
| | - Frank Fan
- Promega Corporation, Madison, WI 53711, USA
| | - Yinduo Ji
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota. 1971 Commonwealth Ave. St. Paul. MN 55108, USA
| |
Collapse
|
48
|
Zhou X, Halladin DK, Rojas ER, Koslover EF, Lee TK, Huang KC, Theriot JA. Bacterial division. Mechanical crack propagation drives millisecond daughter cell separation in Staphylococcus aureus. Science 2015; 348:574-8. [PMID: 25931560 DOI: 10.1126/science.aaa1511] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
When Staphylococcus aureus undergoes cytokinesis, it builds a septum, generating two hemispherical daughters whose cell walls are only connected via a narrow peripheral ring. We found that resolution of this ring occurred within milliseconds ("popping"), without detectable changes in cell volume. The likelihood of popping depended on cell-wall stress, and the separating cells split open asymmetrically, leaving the daughters connected by a hinge. An elastostatic model of the wall indicated high circumferential stress in the peripheral ring before popping. Last, we observed small perforations in the peripheral ring that are likely initial points of mechanical failure. Thus, the ultrafast daughter cell separation in S. aureus appears to be driven by accumulation of stress in the peripheral ring and exhibits hallmarks of mechanical crack propagation.
Collapse
Affiliation(s)
- Xiaoxue Zhou
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA. Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305, USA. Howard Hughes Medical Institute (HHMI), Stanford University School of Medicine, Stanford, CA 94305, USA
| | - David K Halladin
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305, USA. Howard Hughes Medical Institute (HHMI), Stanford University School of Medicine, Stanford, CA 94305, USA. Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Enrique R Rojas
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305, USA. Howard Hughes Medical Institute (HHMI), Stanford University School of Medicine, Stanford, CA 94305, USA. Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Elena F Koslover
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305, USA. Howard Hughes Medical Institute (HHMI), Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Timothy K Lee
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Kerwyn Casey Huang
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA. Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Julie A Theriot
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA 94305, USA. Howard Hughes Medical Institute (HHMI), Stanford University School of Medicine, Stanford, CA 94305, USA. Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
49
|
van den Berg S, Koedijk DGAM, Back JW, Neef J, Dreisbach A, van Dijl JM, Bakker-Woudenberg IAJM, Buist G. Active immunization with an octa-valent Staphylococcus aureus antigen mixture in models of S. aureus bacteremia and skin infection in mice. PLoS One 2015; 10:e0116847. [PMID: 25710376 PMCID: PMC4339199 DOI: 10.1371/journal.pone.0116847] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 12/15/2014] [Indexed: 11/18/2022] Open
Abstract
Proteomic studies with different Staphylococcus aureus isolates have shown that the cell surface-exposed and secreted proteins IsaA, LytM, Nuc, the propeptide of Atl (pro-Atl) and four phenol-soluble modulins α (PSMα) are invariantly produced by this pathogen. Therefore the present study was aimed at investigating whether these proteins can be used for active immunization against S. aureus infection in mouse models of bacteremia and skin infection. To this end, recombinant His-tagged fusions of IsaA, LytM, Nuc and pro-Atl were isolated from Lactococcus lactis or Escherichia coli, while the PSMα1-4 peptides were chemically synthesized. Importantly, patients colonized by S. aureus showed significant immunoglobulin G (IgG) responses against all eight antigens. BALB/cBYJ mice were immunized subcutaneously with a mixture of the antigens at day one (5 μg each), and boosted twice (25 μg of each antigen) with 28 days interval. This resulted in high IgG responses against all antigens although the response against pro-Atl was around one log lower compared to the other antigens. Compared to placebo-immunized mice, immunization with the octa-valent antigen mixture did not reduce the S. aureus isolate P load in blood, lungs, spleen, liver, and kidneys in a bacteremia model in which the animals were challenged for 14 days with a primary load of 3 × 105 CFU. Discomfort scores and animal survival rates over 14 days did not differ between immunized mice and placebo-immunized mice upon bacteremia with S. aureus USA300 (6 × 105 CFU). In addition, this immunization did not reduce the S. aureus isolate P load in mice with skin infection. These results show that the target antigens are immunogenic in both humans and mice, but in the used animal models do not result in protection against S. aureus infection.
Collapse
Affiliation(s)
- Sanne van den Berg
- Department of Medical Microbiology and Infectious Diseases, Erasmus University Medical Center, Rotterdam, the Netherlands
- * E-mail:
| | - Dennis G. A. M. Koedijk
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | | | - Jolanda Neef
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Annette Dreisbach
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Jan Maarten van Dijl
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | | | - Girbe Buist
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
50
|
McCarthy H, Rudkin JK, Black NS, Gallagher L, O'Neill E, O'Gara JP. Methicillin resistance and the biofilm phenotype in Staphylococcus aureus. Front Cell Infect Microbiol 2015; 5:1. [PMID: 25674541 PMCID: PMC4309206 DOI: 10.3389/fcimb.2015.00001] [Citation(s) in RCA: 240] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 01/05/2015] [Indexed: 12/05/2022] Open
Abstract
Antibiotic resistance and biofilm-forming capacity contribute to the success of Staphylococcus aureus as a human pathogen in both healthcare and community settings. These virulence factors do not function independently of each other and the biofilm phenotype expressed by clinical isolates of S. aureus is influenced by acquisition of the methicillin resistance gene mecA. Methicillin-sensitive S. aureus (MSSA) strains commonly produce an icaADBC operon-encoded polysaccharide intercellular adhesin (PIA)-dependent biofilm. In contrast, the release of extracellular DNA (eDNA) and cell surface expression of a number of sortase-anchored proteins, and the major autolysin have been implicated in the biofilm phenotype of methicillin-resistant S. aureus (MRSA) isolates. Expression of high level methicillin resistance in a laboratory MSSA strain resulted in (i) repression of PIA-mediated biofilm production, (ii) down-regulation of the accessory gene regulator (Agr) system, and (iii) attenuation of virulence in murine sepsis and device infection models. Here we review the mechanisms of MSSA and MRSA biofilm production and the relationships between antibiotic resistance, biofilm and virulence gene regulation in S. aureus.
Collapse
Affiliation(s)
- Hannah McCarthy
- Department of Microbiology, School of Natural Sciences, National University of Ireland Galway, Ireland
| | - Justine K Rudkin
- Department of Microbiology, School of Natural Sciences, National University of Ireland Galway, Ireland
| | - Nikki S Black
- Department of Microbiology, School of Natural Sciences, National University of Ireland Galway, Ireland
| | - Laura Gallagher
- Department of Microbiology, School of Natural Sciences, National University of Ireland Galway, Ireland
| | - Eoghan O'Neill
- Department of Clinical Microbiology, Royal College of Surgeons in Ireland Dublin, Ireland
| | - James P O'Gara
- Department of Microbiology, School of Natural Sciences, National University of Ireland Galway, Ireland
| |
Collapse
|