1
|
Zhu S, Hu C, Wang Y, Jin M, Zhang Q, Han S, Tang Y, Wu D, Fu D, Jiang S, Song D, Wei L, Song W, Zhang C, Zhang W. Daphnetin weakened the pathogenicity of methicillin-resistant Staphylococcus aureus by inhibiting Sortase A and α-hemolysin. Biochimie 2024:S0300-9084(24)00236-0. [PMID: 39424258 DOI: 10.1016/j.biochi.2024.10.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 07/31/2024] [Accepted: 10/16/2024] [Indexed: 10/21/2024]
Abstract
The increasing prevalence of antibiotic-resistant bacteria, represented by Methicillin-resistant Staphylococcus aureus (MRSA), has necessitated a shift towards anti-virulence strategies in treatment approaches. This research demonstrated that daphnetin effectively disrupted MRSA virulence by targeting Sortase A (SrtA), an enzyme in Staphylococcus aureus (S. aureus) responsible for adhesion and invasion, as well as the toxin α-hemolysin (Hla) that leads to cell lysis. Utilizing Fluorescence Resonance Energy Transfer, daphnetin showed direct inhibitory effect on SrtA activity, with an IC50 of 25.98 μg/mL. Additionally, daphnetin hindered various SrtA-mediated processes in S. aureus, such as fibronectin adherence, A549 cell invasion, biofilm formation, and bacterial motility. Daphnetin inhibited S. aureus-induced hemolysis and reduced Hla expression as confirmed by Western blot analysis. Molecular docking studies identified specific binding sites of daphnetin with SrtA, highlighting key amino acid residues like GLU-77, TYR-75, and LYS-145, with a docking score of -7.139 kcal/mol. Besides that, daphnetin exhibited a protective effect on MRSA-induced pneumonia in vivo. In summary, daphnetin, a natural compound, effectively inhibited SrtA and Hla activities, attenuating MRSA virulence and showcasing potential for treating bacterial infections.
Collapse
Affiliation(s)
- Shuyue Zhu
- Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Chunjie Hu
- Proctology Department, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Yan Wang
- Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Mengli Jin
- Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Qiuyue Zhang
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, 130021, China
| | - Shaoyu Han
- Shanghai Lixin University of Accounting and Finance, Shanghai 201209, China
| | - Yating Tang
- Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Desheng Wu
- Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Di Fu
- Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Shuang Jiang
- Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Danning Song
- Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Lin Wei
- Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Wu Song
- Changchun University of Chinese Medicine, Changchun, 130117, China.
| | - Chi Zhang
- Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Wenfeng Zhang
- Changchun University of Chinese Medicine, Changchun, 130117, China
| |
Collapse
|
2
|
Ilmain JK, Perelman SS, Panepinto MC, Irnov I, Coudray N, Samhadaneh N, Pironti A, Ueberheide B, Ekiert DC, Bhabha G, Torres VJ. Unlatching of the stem domains in the Staphylococcus aureus pore-forming leukocidin LukAB influences toxin oligomerization. J Biol Chem 2023; 299:105321. [PMID: 37802313 PMCID: PMC10665946 DOI: 10.1016/j.jbc.2023.105321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 09/22/2023] [Accepted: 09/27/2023] [Indexed: 10/08/2023] Open
Abstract
Staphylococcus aureus (S. aureus) is a serious global pathogen that causes a diverse range of invasive diseases. S. aureus utilizes a family of pore-forming toxins, known as bi-component leukocidins, to evade the host immune response and promote infection. Among these is LukAB (leukocidin A/leukocidin B), a toxin that assembles into an octameric β-barrel pore in the target cell membrane, resulting in host cell death. The established cellular receptor for LukAB is CD11b of the Mac-1 complex. Here, we show that hydrogen voltage-gated channel 1 is also required for the cytotoxicity of all major LukAB variants. We demonstrate that while each receptor is sufficient to recruit LukAB to the plasma membrane, both receptors are required for maximal lytic activity. Why LukAB requires two receptors, and how each of these receptors contributes to pore-formation remains unknown. To begin to resolve this, we performed an alanine scanning mutagenesis screen to identify mutations that allow LukAB to maintain cytotoxicity without CD11b. We discovered 30 mutations primarily localized in the stem domains of LukA and LukB that enable LukAB to exhibit full cytotoxicity in the absence of CD11b. Using crosslinking, electron microscopy, and hydroxyl radical protein footprinting, we show these mutations increase the solvent accessibility of the stem domain, priming LukAB for oligomerization. Together, our data support a model in which CD11b binding unlatches the membrane penetrating stem domains of LukAB, and this change in flexibility promotes toxin oligomerization.
Collapse
Affiliation(s)
- Juliana K Ilmain
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Sofya S Perelman
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Maria C Panepinto
- Proteomics Laboratory, Division of Advanced Research Technologies, New York University Grossman School of Medicine, New York, New York, USA
| | - Irnov Irnov
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Nicolas Coudray
- Applied Bioinformatics Laboratories, New York University Grossman School of Medicine, New York, New York, USA; Department of Cell Biology, New York University Grossman School of Medicine, New York, New York, USA
| | - Nora Samhadaneh
- Antimicrobial-Resistant Pathogens Program, New York University Langone Health, New York, New York, USA
| | - Alejandro Pironti
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA; Antimicrobial-Resistant Pathogens Program, New York University Langone Health, New York, New York, USA
| | - Beatrix Ueberheide
- Proteomics Laboratory, Division of Advanced Research Technologies, New York University Grossman School of Medicine, New York, New York, USA; Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, New York, USA; Department of Neurology, Center for Cognitive Neurology, New York University Grossman School of Medicine, New York, New York, USA
| | - Damian C Ekiert
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA; Department of Cell Biology, New York University Grossman School of Medicine, New York, New York, USA; Antimicrobial-Resistant Pathogens Program, New York University Langone Health, New York, New York, USA
| | - Gira Bhabha
- Department of Cell Biology, New York University Grossman School of Medicine, New York, New York, USA; Antimicrobial-Resistant Pathogens Program, New York University Langone Health, New York, New York, USA
| | - Victor J Torres
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, USA; Department of Host-Microbe Interactions, St Jude Children's Research Hospital, Memphis, Tennessee, USA.
| |
Collapse
|
3
|
Yu S, Zhou Y, Feng D, Jiang Q, Li T, Jiang G, Zhou Z, Li H. Whole genome sequence-based characterization of virulence and antimicrobial resistance gene profiles of Staphylococcus aureus isolated from food poisoning incidents in eastern China. Front Microbiol 2023; 14:1225472. [PMID: 37795294 PMCID: PMC10546991 DOI: 10.3389/fmicb.2023.1225472] [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: 05/19/2023] [Accepted: 09/04/2023] [Indexed: 10/06/2023] Open
Abstract
Staphylococcus aureus is an opportunistic foodborne pathogen occasionally isolated from diarrhea patients. In recent years, increasing studies have reported the detection of S. aureus in food poisoning incidents due to food contamination in the North and South of China. However, the epidemiology and genetic characteristics of S. aureus from food poisoning incidents in Eastern China remain unknown. The present study examined the genetic characteristics, antimicrobial resistance, and virulent factors of multidrug-resistant S. aureus isolated from 22 food poisoning incidents reported by the hospitals and health centers in Eastern China from 2011 to 2021. A total of 117 resistant and enterotoxigenic S. aureus isolates were collected and sequenced, among which 20 isolates were identified as methicillin resistant. Genetic analysis revealed 19 distinct CC/ST types, with CC6, CC22, CC59, CC88, and CC398 being the most frequent variants in methicillin-resistant S. aureus (MRSA). A considerable shift in CC types from CC1 to CC398 between 2011 and 2021 was observed in this study, indicating that CC398 may be the main epidemic strain circulating in the current food poisoning incidents. Additionally, genes for enterotoxins were detected in 55 isolates, with a prevalence of 27.8% (27/97) for methicillin-sensitive variants and 35.0% (7/20) for MRSA. The scn gene was detected in 59.0% of the isolates, demonstrating diverse contaminations of S. aureus among livestock-to-human transmission. Of the 117 isolates, only ten isolates displayed multi-drug resistance (MDR) to penicillin, tetracycline, and macrolides. None of the 117 foodborne S. aureus isolates tested positive for vanA in this study. Together, the present study provided phylogenetic characteristics of S. aureus from food poisoning incidents that emerged in Eastern China from 2011 to 2021. Our results suggested that these diarrhea episodes were hypotonic and merely transient low-MDR infections, however, further research for continued surveillance given the detection of virulence and antimicrobial resistance determinants is required to elucidate the genomic characteristics of pathogenic S. aureus in food poisoning incidents in the context of public health.
Collapse
|
4
|
Ogata A, Hayashi K, Kitano T, Onozaki K, Itoh S, Hida S. Staphylococcal γ-hemolysins induce IL-4 production in murine basophils. Biochem Biophys Res Commun 2022; 632:107-112. [DOI: 10.1016/j.bbrc.2022.09.070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 09/18/2022] [Indexed: 11/02/2022]
|
5
|
Ghanem N, Kanagami N, Matsui T, Takeda K, Kaneko J, Shiraishi Y, Choe CA, Uchikubo‐Kamo T, Shirouzu M, Hashimoto T, Ogawa T, Matsuura T, Huang P, Yokoyama T, Tanaka Y. Chimeric mutants of staphylococcal hemolysin, which act as both one‐component and two‐component hemolysin, created by grafting the stem domain. FEBS J 2022; 289:3505-3520. [DOI: 10.1111/febs.16354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 12/03/2021] [Accepted: 01/12/2022] [Indexed: 11/29/2022]
Affiliation(s)
- Nouran Ghanem
- Graduate School of Life Sciences Tohoku University Sendai Japan
- Laboratory for Protein Functional and Structural Biology RIKEN Center for Biosystems Dynamics Research Yokohama Japan
| | - Natsuki Kanagami
- Graduate School of Life Sciences Tohoku University Sendai Japan
| | - Takashi Matsui
- Graduate School of Life Sciences Tohoku University Sendai Japan
- School of Science Kitasato University Sagamihara Japan
| | - Kein Takeda
- Department of Microbial Biotechnology Graduate School of Agricultural Science Tohoku University Sendai Japan
| | - Jun Kaneko
- Department of Microbial Biotechnology Graduate School of Agricultural Science Tohoku University Sendai Japan
| | - Yasuyuki Shiraishi
- Pre‐Clinical Research Center Institute of Development, Aging and Cancer Tohoku University Sendai Japan
| | | | - Tomomi Uchikubo‐Kamo
- Laboratory for Protein Functional and Structural Biology RIKEN Center for Biosystems Dynamics Research Yokohama Japan
| | - Mikako Shirouzu
- Laboratory for Protein Functional and Structural Biology RIKEN Center for Biosystems Dynamics Research Yokohama Japan
| | | | - Tomohisa Ogawa
- Graduate School of Life Sciences Tohoku University Sendai Japan
- Department of Microbial Biotechnology Graduate School of Agricultural Science Tohoku University Sendai Japan
| | - Tomoaki Matsuura
- Department of Biotechnology Graduate School of Engineering Osaka University Suita Japan
| | - Po‐Ssu Huang
- Department of Bioengineering Stanford University CA USA
| | - Takeshi Yokoyama
- Graduate School of Life Sciences Tohoku University Sendai Japan
- Laboratory for Protein Functional and Structural Biology RIKEN Center for Biosystems Dynamics Research Yokohama Japan
| | - Yoshikazu Tanaka
- Graduate School of Life Sciences Tohoku University Sendai Japan
| |
Collapse
|
6
|
Recent Advances in Aptamer‐Based Nanopore Sensing at Single‐Molecule Resolution. Chem Asian J 2022; 17:e202200364. [DOI: 10.1002/asia.202200364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/20/2022] [Indexed: 11/07/2022]
|
7
|
Popovich J, Chen S, Iannuzo N, Ganser C, Seo DK, Haydel SE. Synthesized Geopolymers Adsorb Bacterial Proteins, Toxins, and Cells. Front Bioeng Biotechnol 2020; 8:527. [PMID: 32582660 PMCID: PMC7283576 DOI: 10.3389/fbioe.2020.00527] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 05/04/2020] [Indexed: 11/30/2022] Open
Abstract
Pore-forming and hemolytic toxins are bacterial cytotoxic proteins required for virulence in many pathogens, including staphylococci and streptococci, and are notably associated with clinical manifestations of disease. Inspired by adsorption properties of naturally occurring aluminosilicates, we engineered inexpensive, laboratory-synthesized, aluminosilicate geopolymers with controllable pore and surface characteristics to remove pathogenic or cytotoxic material from the surrounding environment. In this study, macroporous and mesoporous geopolymers were produced with and without stearic acid surface modifications. Geopolymer binding efficacies were assessed by measuring adsorption of methicillin-resistant Staphylococcus aureus (MRSA) culture filtrate proteins, α-hemolysin and streptolysin-O toxins, MRSA whole cells, and antibiotics. Macroporous and mesoporous geopolymers were strong non-selective adsorbents for bacterial protein, protein toxins, and bacteria. Although some geopolymers adsorbed antibiotics, these synthesized geopolymers could potentially be used in non-selective adsorptive applications and optimized for adsorption of specific biomolecules.
Collapse
Affiliation(s)
- John Popovich
- Center for Immunotherapy, Vaccines and Virotherapy, The Biodesign Institute, Arizona State University, Tempe, AZ, United States
| | - Shaojiang Chen
- School of Molecular Sciences, Arizona State University, Tempe, AZ, United States
| | - Natalie Iannuzo
- School of Molecular Sciences, Arizona State University, Tempe, AZ, United States.,School of Life Sciences, Arizona State University, Tempe, AZ, United States
| | - Collin Ganser
- School of Life Sciences, Arizona State University, Tempe, AZ, United States.,School of Earth and Space Exploration, Arizona State University, Tempe, AZ, United States
| | - Dong-Kyun Seo
- School of Molecular Sciences, Arizona State University, Tempe, AZ, United States.,Center for Molecular Design and Biomimetics, The Biodesign Institute, Arizona State University, Tempe, AZ, United States
| | - Shelley E Haydel
- Center for Immunotherapy, Vaccines and Virotherapy, The Biodesign Institute, Arizona State University, Tempe, AZ, United States.,School of Life Sciences, Arizona State University, Tempe, AZ, United States.,Center for Bioelectronics and Biosensors, The Biodesign Institute, Arizona State University, Tempe, AZ, United States
| |
Collapse
|
8
|
Liu J, Kozhaya L, Torres VJ, Unutmaz D, Lu M. Structure-based discovery of a small-molecule inhibitor of methicillin-resistant Staphylococcus aureus virulence. J Biol Chem 2020; 295:5944-5959. [PMID: 32179646 PMCID: PMC7196633 DOI: 10.1074/jbc.ra120.012697] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 03/06/2020] [Indexed: 01/07/2023] Open
Abstract
The rapid emergence and dissemination of methicillin-resistant Staphylococcus aureus (MRSA) strains poses a major threat to public health. MRSA possesses an arsenal of secreted host-damaging virulence factors that mediate pathogenicity and blunt immune defenses. Panton-Valentine leukocidin (PVL) and α-toxin are exotoxins that create lytic pores in the host cell membrane. They are recognized as being important for the development of invasive MRSA infections and are thus potential targets for antivirulence therapies. Here, we report the high-resolution X-ray crystal structures of both PVL and α-toxin in their soluble, monomeric, and oligomeric membrane-inserted pore states in complex with n-tetradecylphosphocholine (C14PC). The structures revealed two evolutionarily conserved phosphatidylcholine-binding mechanisms and their roles in modulating host cell attachment, oligomer assembly, and membrane perforation. Moreover, we demonstrate that the soluble C14PC compound protects primary human immune cells in vitro against cytolysis by PVL and α-toxin and hence may serve as the basis for the development of an antivirulence agent for managing MRSA infections.
Collapse
Affiliation(s)
- Jie Liu
- Public Health Research Institute, Department of Microbiology, Biochemistry, and Molecular Genetics, New Jersey Medical School, Rutgers University, Newark, New Jersey 07103
| | - Lina Kozhaya
- Jackson Laboratory for Genomic Medicine, Farmington, Connecticut 06032
| | - Victor J. Torres
- Department of Microbiology, New York University School of Medicine, New York, New York 10016
| | - Derya Unutmaz
- Jackson Laboratory for Genomic Medicine, Farmington, Connecticut 06032
| | - Min Lu
- Public Health Research Institute, Department of Microbiology, Biochemistry, and Molecular Genetics, New Jersey Medical School, Rutgers University, Newark, New Jersey 07103, To whom correspondence should be addressed:
Public Health Research Institute, Dept. of Microbiology, Biochemistry, and Molecular Genetics, New Jersey Medical School, Newark, NJ 07103. Tel.:
973-854-3260; E-mail:
| |
Collapse
|
9
|
Barrera EE, Machado MR, Pantano S. Fat SIRAH: Coarse-Grained Phospholipids To Explore Membrane-Protein Dynamics. J Chem Theory Comput 2019; 15:5674-5688. [PMID: 31433946 DOI: 10.1021/acs.jctc.9b00435] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The capability to handle highly heterogeneous molecular assemblies in a consistent manner is among the greatest challenges faced when deriving simulation parameters. This is particularly the case for coarse-grained (CG) simulations in which chemical functional groups are lumped into effective interaction centers for which transferability between different chemical environments is not guaranteed. Here, we introduce the parametrization of a set of CG phospholipids compatible with the latest version of the SIRAH force field for proteins. The newly introduced lipid species include different acylic chain lengths and partial unsaturation, as well as polar and acidic head groups that show a very good reproduction of structural membrane determinants, such as areas per lipid, thickness, order parameter, etc., and their dependence with temperature. Simulation of membrane proteins showed unprecedented accuracy in the unbiased description of the thickness-dependent membrane-protein orientation in systems where this information is experimentally available (namely, the SarcoEndoplasmic Reticulum Calcium-SERCA-pump and its regulator Phospholamban). The interactions that lead to this faithful reproduction can be traced down to the single amino acid-lipid interaction level and show full agreement with biochemical data present in the literature. Finally, the present parametrization is implemented in the GROMACS and AMBER simulation packages facilitating its use by a wide portion of the biocomputing community.
Collapse
Affiliation(s)
- Exequiel E Barrera
- Biomolecular Simulations Group , Institut Pasteur de Montevideo , Mataojo 2020 , CP 11400 Montevideo , Uruguay
| | - Matías R Machado
- Biomolecular Simulations Group , Institut Pasteur de Montevideo , Mataojo 2020 , CP 11400 Montevideo , Uruguay
| | - Sergio Pantano
- Biomolecular Simulations Group , Institut Pasteur de Montevideo , Mataojo 2020 , CP 11400 Montevideo , Uruguay.,Shanghai Institute for Advanced Immunochemical Studies , ShanghaiTech University , Shanghai 201210 , China
| |
Collapse
|
10
|
Ryndak MB, Laal S. Mycobacterium tuberculosis Primary Infection and Dissemination: A Critical Role for Alveolar Epithelial Cells. Front Cell Infect Microbiol 2019; 9:299. [PMID: 31497538 PMCID: PMC6712944 DOI: 10.3389/fcimb.2019.00299] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 08/02/2019] [Indexed: 12/28/2022] Open
Abstract
Globally, tuberculosis (TB) has reemerged as a major cause of morbidity and mortality, despite the use of the Mycobacterium bovis BCG vaccine and intensive attempts to improve upon BCG or develop new vaccines. Two lacunae in our understanding of the Mycobacterium tuberculosis (M. tb)-host pathogenesis have mitigated the vaccine efforts; the bacterial-host interaction that enables successful establishment of primary infection and the correlates of protection against TB. The vast majority of vaccine efforts are based on the premise that cell-mediated immunity (CMI) is the predominating mode of protection against TB. However, studies in animal models and in humans demonstrate that post-infection, a period of several weeks precedes the initiation of CMI during which the few inhaled bacteria replicate dramatically and disseminate systemically. The “Trojan Horse” mechanism, wherein M. tb is phagocytosed and transported across the alveolar barrier by infected alveolar macrophages has been long postulated as the sole, primary M. tb:host interaction. In the current review, we present evidence from our studies of transcriptional profiles of M. tb in sputum as it emerges from infectious patients where the bacteria are in a quiescent state, to its adaptations in alveolar epithelial cells where the bacteria transform to a highly replicative and invasive phenotype, to its maintenance of the invasive phenotype in whole blood to the downregulation of invasiveness upon infection of epithelial cells at an extrapulmonary site. Evidence for this alternative mode of infection and dissemination during primary infection is supported by in vivo, in vitro cell-based, and transcriptional studies from multiple investigators in recent years. The proposed alternative mechanism of primary infection and dissemination across the alveolar barrier parallels our understanding of infection and dissemination of other Gram-positive pathogens across their relevant mucosal barriers in that barrier-specific adhesins, toxins, and enzymes synergize to facilitate systemic establishment of infection prior to the emergence of CMI. Further exploration of this M. tb:non-phagocytic cell interaction can provide alternative approaches to vaccine design to prevent infection with M. tb and not only decrease clinical disease but also decrease the overwhelming reservoir of latent TB infection.
Collapse
Affiliation(s)
- Michelle B Ryndak
- Department of Pathology, New York University School of Medicine, New York, NY, United States
| | - Suman Laal
- Department of Pathology, New York University School of Medicine, New York, NY, United States
| |
Collapse
|
11
|
Raimi AR, Ezeokoli OT, Adeleke RA. High-throughput sequence analysis of bacterial communities in commercial biofertiliser products marketed in South Africa: an independent snapshot quality assessment. 3 Biotech 2019; 9:108. [PMID: 30863692 DOI: 10.1007/s13205-019-1643-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 02/20/2019] [Indexed: 02/03/2023] Open
Abstract
The genetic and predicted functional diversity of bacterial communities in 12 commercial biofertiliser products were evaluated using high-throughput sequencing of the 16S rRNA gene. Proteobacteria, Firmicutes and Bacteroides dominated the bacterial communities, with the genera Pseudomonas, Lactobacillus, Bacillus, Bradyrhizobium and Rhizobium being prevalent. The manufacturer-specified species were detected in relatively high abundance in two of the products while a few or none of the specified species were detected in some products. A number of unspecified microbes were detected, including potential human and crop pathogens such as Alcaligenes, Clostridium, Escherichia-Shigella and Proteus. The functional prediction unravelled high prevalence of enzyme-coding genes such as nitrogenase, NifT, alkaline phosphatase and reductases of nitric oxide, nitrate and nitrite which contribute to nitrogen-fixation, phosphorus solubilisation and degradation of nitrates and nitrites. In addition, toxins such as leukocidin/hemolysin and colicin V protein that cause product quality damage were highly predicted in over 67% of the products. Overall, high-throughput sequence analysis of bacterial communities in biofertiliser products revealed that majority of the products were of poor quality. This result justifies the need for regular quality assessment and improvement in quality control systems during biofertiliser formulation.
Collapse
|
12
|
Characterization of Host and Bacterial Contributions to Lung Barrier Dysfunction Following Co-infection with 2009 Pandemic Influenza and Methicillin Resistant Staphylococcus aureus. Viruses 2019; 11:v11020116. [PMID: 30699912 PMCID: PMC6409999 DOI: 10.3390/v11020116] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 01/26/2019] [Indexed: 12/12/2022] Open
Abstract
Influenza viruses are a threat to global public health resulting in ~500,000 deaths each year. Despite an intensive vaccination program, influenza infections remain a recurrent, yet unsolved public health problem. Secondary bacterial infections frequently complicate influenza infections during seasonal outbreaks and pandemics, resulting in increased morbidity and mortality. Staphylococcus aureus, including methicillin-resistant S. aureus (MRSA), is frequently associated with these co-infections, including the 2009 influenza pandemic. Damage to alveolar epithelium is a major contributor to severe influenza-bacterial co-infections and can result in gas exchange abnormalities, fluid leakage, and respiratory insufficiency. These deleterious manifestations likely involve both pathogen- and host-mediated mechanisms. However, there is a paucity of information regarding the mechanisms (pathogen- and/or host-mediated) underlying influenza-bacterial co-infection pathogenesis. To address this, we characterized the contributions of viral-, bacterial-, and host-mediated factors to the altered structure and function of alveolar epithelial cells during co-infection with a focus on the 2009 pandemic influenza (pdm2009) and MRSA. Here, we characterized pdm2009 and MRSA replication kinetics, temporal host kinome responses, modulation of MRSA virulence factors, and disruption of alveolar barrier integrity in response to pdm2009-MRSA co-infection. Our results suggest that alveolar barrier disruption during co-infection is mediated primarily through host response dysregulation, resulting in loss of alveolar barrier integrity.
Collapse
|
13
|
Divyakolu S, Chikkala R, Ratnakar KS, Sritharan V. Hemolysins of <i>Staphylococcus aureus</i>—An Update on Their Biology, Role in Pathogenesis and as Targets for Anti-Virulence Therapy. ACTA ACUST UNITED AC 2019. [DOI: 10.4236/aid.2019.92007] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
14
|
Tan CS, Fleming AM, Ren H, Burrows CJ, White HS. γ-Hemolysin Nanopore Is Sensitive to Guanine-to-Inosine Substitutions in Double-Stranded DNA at the Single-Molecule Level. J Am Chem Soc 2018; 140:14224-14234. [PMID: 30269492 DOI: 10.1021/jacs.8b08153] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Biological nanopores provide a unique single-molecule sensing platform to detect target molecules based on their specific electrical signatures. The γ-hemolysin (γ-HL) protein produced by Staphylococcus aureus is able to assemble into an octamer nanopore with a ∼2.3 nm diameter β-barrel. Herein, we demonstrate the first application of γ-HL nanopore for DNA structural analysis. To optimize conditions for ion-channel recording, the properties of the γ-HL pore (e.g., conductance, voltage-dependent gating, and ion-selectivity) were characterized at different pH, temperature, and electrolyte concentrations. The optimal condition for DNA analysis using γ-HL corresponds to 3 M KCl, pH 5, and T = 20 °C. The γ-HL protein nanopore is able to translocate dsDNA at about ∼20 bp/ms, and the unique current-signature of captured dsDNA can directly distinguish guanine-to-inosine substitutions at the single-molecule level with ∼99% accuracy. The slow dsDNA threading and translocation processes indicate this wild-type γ-HL channel has potential to detect other base modifications in dsDNA.
Collapse
Affiliation(s)
- Cherie S Tan
- Department of Chemistry , University of Utah , 315 South 1400 East , Salt Lake City , Utah 84112-0850 , United States
| | - Aaron M Fleming
- Department of Chemistry , University of Utah , 315 South 1400 East , Salt Lake City , Utah 84112-0850 , United States
| | - Hang Ren
- Department of Chemistry , University of Utah , 315 South 1400 East , Salt Lake City , Utah 84112-0850 , United States
| | - Cynthia J Burrows
- Department of Chemistry , University of Utah , 315 South 1400 East , Salt Lake City , Utah 84112-0850 , United States
| | - Henry S White
- Department of Chemistry , University of Utah , 315 South 1400 East , Salt Lake City , Utah 84112-0850 , United States
| |
Collapse
|
15
|
Tanaka J, Hirayama F, Yanase S, Uno S, Nakae T, Kamizono A, Hanaki H. Effective concentration of intravenous immunoglobulin for neutralizing Panton-Valentine leukocidin in human blood. J Infect Chemother 2018; 24:383-388. [DOI: 10.1016/j.jiac.2017.12.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 12/01/2017] [Accepted: 12/30/2017] [Indexed: 10/17/2022]
|
16
|
de Wit J, Totté J, van Buchem F, Pasmans S. The prevalence of antibody responses againstStaphylococcus aureusantigens in patients with atopic dermatitis: a systematic review and meta-analysis. Br J Dermatol 2018; 178:1263-1271. [DOI: 10.1111/bjd.16251] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/04/2017] [Indexed: 12/21/2022]
Affiliation(s)
- J. de Wit
- Department of Dermatology; Erasmus MC University Medical Centre; Wytemaweg 80 3015 CN Rotterdam the Netherlands
| | - J.E.E. Totté
- Department of Dermatology; Erasmus MC University Medical Centre; Wytemaweg 80 3015 CN Rotterdam the Netherlands
| | - F.J.M. van Buchem
- Department of Dermatology; Erasmus MC University Medical Centre; Wytemaweg 80 3015 CN Rotterdam the Netherlands
| | - S.G.M.A. Pasmans
- Department of Dermatology; Erasmus MC University Medical Centre; Wytemaweg 80 3015 CN Rotterdam the Netherlands
| |
Collapse
|
17
|
Vaishampayan A, de Jong A, Wight DJ, Kok J, Grohmann E. A Novel Antimicrobial Coating Represses Biofilm and Virulence-Related Genes in Methicillin-Resistant Staphylococcus aureus. Front Microbiol 2018; 9:221. [PMID: 29497410 PMCID: PMC5818464 DOI: 10.3389/fmicb.2018.00221] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 01/30/2018] [Indexed: 12/20/2022] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) has become an important cause of hospital-acquired infections worldwide. It is one of the most threatening pathogens due to its multi-drug resistance and strong biofilm-forming capacity. Thus, there is an urgent need for novel alternative strategies to combat bacterial infections. Recently, we demonstrated that a novel antimicrobial surface coating, AGXX®, consisting of micro-galvanic elements of the two noble metals, silver and ruthenium, surface-conditioned with ascorbic acid, efficiently inhibits MRSA growth. In this study, we demonstrated that the antimicrobial coating caused a significant reduction in biofilm formation (46%) of the clinical MRSA isolate, S. aureus 04-02981. To understand the molecular mechanism of the antimicrobial coating, we exposed S. aureus 04-02981 for different time-periods to the coating and investigated its molecular response via next-generation RNA-sequencing. A conventional antimicrobial silver coating served as a control. RNA-sequencing demonstrated down-regulation of many biofilm-associated genes and of genes related to virulence of S. aureus. The antimicrobial substance also down-regulated the two-component quorum-sensing system agr suggesting that it might interfere with quorum-sensing while diminishing biofilm formation in S. aureus 04-02981.
Collapse
Affiliation(s)
- Ankita Vaishampayan
- Life Sciences and Technology, Beuth University of Applied Sciences Berlin, Berlin, Germany
| | - Anne de Jong
- Department of Molecular Genetics, University of Groningen, Groningen, Netherlands
| | - Darren J. Wight
- Institute of Virology, Free University of Berlin, Berlin, Germany
| | - Jan Kok
- Department of Molecular Genetics, University of Groningen, Groningen, Netherlands
| | - Elisabeth Grohmann
- Life Sciences and Technology, Beuth University of Applied Sciences Berlin, Berlin, Germany
- Division of Infectious Diseases, University Medical Center Freiburg, Freiburg, Germany
| |
Collapse
|
18
|
Humoral immune consequences of Staphylococcus aureus ST239-associated bacteremia. Eur J Clin Microbiol Infect Dis 2017; 37:255-263. [PMID: 29103153 DOI: 10.1007/s10096-017-3124-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 10/16/2017] [Indexed: 01/21/2023]
Abstract
The humoral immune responses against 46 different staphylococcal antigens in 27 bacteremia patients infected by clonally related methicillin-resistant Staphylococcus aureus (MRSA) strains of a single sequence type (ST) 239 were investigated. A group of non-infected patients (n = 31) hospitalized for different reasons served as controls. All strains were confirmed as ST 239 by S. aureus and mecA-specific PCR, spa, and multi-locus sequence typing (MLST). In each bacteremia patient, a unique pattern of S. aureus antigen-specific immune responses after infection was observed. Antibody levels among bacteremia patients were significantly higher than controls for HlgB (P = 0.001), LukD (P = 0.009), LukF (P = 0.0001), SEA (P = 0.0001), SEB (P = 0.011), SEC (P = 0.010), SEQ (P = 0.049), IsaA (P = 0.043), IsdA (P = 0.038), IsdH (P = 0.01), SdrD (P = 0.001), SdrE (P = 0.046), EsxA (P = 0.0001), and SA0104 (P = 0.0001). On the other hand, the antibody levels were significantly higher among controls for SSL3 (P = 0.009), SSL9 (P = 0.002), and SSL10 (P = 0.007) when the IgG level on the day of infection was compared with that measured on the day of admission. Diversity was observed in the immune response against the antigens. However, a set of antigens (IsaA, IsdA, IsdH, SdrD, and HlgB) triggered a similar type of immune response in different individuals. We suggest that these antigens could be considered when developing a multi-component (passive) vaccine. SEA and/or its specific antibodies seem to play a critical role during ST239 MRSA bacteremia and SEA-targeted therapy may be a strategy to be considered.
Collapse
|
19
|
Adamala KP, Martin-Alarcon DA, Guthrie-Honea KR, Boyden ES. Engineering genetic circuit interactions within and between synthetic minimal cells. Nat Chem 2017; 9:431-439. [PMID: 28430194 PMCID: PMC5407321 DOI: 10.1038/nchem.2644] [Citation(s) in RCA: 225] [Impact Index Per Article: 32.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2015] [Accepted: 09/12/2016] [Indexed: 12/14/2022]
Abstract
Genetic circuits and reaction cascades are of great importance for synthetic biology, biochemistry and bioengineering. An open question is how to maximize the modularity of their design to enable the integration of different reaction networks and to optimize their scalability and flexibility. One option is encapsulation within liposomes, which enables chemical reactions to proceed in well-isolated environments. Here we adapt liposome encapsulation to enable the modular, controlled compartmentalization of genetic circuits and cascades. We demonstrate that it is possible to engineer genetic circuit-containing synthetic minimal cells (synells) to contain multiple-part genetic cascades, and that these cascades can be controlled by external signals as well as inter-liposomal communication without crosstalk. We also show that liposomes that contain different cascades can be fused in a controlled way so that the products of incompatible reactions can be brought together. Synells thus enable a more modular creation of synthetic biology cascades, an essential step towards their ultimate programmability.
Collapse
Affiliation(s)
| | - Daniel A. Martin-Alarcon
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | | | - Edward S. Boyden
- Media Lab, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| |
Collapse
|
20
|
Trouillet-Assant S, Lelièvre L, Martins-Simões P, Gonzaga L, Tasse J, Valour F, Rasigade JP, Vandenesch F, Muniz Guedes RL, Ribeiro de Vasconcelos AT, Caillon J, Lustig S, Ferry T, Jacqueline C, Loss de Morais G, Laurent F. Adaptive processes of Staphylococcus aureus isolates during the progression from acute to chronic bone and joint infections in patients. Cell Microbiol 2016; 18:1405-14. [PMID: 26918656 DOI: 10.1111/cmi.12582] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 02/20/2016] [Indexed: 11/28/2022]
Abstract
Staphylococcus aureus bone and joint infection (BJI) is associated with significant rates of chronicity and relapse. In this study, we investigated how S. aureus is able to adapt to the human environment by comparing isolates from single patients with persisting or relapsing BJIs that were recovered during the initial and recurrent BJI episodes. In vitro and in vivo assays and whole-genome sequencing analyses revealed that the recurrent isolates induced a reduced inflammatory response, formed more biofilms, persisted longer in the intracellular compartments of host bone cells, were less cytotoxic and induced less mortality in a mouse infection model compared with the initial isolates despite the lack of significant changes at the genomic level. These findings suggest that S. aureus BJI chronicization is associated with an in vivo bacterial phenotypical adaptation that leads to decreased virulence and host immune escape, which is linked to increased intraosteoblastic persistence and biofilm formation.
Collapse
Affiliation(s)
- Sophie Trouillet-Assant
- Centre International de Recherche en Infectiologie, INSERM U1111, Pathogenesis of staphylococcal infections, University of Lyon 1, Lyon, France. .,Department of Clinical Microbiology, Northern Hospital Group, Hospices Civils de Lyon, Lyon, France.
| | - Lucie Lelièvre
- Centre International de Recherche en Infectiologie, INSERM U1111, Pathogenesis of staphylococcal infections, University of Lyon 1, Lyon, France
| | - Patrícia Martins-Simões
- Centre International de Recherche en Infectiologie, INSERM U1111, Pathogenesis of staphylococcal infections, University of Lyon 1, Lyon, France.,Department of Clinical Microbiology, Northern Hospital Group, Hospices Civils de Lyon, Lyon, France
| | - Luiz Gonzaga
- Bioinformatics Laboratory - LABINFO, National Laboratory of Scientific Computation - LNCC/MCTI, Petrópolis, Brazil
| | - Jason Tasse
- Centre International de Recherche en Infectiologie, INSERM U1111, Pathogenesis of staphylococcal infections, University of Lyon 1, Lyon, France.,Department of Clinical Microbiology, Northern Hospital Group, Hospices Civils de Lyon, Lyon, France
| | - Florent Valour
- Centre International de Recherche en Infectiologie, INSERM U1111, Pathogenesis of staphylococcal infections, University of Lyon 1, Lyon, France.,Infectious Diseases Department, Northern Hospital Group, Hospices Civils de Lyon, Lyon, France
| | - Jean-Philippe Rasigade
- Centre International de Recherche en Infectiologie, INSERM U1111, Pathogenesis of staphylococcal infections, University of Lyon 1, Lyon, France.,Department of Clinical Microbiology, Northern Hospital Group, Hospices Civils de Lyon, Lyon, France.,National Reference Center for Staphylococci, Hospices Civils de Lyon, Lyon, France
| | - François Vandenesch
- Centre International de Recherche en Infectiologie, INSERM U1111, Pathogenesis of staphylococcal infections, University of Lyon 1, Lyon, France.,Department of Clinical Microbiology, Northern Hospital Group, Hospices Civils de Lyon, Lyon, France.,National Reference Center for Staphylococci, Hospices Civils de Lyon, Lyon, France
| | - Rafael Lucas Muniz Guedes
- Bioinformatics Laboratory - LABINFO, National Laboratory of Scientific Computation - LNCC/MCTI, Petrópolis, Brazil
| | | | - Jocelyne Caillon
- University of Nantes, Medical School, UPRES EA, 3826, Nantes, France
| | - Sebastien Lustig
- Orthopedic Surgery Department, Northern Hospital Group, Hospices Civils de Lyon, Lyon, France
| | - Tristan Ferry
- Centre International de Recherche en Infectiologie, INSERM U1111, Pathogenesis of staphylococcal infections, University of Lyon 1, Lyon, France.,Infectious Diseases Department, Northern Hospital Group, Hospices Civils de Lyon, Lyon, France
| | - Cédric Jacqueline
- University of Nantes, Medical School, UPRES EA, 3826, Nantes, France
| | - Guilherme Loss de Morais
- Bioinformatics Laboratory - LABINFO, National Laboratory of Scientific Computation - LNCC/MCTI, Petrópolis, Brazil
| | - Frédéric Laurent
- Centre International de Recherche en Infectiologie, INSERM U1111, Pathogenesis of staphylococcal infections, University of Lyon 1, Lyon, France.,Department of Clinical Microbiology, Northern Hospital Group, Hospices Civils de Lyon, Lyon, France.,National Reference Center for Staphylococci, Hospices Civils de Lyon, Lyon, France
| |
Collapse
|
21
|
den Reijer PM, Haisma EM, Lemmens-den Toom NA, Willemse J, Koning RA, Demmers JAA, Dekkers DHW, Rijkers E, El Ghalbzouri A, Nibbering PH, van Wamel W. Detection of Alpha-Toxin and Other Virulence Factors in Biofilms of Staphylococcus aureus on Polystyrene and a Human Epidermal Model. PLoS One 2016; 11:e0145722. [PMID: 26741798 PMCID: PMC4704740 DOI: 10.1371/journal.pone.0145722] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 12/07/2015] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND & AIM The ability of Staphylococcus aureus to successfully colonize (a)biotic surfaces may be explained by biofilm formation and the actions of virulence factors. The aim of the present study was to establish the presence of 52 proteins, including virulence factors such as alpha-toxin, during biofilm formation of five different (methicillin resistant) S. aureus strains on Leiden human epidermal models (LEMs) and polystyrene surfaces (PS) using a competitive Luminex-based assay. RESULTS All five S. aureus strains formed biofilms on PS, whereas only three out of five strains formed biofilms on LEMs. Out of the 52 tested proteins, six functionally diverse proteins (ClfB, glucosaminidase, IsdA, IsaA, SACOL0688 and nuclease) were detected in biofilms of all strains on both PS and LEMs. At the same time, four toxins (alpha-toxin, gamma-hemolysin B and leukocidins D and E), two immune modulators (formyl peptide receptor-like inhibitory protein and Staphylococcal superantigen-like protein 1), and two other proteins (lipase and LytM) were detectable in biofilms by all five S. aureus strains on LEMs, but not on PS. In contrast, fibronectin-binding protein B (FnbpB) was detectable in biofilms by all S. aureus biofilms on PS, but not on LEMs. These data were largely confirmed by the results from proteomic and transcriptomic analyses and in case of alpha-toxin additionally by GFP-reporter technology. CONCLUSION Functionally diverse virulence factors of (methicillin-resistant) S. aureus are present during biofilm formation on LEMs and PS. These results could aid in identifying novel targets for future treatment strategies against biofilm-associated infections.
Collapse
Affiliation(s)
- P. M. den Reijer
- Department of Medical Microbiology and Infectious Diseases, Erasmus University Medical Center, Rotterdam, The Netherlands
- * E-mail:
| | - E. M. Haisma
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
- Department of Dermatology, Leiden University Medical Center, Leiden, The Netherlands
| | - N. A. Lemmens-den Toom
- Department of Medical Microbiology and Infectious Diseases, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - J. Willemse
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - R. A. Koning
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - J. A. A. Demmers
- Proteomics Centre, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - D. H. W. Dekkers
- Proteomics Centre, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - E. Rijkers
- Proteomics Centre, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - A. El Ghalbzouri
- Department of Dermatology, Leiden University Medical Center, Leiden, The Netherlands
| | - P. H. Nibbering
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - W. van Wamel
- Department of Medical Microbiology and Infectious Diseases, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
22
|
Nocadello S, Minasov G, Shuvalova L, Dubrovska I, Sabini E, Bagnoli F, Grandi G, Anderson WF. Crystal structures of the components of the Staphylococcus aureus leukotoxin ED. Acta Crystallogr D Struct Biol 2016; 72:113-20. [PMID: 26894539 PMCID: PMC4756620 DOI: 10.1107/s2059798315023207] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 12/02/2015] [Indexed: 12/31/2022] Open
Abstract
Staphylococcal leukotoxins are a family of β-barrel, bicomponent, pore-forming toxins with membrane-damaging functions. These bacterial exotoxins share sequence and structural homology and target several host-cell types. Leukotoxin ED (LukED) is one of these bicomponent pore-forming toxins that Staphylococcus aureus produces in order to suppress the ability of the host to contain the infection. The recent delineation of the important role that LukED plays in S. aureus pathogenesis and the identification of its protein receptors, combined with its presence in S. aureus methicillin-resistant epidemic strains, establish this leukocidin as a possible target for the development of novel therapeutics. Here, the crystal structures of the water-soluble LukE and LukD components of LukED have been determined. The two structures illustrate the tertiary-structural variability with respect to the other leukotoxins while retaining the conservation of the residues involved in the interaction of the protomers in the bipartite leukotoxin in the pore complex.
Collapse
Affiliation(s)
- S. Nocadello
- Center for Structural Genomics of Infectious Diseases, Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - G. Minasov
- Center for Structural Genomics of Infectious Diseases, Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - L. Shuvalova
- Center for Structural Genomics of Infectious Diseases, Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - I. Dubrovska
- Center for Structural Genomics of Infectious Diseases, Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - E. Sabini
- Center for Structural Genomics of Infectious Diseases, Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - F. Bagnoli
- Novartis Vaccines and Diagnostics, Research Centre, Siena, Italy
| | - G. Grandi
- Novartis Vaccines and Diagnostics, Research Centre, Siena, Italy
| | - W. F. Anderson
- Center for Structural Genomics of Infectious Diseases, Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
23
|
Melehani JH, Duncan JA. Inflammasome Activation Can Mediate Tissue-Specific Pathogenesis or Protection in Staphylococcus aureus Infection. Curr Top Microbiol Immunol 2016; 397:257-82. [PMID: 27460814 DOI: 10.1007/978-3-319-41171-2_13] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Staphylococcus aureus is a Gram-positive coccus that interacts with human hosts on a spectrum from quiet commensal to deadly pathogen. S. aureus is capable of infecting nearly every tissue in the body resulting in cellulitis, pneumonia, osteomyelitis, endocarditis, brain abscesses, bacteremia, and more. S. aureus has a wide range of factors that promote infection, and each site of infection triggers a different response in the human host. In particular, the different patterns of inflammasome activation mediate tissue-specific pathogenesis or protection in S. aureus infection. Although still a nascent field, understanding the unique host-pathogen interactions in each infection and the role of inflammasomes in mediating pathogenesis may lead to novel strategies for treating S. aureus infections. Reviews addressing S. aureus virulence and pathogenesis (Thammavongsa et al. 2015), as well as epidemiology and pathophysiology (Tong et al. 2015), have recently been published. This review will focus on S. aureus factors that activate inflammasomes and their impact on innate immune signaling and bacterial survival.
Collapse
Affiliation(s)
- Jason H Melehani
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Joseph A Duncan
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA. .,Department of Medicine, Division of Infectious Diseases, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA. .,Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
24
|
Protection of mice against Staphylococcus aureus infection by a recombinant protein ClfA-IsdB-Hlg as a vaccine candidate. Med Microbiol Immunol 2015; 205:47-55. [PMID: 26155981 DOI: 10.1007/s00430-015-0425-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Accepted: 06/24/2015] [Indexed: 10/23/2022]
Abstract
Staphylococcus aureus is one of the most important causes of nosocomial infections. An effective vaccine to prevent S. aureus infections is urgently required due to the dramatic increase in the number of antibiotic-resistant strains. In this report, we evaluated a newly recombinant protein composed of selected antigenic regions of clumping factor A (ClfA), iron surface determinant B (IsdB) and gamma hemolysin B (HlgB) of S. aureus and sequence coding for hydrophobic linkers between three domains. The recombinant gene was constructed in pET-28a (+) and expressed in Escherichia coli BL21. In addition, sequence coding for a His(6)-tag was added followed by a hybrid procedure of nickel chelate protein purification. Immunization of BALB/c mice with the recombinant protein ClfA-IsdB-Hlg evoked antigen-specific antibodies that could opsonize S. aureus cells, enhancing in vitro phagocytosis by macrophages. Vaccination with the recombinant protein also reduced the bacterial load recovered from mice spleen samples and increased survival following the intraperitoneal challenge with pathogenic S. aureus compared to the control mice. Our results showed that the recombinant protein ClfA-IsdB-Hlg is a promising vaccine candidate for the prevention of S. aureus bacteremia infections.
Collapse
|
25
|
The bicomponent pore-forming leucocidins of Staphylococcus aureus. Microbiol Mol Biol Rev 2015; 78:199-230. [PMID: 24847020 DOI: 10.1128/mmbr.00055-13] [Citation(s) in RCA: 192] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The ability to produce water-soluble proteins with the capacity to oligomerize and form pores within cellular lipid bilayers is a trait conserved among nearly all forms of life, including humans, single-celled eukaryotes, and numerous bacterial species. In bacteria, some of the most notable pore-forming molecules are protein toxins that interact with mammalian cell membranes to promote lysis, deliver effectors, and modulate cellular homeostasis. Of the bacterial species capable of producing pore-forming toxic molecules, the Gram-positive pathogen Staphylococcus aureus is one of the most notorious. S. aureus can produce seven different pore-forming protein toxins, all of which are believed to play a unique role in promoting the ability of the organism to cause disease in humans and other mammals. The most diverse of these pore-forming toxins, in terms of both functional activity and global representation within S. aureus clinical isolates, are the bicomponent leucocidins. From the first description of their activity on host immune cells over 100 years ago to the detailed investigations of their biochemical function today, the leucocidins remain at the forefront of S. aureus pathogenesis research initiatives. Study of their mode of action is of immediate interest in the realm of therapeutic agent design as well as for studies of bacterial pathogenesis. This review provides an updated perspective on our understanding of the S. aureus leucocidins and their function, specificity, and potential as therapeutic targets.
Collapse
|
26
|
Richter E, Harms M, Ventz K, Gierok P, Chilukoti RK, Hildebrandt JP, Mostertz J, Hochgräfe F. A multi-omics approach identifies key hubs associated with cell type-specific responses of airway epithelial cells to staphylococcal alpha-toxin. PLoS One 2015; 10:e0122089. [PMID: 25816343 PMCID: PMC4376684 DOI: 10.1371/journal.pone.0122089] [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: 11/18/2014] [Accepted: 02/17/2015] [Indexed: 12/18/2022] Open
Abstract
Responsiveness of cells to alpha-toxin (Hla) from Staphylococcus aureus appears to occur in a cell-type dependent manner. Here, we compare two human bronchial epithelial cell lines, i.e. Hla-susceptible 16HBE14o- and Hla-resistant S9 cells, by a quantitative multi-omics strategy for a better understanding of Hla-induced cellular programs. Phosphoproteomics revealed a substantial impact on phosphorylation-dependent signaling in both cell models and highlights alterations in signaling pathways associated with cell-cell and cell-matrix contacts as well as the actin cytoskeleton as key features of early rHla-induced effects. Along comparable changes in down-stream activity of major protein kinases significant differences between both models were found upon rHla-treatment including activation of the epidermal growth factor receptor EGFR and mitogen-activated protein kinases MAPK1/3 signaling in S9 and repression in 16HBE14o- cells. System-wide transcript and protein expression profiling indicate induction of an immediate early response in either model. In addition, EGFR and MAPK1/3-mediated changes in gene expression suggest cellular recovery and survival in S9 cells but cell death in 16HBE14o- cells. Strikingly, inhibition of the EGFR sensitized S9 cells to Hla indicating that the cellular capacity of activation of the EGFR is a major protective determinant against Hla-mediated cytotoxic effects.
Collapse
Affiliation(s)
- Erik Richter
- Competence Center Functional Genomics, Junior Research Group Pathoproteomics, University of Greifswald, 17489, Greifswald, Germany
| | - Manuela Harms
- Competence Center Functional Genomics, Junior Research Group Pathoproteomics, University of Greifswald, 17489, Greifswald, Germany
| | - Katharina Ventz
- Competence Center Functional Genomics, Junior Research Group Pathoproteomics, University of Greifswald, 17489, Greifswald, Germany
| | - Philipp Gierok
- Department of Biochemistry, University of Greifswald, 17487, Greifswald, Germany
| | - Ravi Kumar Chilukoti
- Interfaculty Institute for Genetics and Functional Genomics, Department of Functional Genomics, University of Greifswald, 17489, Greifswald, Germany
| | - Jan-Peter Hildebrandt
- Animal Physiology and Biochemistry, Zoological Institute, University of Greifswald, 17487, Greifswald, Germany
| | - Jörg Mostertz
- Competence Center Functional Genomics, Junior Research Group Pathoproteomics, University of Greifswald, 17489, Greifswald, Germany
| | - Falko Hochgräfe
- Competence Center Functional Genomics, Junior Research Group Pathoproteomics, University of Greifswald, 17489, Greifswald, Germany
- * E-mail:
| |
Collapse
|
27
|
Arroliga AC, Velazco JF, Midturi JK, Ghamande SA. Back to the future: α-hemolysin activity on blood agar to predict ventilator-associated pneumonia caused by Staphylococcus aureus. Am J Respir Crit Care Med 2015; 190:1086-8. [PMID: 25398106 DOI: 10.1164/rccm.201410-1886ed] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
28
|
Delfani S, Imani Fooladi AA, Mobarez AM, Emaneini M, Amani J, Sedighian H. In silico analysis for identifying potential vaccine candidates against Staphylococcus aureus. Clin Exp Vaccine Res 2015; 4:99-106. [PMID: 25649548 PMCID: PMC4313115 DOI: 10.7774/cevr.2015.4.1.99] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 10/24/2014] [Accepted: 10/27/2014] [Indexed: 01/08/2023] Open
Abstract
Purpose Staphylococcus aureus is one of the most important causes of nosocomial and community-acquired infections. The increasing incidence of multiple antibiotic-resistant S. aureus strains and the emergence of vancomycin resistant S. aureus strains have placed renewed interest on alternative means of prevention and control of infection. S. aureus produces a variety of virulence factors, so a multi-subunit vaccine will be more successful for preventing S. aureus infections than a mono-subunit vaccine. Materials and Methods We selected three important virulence factors of S. aureus, clumping factor A (ClfA), iron-regulated surface determinant (IsdB), and gamma hemolysin (Hlg) that are potential candidates for vaccine development. We designed synthetic genes encoding the clfA, isdB, and hlg and used bioinformatics tools to predict structure of the synthetic construct and its stabilities. VaxiJen analysis of the protein showed a high antigenicity. Linear and conformational B-cell epitopes were identified. Results The proteins encoded by these genes were useful as vaccine candidates against S. aureus infections. Conclusion In silico tools are highly suited to study, design, and evaluate vaccine strategies.
Collapse
Affiliation(s)
- Somayeh Delfani
- Department of Bacteriology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Abbas Ali Imani Fooladi
- Applied Microbiology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Ashraf Mohabati Mobarez
- Department of Bacteriology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mohammad Emaneini
- Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Jafar Amani
- Applied Microbiology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Hamid Sedighian
- Applied Microbiology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
29
|
Perlińska A, Grynberg M. Bacillus anthracis pXO1 plasmid encodes a putative membrane-bound bacteriocin. PeerJ 2014; 2:e679. [PMID: 25426338 PMCID: PMC4243335 DOI: 10.7717/peerj.679] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Accepted: 11/06/2014] [Indexed: 11/20/2022] Open
Abstract
Evolutionary advantages over cousin cells in bacterial pathogens may decide about the success of a specific cell in its environment. Bacteria use a plethora of methods to defend against other cells and many devices to attack their opponents when competing for resources. Bacteriocins are antibacterial proteins that are used to eliminate competition. We report the discovery of a putative membrane-bound bacteriocin encoded by the Bacillus anthracis pathogenic pXO1 plasmid. We analyze the genomic structure of the bacteriocin operon. The proposed mechanisms of action predestine this operon as a potent competitive advantage over cohabitants of the same niche.
Collapse
Affiliation(s)
- Agata Perlińska
- Centre of New Technologies, University of Warsaw , Banacha, Warsaw , Poland
| | - Marcin Grynberg
- Institute of Biochemistry and Biophysics PAS , Pawińskiego, Warsaw , Poland
| |
Collapse
|
30
|
Badarau A, Rouha H, Malafa S, Logan DT, Håkansson M, Stulik L, Dolezilkova I, Teubenbacher A, Gross K, Maierhofer B, Weber S, Jägerhofer M, Hoffman D, Nagy E. Structure-function analysis of heterodimer formation, oligomerization, and receptor binding of the Staphylococcus aureus bi-component toxin LukGH. J Biol Chem 2014; 290:142-56. [PMID: 25371205 DOI: 10.1074/jbc.m114.598110] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
The bi-component leukocidins of Staphylococcus aureus are important virulence factors that lyse human phagocytic cells and contribute to immune evasion. The γ-hemolysins (HlgAB and HlgCB) and Panton-Valentine leukocidin (PVL or LukSF) were shown to assemble from soluble subunits into membrane-bound oligomers on the surface of target cells, creating barrel-like pore structures that lead to cell lysis. LukGH is the most distantly related member of this toxin family, sharing only 30-40% amino acid sequence identity with the others. We observed that, unlike other leukocidin subunits, recombinant LukH and LukG had low solubility and were unable to bind to target cells, unless both components were present. Using biolayer interferometry and intrinsic tryptophan fluorescence we detected binding of LukH to LukG in solution with an affinity in the low nanomolar range and dynamic light scattering measurements confirmed formation of a heterodimer. We elucidated the structure of LukGH by x-ray crystallography at 2.8-Å resolution. This revealed an octameric structure that strongly resembles that reported for HlgAB, but with important structural differences. Structure guided mutagenesis studies demonstrated that three salt bridges, not found in other bi-component leukocidins, are essential for dimer formation in solution and receptor binding. We detected weak binding of LukH, but not LukG, to the cellular receptor CD11b by biolayer interferometry, suggesting that in common with other members of this toxin family, the S-component has the primary contact role with the receptor. These new insights provide the basis for novel strategies to counteract this powerful toxin and Staphylococcus aureus pathogenesis.
Collapse
Affiliation(s)
- Adriana Badarau
- From Arsanis Biosciences, Vienna Biocenter Campus, Helmut-Qualtinger-Gasse 2, 1030 Vienna, Austria and
| | - Harald Rouha
- From Arsanis Biosciences, Vienna Biocenter Campus, Helmut-Qualtinger-Gasse 2, 1030 Vienna, Austria and
| | - Stefan Malafa
- From Arsanis Biosciences, Vienna Biocenter Campus, Helmut-Qualtinger-Gasse 2, 1030 Vienna, Austria and
| | - Derek T Logan
- SARomics Biostructures AB, Medicon Village, S-223 81 Lund, Sweden
| | - Maria Håkansson
- SARomics Biostructures AB, Medicon Village, S-223 81 Lund, Sweden
| | - Lukas Stulik
- From Arsanis Biosciences, Vienna Biocenter Campus, Helmut-Qualtinger-Gasse 2, 1030 Vienna, Austria and
| | - Ivana Dolezilkova
- From Arsanis Biosciences, Vienna Biocenter Campus, Helmut-Qualtinger-Gasse 2, 1030 Vienna, Austria and
| | - Astrid Teubenbacher
- From Arsanis Biosciences, Vienna Biocenter Campus, Helmut-Qualtinger-Gasse 2, 1030 Vienna, Austria and
| | - Karin Gross
- From Arsanis Biosciences, Vienna Biocenter Campus, Helmut-Qualtinger-Gasse 2, 1030 Vienna, Austria and
| | - Barbara Maierhofer
- From Arsanis Biosciences, Vienna Biocenter Campus, Helmut-Qualtinger-Gasse 2, 1030 Vienna, Austria and
| | - Susanne Weber
- From Arsanis Biosciences, Vienna Biocenter Campus, Helmut-Qualtinger-Gasse 2, 1030 Vienna, Austria and
| | - Michaela Jägerhofer
- From Arsanis Biosciences, Vienna Biocenter Campus, Helmut-Qualtinger-Gasse 2, 1030 Vienna, Austria and
| | - David Hoffman
- From Arsanis Biosciences, Vienna Biocenter Campus, Helmut-Qualtinger-Gasse 2, 1030 Vienna, Austria and
| | - Eszter Nagy
- From Arsanis Biosciences, Vienna Biocenter Campus, Helmut-Qualtinger-Gasse 2, 1030 Vienna, Austria and
| |
Collapse
|
31
|
Oganesyan V, Peng L, Damschroder MM, Cheng L, Sadowska A, Tkaczyk C, Sellman BR, Wu H, Dall'Acqua WF. Mechanisms of neutralization of a human anti-α-toxin antibody. J Biol Chem 2014; 289:29874-80. [PMID: 25210036 PMCID: PMC4207998 DOI: 10.1074/jbc.m114.601328] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
MEDI4893 is a neutralizing human monoclonal antibody that targets α-toxin (AT) and is currently undergoing evaluation in the field of Staphylococcus aureus-mediated diseases. We have solved the crystal structure of MEDI4893 Fab bound to monomeric AT at a resolution of 2.56 Å and further characterized its epitope using various engineered AT variants. We have found that MEDI4893 recognizes a novel epitope in the so-called “rim” domain of AT and exerts its neutralizing effect through a dual mechanism. In particular, MEDI4893 not only sterically blocks binding of AT to its cell receptor but also prevents it from adopting a lytic heptameric trans-membrane conformation.
Collapse
Affiliation(s)
- Vaheh Oganesyan
- From the Departments of Antibody Discovery and Protein Engineering and
| | - Li Peng
- From the Departments of Antibody Discovery and Protein Engineering and
| | | | - Li Cheng
- From the Departments of Antibody Discovery and Protein Engineering and
| | | | | | - Bret R Sellman
- Infectious Diseases, MedImmune, Gaithersburg, Maryland 20878
| | - Herren Wu
- From the Departments of Antibody Discovery and Protein Engineering and
| | | |
Collapse
|
32
|
Genome of Staphylococcus xylosus and Comparison with S. aureus and S. epidermidis. J Genet Genomics 2014; 41:413-6. [DOI: 10.1016/j.jgg.2014.03.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Revised: 03/21/2014] [Accepted: 03/25/2014] [Indexed: 11/18/2022]
|
33
|
Fernandes da Costa SP, Savva CG, Bokori-Brown M, Naylor CE, Moss DS, Basak AK, Titball RW. Identification of a key residue for oligomerisation and pore-formation of Clostridium perfringens NetB. Toxins (Basel) 2014; 6:1049-61. [PMID: 24625763 PMCID: PMC3968376 DOI: 10.3390/toxins6031049] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 02/26/2014] [Accepted: 03/03/2014] [Indexed: 11/22/2022] Open
Abstract
Necrotic enteritis toxin B (NetB) is a β-pore-forming toxin produced by Clostridium perfringens and has been identified as a key virulence factor in the pathogenesis of avian necrotic enteritis, a disease causing significant economic damage to the poultry industry worldwide. In this study, site-directed mutagenesis was used to identify amino acids that play a role in NetB oligomerisation and pore-formation. NetB K41H showed significantly reduced toxicity towards LMH cells and human red blood cells relative to wild type toxin. NetB K41H was unable to oligomerise and form pores in liposomes. These findings suggest that NetB K41H could be developed as a genetic toxoid vaccine to protect against necrotic enteritis.
Collapse
Affiliation(s)
| | - Christos G Savva
- Department of Biological Sciences, School of Crystallography, Institute of Structural and Molecular Biology, Birkbeck College, Malet Street, London WC1E 7HX, UK.
| | - Monika Bokori-Brown
- College of Life and Environmental Sciences, University of Exeter, Stocker Road, Exeter EX4 4QD, UK.
| | - Claire E Naylor
- Department of Biological Sciences, School of Crystallography, Institute of Structural and Molecular Biology, Birkbeck College, Malet Street, London WC1E 7HX, UK.
| | - David S Moss
- Department of Biological Sciences, School of Crystallography, Institute of Structural and Molecular Biology, Birkbeck College, Malet Street, London WC1E 7HX, UK.
| | - Ajit K Basak
- Department of Biological Sciences, School of Crystallography, Institute of Structural and Molecular Biology, Birkbeck College, Malet Street, London WC1E 7HX, UK.
| | - Richard W Titball
- College of Life and Environmental Sciences, University of Exeter, Stocker Road, Exeter EX4 4QD, UK.
| |
Collapse
|
34
|
Induction of virulence gene expression in Staphylococcus aureus by pulmonary surfactant. Infect Immun 2014; 82:1500-10. [PMID: 24452679 DOI: 10.1128/iai.01635-13] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We performed a genomewide analysis using a next-generation sequencer to investigate the effect of pulmonary surfactant on gene expression in Staphylococcus aureus, a clinically important opportunistic pathogen. RNA sequence (RNA-seq) analysis of bacterial transcripts at late log phase revealed 142 genes that were upregulated >2-fold following the addition of pulmonary surfactant to the culture medium. Among these genes, we confirmed by quantitative reverse transcription-PCR analysis that mRNA amounts for genes encoding ESAT-6 secretion system C (EssC), an unknown hypothetical protein (NWMN_0246; also called pulmonary surfactant-inducible factor A [PsiA] in this study), and hemolysin gamma subunit B (HlgB) were increased 3- to 10-fold by the surfactant treatment. Among the major constituents of pulmonary surfactant, i.e., phospholipids and palmitate, only palmitate, which is the most abundant fatty acid in the pulmonary surfactant and a known antibacterial substance, stimulated the expression of these three genes. Moreover, these genes were also induced by supplementing the culture with detergents. The induction of gene expression by surfactant or palmitate was not observed in a disruption mutant of the sigB gene, which encodes an alternative sigma factor involved in bacterial stress responses. Furthermore, each disruption mutant of the essC, psiA, and hlgB genes showed attenuation of both survival in the lung and host-killing ability in a murine pneumonia model. These findings suggest that S. aureus resists membrane stress caused by free fatty acids present in the pulmonary surfactant through the regulation of virulence gene expression, which contributes to its pathogenesis within the lungs of the host animal.
Collapse
|
35
|
Abstract
Staphylococcus aureus secretes a number of host-injurious toxins, among the most prominent of which is the small β-barrel pore-forming toxin α-hemolysin. Initially named based on its properties as a red blood cell lytic toxin, early studies suggested a far greater complexity of α-hemolysin action as nucleated cells also exhibited distinct responses to intoxication. The hemolysin, most aptly referred to as α-toxin based on its broad range of cellular specificity, has long been recognized as an important cause of injury in the context of both skin necrosis and lethal infection. The recent identification of ADAM10 as a cellular receptor for α-toxin has provided keen insight on the biology of toxin action during disease pathogenesis, demonstrating the molecular mechanisms by which the toxin causes tissue barrier disruption at host interfaces lined by epithelial or endothelial cells. This review highlights both the historical studies that laid the groundwork for nearly a century of research on α-toxin and key findings on the structural and functional biology of the toxin, in addition to discussing emerging observations that have significantly expanded our understanding of this toxin in S. aureus disease. The identification of ADAM10 as a proteinaceous receptor for the toxin not only provides a greater appreciation of truths uncovered by many historic studies, but now affords the opportunity to more extensively probe and understand the role of α-toxin in modulation of the complex interaction of S. aureus with its human host.
Collapse
Affiliation(s)
- Bryan J. Berube
- Department of Microbiology, The University of Chicago, 920 E. 58th Street Chicago, IL 60637, USA; E-Mail:
| | - Juliane Bubeck Wardenburg
- Department of Microbiology, The University of Chicago, 920 E. 58th Street Chicago, IL 60637, USA; E-Mail:
- Department of Pediatrics, The University of Chicago, 5721 S. Maryland Ave. Chicago, IL 60637, USA
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-773-834-9763; Fax: +1-773-834-8150
| |
Collapse
|
36
|
Genetic nature and virulence of community-associated methicillin-resistant Staphylococcus aureus. Biomedicine (Taipei) 2013. [DOI: 10.1016/j.biomed.2012.12.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
37
|
Characterization of the humoral immune response during Staphylococcus aureus bacteremia and global gene expression by Staphylococcus aureus in human blood. PLoS One 2013; 8:e53391. [PMID: 23308212 PMCID: PMC3538780 DOI: 10.1371/journal.pone.0053391] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Accepted: 11/27/2012] [Indexed: 01/06/2023] Open
Abstract
Attempts to develop an efficient anti-staphylococcal vaccine in humans have so far been unsuccessful. Therefore, more knowledge of the antigens that are expressed by Staphylococcus aureus in human blood and induce an immune response in patients is required. In this study we further characterize the serial levels of IgG and IgA antibodies against 56 staphylococcal antigens in multiple serum samples of 21 patients with a S. aureus bacteremia, compare peak IgG levels between patients and 30 non-infected controls, and analyze the expression of 3626 genes by two genetically distinct isolates in human blood. The serum antibody levels were measured using a bead-based flow cytometry technique (xMAP®, Luminex corporation). Gene expression levels were analyzed using a microarray (BµG@s microarray). The initial levels and time taken to reach peak IgG and IgA antibody levels were heterogeneous in bacteremia patients. The antigen SA0688 was associated with the highest median initial-to-peak antibody fold-increase for IgG (5.05-fold) and the second highest increase for IgA (2.07-fold). Peak IgG levels against 27 antigens, including the antigen SA0688, were significantly elevated in bacteremia patients versus controls (P≤0.05). Expression of diverse genes, including SA0688, was ubiquitously high in both isolates at all time points during incubation in blood. However, only a limited number of genes were specifically up- or downregulated in both isolates when cultured in blood, compared to the start of incubation in blood or during incubation in BHI broth. In conclusion, most staphylococcal antigens tested in this study, including many known virulence factors, do not induce uniform increases in the antibody levels in bacteremia patients. In addition, the expression of these antigens by S. aureus is not significantly altered by incubation in human blood over time. One immunogenic and ubiquitously expressed antigen is the putative iron-regulated ABC transporter SA0688.
Collapse
|
38
|
Abstract
Bacterial pathogens have specific virulence factors (e.g., toxins) that contribute significantly to the virulence and infectivity of microorganisms within the human hosts. Virulence factors are molecules expressed by pathogens that enable colonization, immunoevasion, and immunosuppression, obtaining nutrients from the host or gaining entry into host cells. They can cause pathogenesis by inhibiting or stimulating certain host functions. For example, in systemic Staphylococcus aureus infections, virulence factors such as toxic shock syndrome toxin 1 (TSST-1), staphylococcal enterotoxin A (SEA), and staphylococcal enterotoxin B (SEB) cause sepsis or toxic shock by uncontrolled stimulation of T lymphocytes and by triggering a cytokine storm. In vitro, these superantigens stimulate the proliferation of human peripheral blood mononuclear cells (PBMC) and the release of many cytokines. NVC-422 (N,N-dichloro-2,2-dimethyltaurine) is a broad-spectrum, fast-acting topical anti-infective agent against microbial pathogens, including antibiotic-resistant microbes. Using mass spectrometry, we demonstrate here that NVC-422 oxidizes methionine residues of TSST-1, SEA, SEB, and exfoliative toxin A (ETA). Exposure of virulence factors to 0.1% NVC-422 for 1 h prevented TSST-1-, SEA-, SEB-, and ETA-induced cell proliferation and cytokine release. Moreover, NVC-422 also delayed and reduced the protein A- and clumping factor-associated agglutination of S. aureus cultures. These results show that, in addition to its well-described direct microbicidal activity, NVC-422 can inactivate S. aureus virulence factors through rapid oxidation of methionines.
Collapse
|
39
|
Robertson JWF, Kasianowicz JJ, Banerjee S. Analytical Approaches for Studying Transporters, Channels and Porins. Chem Rev 2012; 112:6227-49. [DOI: 10.1021/cr300317z] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Joseph W. F. Robertson
- Physical Measurement Laboratory,
National Institute of Standards and Technology, Gaithersburg, Maryland
20899, United States
| | - John J. Kasianowicz
- Physical Measurement Laboratory,
National Institute of Standards and Technology, Gaithersburg, Maryland
20899, United States
| | - Soojay Banerjee
- National
Institute of Neurological
Disorders and Stroke, Bethesda, Maryland 20824, United States
| |
Collapse
|
40
|
Alessandrini A, Viero G, Dalla Serra M, Prévost G, Facci P. γ-Hemolysin oligomeric structure and effect of its formation on supported lipid bilayers: an AFM investigation. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2012; 1828:405-11. [PMID: 23036932 DOI: 10.1016/j.bbamem.2012.09.027] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2012] [Revised: 09/05/2012] [Accepted: 09/25/2012] [Indexed: 11/28/2022]
Abstract
γ-Hemolysins are bicomponent β-barrel pore forming toxins produced by Staphylococcus aureus as water-soluble monomers, which assemble into oligomeric pores on the surface of lipid bilayers. Here, after investigating the oligomeric structure of γ-hemolysins on supported lipid bilayers (SLBs) by atomic force microscopy (AFM), we studied the effect produced by this toxin on the structure of SLBs. We found that oligomeric structures with different number of monomers can assemble on the lipid bilayer being the octameric form the stablest one. Moreover, in this membrane model we found that γ-hemolysins can form clusters of oligomers inducing a curvature in the lipid bilayer, which could probably enhance the aggressiveness of these toxins at high concentrations.
Collapse
Affiliation(s)
- Andrea Alessandrini
- Centro S3, CNR-Istituto di Nanoscienze, Via Campi 213/A, 41125 Modena, Italy.
| | | | | | | | | |
Collapse
|
41
|
Nishiyama A, Isobe H, Iwao Y, Takano T, Hung WC, Taneike I, Nakagawa S, Dohmae S, Iwakura N, Yamamoto T. Accumulation of staphylococcal Panton-Valentine leukocidin in the detergent-resistant membrane microdomains on the target cells is essential for its cytotoxicity. ACTA ACUST UNITED AC 2012; 66:343-52. [PMID: 22924956 DOI: 10.1111/j.1574-695x.2012.01027.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2011] [Revised: 07/24/2012] [Accepted: 07/27/2012] [Indexed: 12/23/2022]
Abstract
The mechanisms for the cytotoxicity of staphylococcal Panton-Valentine leukocidin (PVL), a pore-forming toxin consisting of LukS-PV and LukF-PV, in human immune cells are still unclear. Because LukS-PV binds to ganglioside GM1, a constituent of detergent-resistant membrane microdomains (DRMs) of the plasma membrane, the role of DRMs in PVL cytotoxicity was examined in human polymorphonuclear neutrophils (PMNs), monocytes, HL-60 cells, and THP-1 cells. PVL binding capacities in HL-60 and THP-1 cells were higher than those in PMNs and monocytes; however, the PVL concentration to obtain more than 80% cell lysis in HL-60 cells was 10 times higher than that in PMNs and PVL even at such concentration induced < 10% cell lysis in THP-1 cells. After incubation of PMNs with LukS-PV, more than 90% of LukS-PV bound to the detergent-soluble membranes. Subsequent incubation with LukF-PV at 4 °C induced the accumulation of more than 70% of PVL components and 170- to 220-kDa complex formation in DRMs in an actin-independent manner. However, only 30% of PVL was found, and complex formation was under detectable level in DRMs in HL-60 cells. PVL did not accumulate in DRMs in THP-1 cells. Our observations strongly indicate that PVL accumulation in DRMs is essential for PVL cytotoxicity.
Collapse
Affiliation(s)
- Akihito Nishiyama
- Division of Bacteriology, Department of Infectious Disease Control and International Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Wang H, Wu Y, Ojcius DM, Yang XF, Zhang C, Ding S, Lin X, Yan J. Leptospiral hemolysins induce proinflammatory cytokines through Toll-like receptor 2-and 4-mediated JNK and NF-κB signaling pathways. PLoS One 2012; 7:e42266. [PMID: 22870312 PMCID: PMC3411626 DOI: 10.1371/journal.pone.0042266] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Accepted: 07/02/2012] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Infection with pathogenic Leptospira species causes serious systemic inflammation in patients. Although a few leptospiral proinflammatory molecules have been identified, Leptospira likely encodes other unidentified strong inflammation stimulators. The pathogenic L. interrogans genome encodes numerous putative hemolysin genes. Since hemolysins from other bacteria can cause inflammatory reactions, we hypothesized that leptospiral hemolysins may function as proinflammatory stimulators that contribute to the strong inflammation associated with Leptospira infection. METHODOLOGY/PRINCIPAL FINDINGS We first used cytokine protein microarrays for systematic analysis of serum cytokine profiles in leptospirosis patients and leptospire-infected mice. We found that IL-1β, IL-6 and TNF-α were the main proinflammatory cytokines in the sera of both the patients and the mice. We then analyzed eight putative hemolysins in L. interrogans strain Lai. The results showed that five of them, Sph1, Sph2, Sph3, HlpA and TlyA were secreted and had hemolytic activity. More importantly, these five hemolysins induced the strong production of IL-1β, IL-6 and TNF-α in human and mouse macrophages (although a bit lower in the latter). Furthermore, blockade of TLR2 or TLR4 with either antibodies or inhibitors of the NF-κB or JNK signaling pathways significantly reduced the production of hemolysin-induced IL-1β, IL-6 and TNF-α. Macrophages isolated from TLR2-, TLR4-or double TLR2-and 4-deficient mice also confirmed that the leptospiral hemolysins that induce proinflammatory cytokines are both TLR2-and TLR4-dependent. CONCLUSIONS/SIGNIFICANCE Our findings demonstrate that L. interrogans secretes many hemolysins that function as powerful inducers of proinflammatory cytokines through both TLR2-and TLR4-dependent JNK and NF-κB pathways.
Collapse
Affiliation(s)
- Huan Wang
- Department of Medical Microbiology and Parasitology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Division of Basic Medical Microbiology, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yifei Wu
- Department of Medical Microbiology and Parasitology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - David M. Ojcius
- Health Sciences Research Institute and Molecular Cell Biology, University of California, Merced, California, United States of America
| | - X. Frank Yang
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Chenglin Zhang
- Department of Medical Microbiology and Parasitology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Shibiao Ding
- Department of Medical Microbiology and Parasitology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xu’ai Lin
- Department of Medical Microbiology and Parasitology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- * E-mail: (JY); (XL)
| | - Jie Yan
- Department of Medical Microbiology and Parasitology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Division of Basic Medical Microbiology, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- * E-mail: (JY); (XL)
| |
Collapse
|
43
|
Uppalapati SR, Kingston JJ, Murali HS, Batra HV. Generation and characterization of an inter-generic bivalent alpha domain fusion protein αCS from Clostridium perfringens and Staphylococcus aureus for concurrent diagnosis and therapeutic applications. J Appl Microbiol 2012; 113:448-58. [PMID: 22563989 DOI: 10.1111/j.1365-2672.2012.05333.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
AIM To evaluate an inter-generic recombinant alpha domain fusion protein for simultaneous detection and neutralization of Clostridium perfringens and Staphylococcus aureus alpha toxins. METHODS AND RESULTS Truncated portions of clostridial and staphylococcal alpha haemolysin genes were PCR amplified and linked to each other through a hydrophilic flexible Glycine linker sequence using overlap-extension PCR to form a chimeric gene αCS. The recombinant αCS fusion protein was expressed and characterized for its toxicity, cell binding capacity and haemolysis inhibition properties. The fusion protein was nontoxic and effectively retarded staphylococcal alpha haemolysis, probably by competitively interacting with putative staphylococcal alpha haemolysin receptors on erythrocytes. Murine hyperimmune polysera raised against r-αCS specifically detected 42-kDa and 33-kDa proteins when culture supernatants of Cl. perfringens (clostridial alpha toxin) and Staph. aureus (staphylococcal alpha toxin), respectively, were analysed in Western blot. The polyclonal antisera effectively diminished the haemolytic action of both the wild-type toxins in vitro. CONCLUSIONS The r-αCS fusion protein was nontoxic competitive inhibitor of staphylococcal alpha haemolysin. The protein elicited specific immune response against Cl. perfringens and Staph. aureus alpha toxins. The antisera also neutralized the toxicities of both the native wild-type toxins in vitro. SIGNIFICANCE OF THE STUDY The bivalent recombinant αCS protein could be a novel intervention in the field of diagnostics and therapeutics against Cl. perfringens and Staph. aureus infections, particularly, in case of co-infections like gangrenous ischaemia, gangrenous mastitis, etc.
Collapse
Affiliation(s)
- S R Uppalapati
- Microbiology Division, Defence Food Research Laboratory, Siddarthanagar, Mysore, Karnataka, India
| | | | | | | |
Collapse
|
44
|
Pilpa RM, Bayrhuber M, Marlett JM, Riek R, Young JAT. A receptor-based switch that regulates anthrax toxin pore formation. PLoS Pathog 2011; 7:e1002354. [PMID: 22174672 PMCID: PMC3234216 DOI: 10.1371/journal.ppat.1002354] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2011] [Accepted: 09/19/2011] [Indexed: 11/19/2022] Open
Abstract
Cellular receptors can act as molecular switches, regulating the sensitivity of microbial proteins to conformational changes that promote cellular entry. The activities of these receptor-based switches are only partially understood. In this paper, we sought to understand the mechanism that underlies the activity of the ANTXR2 anthrax toxin receptor-based switch that binds to domains 2 and 4 of the protective antigen (PA) toxin subunit. Receptor-binding restricts structural changes within the heptameric PA prepore that are required for pore conversion to an acidic endosomal compartment. The transfer cross-saturation (TCS) NMR approach was used to monitor changes in the heptameric PA-receptor contacts at different steps during prepore-to-pore conversion. These studies demonstrated that receptor contact with PA domain 2 is weakened prior to pore conversion, defining a novel intermediate in this pathway. Importantly, ANTXR2 remained bound to PA domain 4 following pore conversion, suggesting that the bound receptor might influence the structure and/or function of the newly formed pore. These studies provide new insights into the function of a receptor-based molecular switch that controls anthrax toxin entry into cells. The bacterium that causes anthrax produces a toxin called anthrax toxin that is largely responsible for causing disease symptoms. The first step in anthrax intoxication involves binding of the toxin to a specific protein, called a receptor, on the cell surface. Receptor-binding acts like a switch to prevent the toxin from forming a pore in a cell membrane until the toxin-receptor complex is taken up into cells and delivered to a specific location (called an endosome) where it is exposed to an “acid bath”. This acidic environment promotes structural changes in the toxin leading to pore formation in the endosomal membrane. In this report, we have studied how the receptor regulates pore formation by following the associated changes in toxin-receptor contacts. These studies have defined a new toxin-receptor intermediate in the pathway leading to pore conversion and demonstrate that the receptor remains bound after pore conversion. Our results provide important new insights into how the receptor regulates anthrax toxin pore formation, information that could be useful for designing new therapeutic strategies to treat this disease.
Collapse
Affiliation(s)
- Rosemarie M. Pilpa
- Nomis Center for Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, La Jolla, California, United States of America
| | - Monika Bayrhuber
- Structural Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, California, United States of America
- Laboratory of Physical Chemistry, ETH Zürich, Zürich, Switzerland
| | - John M. Marlett
- Nomis Center for Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, La Jolla, California, United States of America
| | - Roland Riek
- Structural Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, California, United States of America
- Laboratory of Physical Chemistry, ETH Zürich, Zürich, Switzerland
- * E-mail: (JATY); (RR)
| | - John A. T. Young
- Nomis Center for Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, La Jolla, California, United States of America
- * E-mail: (JATY); (RR)
| |
Collapse
|
45
|
Li HE, Qiu JZ, Yang ZQ, Dong J, Wang JF, Luo MJ, Pan J, Dai XH, Zhang Y, Song BL, Deng XM. Glycyrrhetinic acid protects mice from Staphylococcus aureus pneumonia. Fitoterapia 2011; 83:241-8. [PMID: 22085765 DOI: 10.1016/j.fitote.2011.10.018] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2011] [Revised: 10/24/2011] [Accepted: 10/31/2011] [Indexed: 11/18/2022]
Abstract
In the present study, the antimicrobial activity of glycyrrhetinic acid (GA) against Staphylococcus aureus, and its influence on the production of S. aureus alpha-haemolysin (Hla) were investigated, along with the in vivo activity of GA against S. aureus-induced pneumonia. GA could not inhibit the growth of S. aureus, but the secretion of Hla by S. aureus was significantly inhibited by low concentrations of GA in a dose-dependent manner. Furthermore, in vivo data show that GA provides protection against staphylococcal pneumonia in a murine model system.
Collapse
Affiliation(s)
- Hong-en Li
- Key Laboratory of Zoonosis, Ministry of Education, Institute of Zoonosis, College of Animal Science and Veterinary Medicine, Jilin University, Changchun 130062, PR China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Prevention and treatment of Staphylococcus aureus pneumonia with a beta-cyclodextrin derivative. Antimicrob Agents Chemother 2009; 54:298-304. [PMID: 19805564 DOI: 10.1128/aac.00973-09] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Staphylococcus aureus pneumonia is a common, potentially life-threatening infection caused by this human pathogen. The only therapies available to treat S. aureus pneumonia are antibiotics, a modality that is jeopardized by the organism's remarkable ability to acquire antimicrobial resistance. S. aureus alpha-hemolysin is a pore-forming cytotoxin that is essential for the pathogenesis of pneumonia. Strains lacking this cytotoxin are avirulent in a murine model of pneumonia; likewise, vaccine-based strategies that antagonize the toxin afford protection against lethal disease. Disruption of the function of this toxin therefore provides a potent mechanism to prevent and/or treat S. aureus pneumonia. beta-Cyclodextrin derivatives are small molecules with a sevenfold symmetry that mirrors the heptameric alpha-hemolysin. These compounds block the assembled alpha-hemolysin pore, compromising toxin function. We report that a modified beta-cyclodextrin compound, IB201, prevents alpha-hemolysin-induced lysis of human alveolar epithelial cells. This protective effect does not result from the ability of the beta-cyclodextrin to impair formation of the oligomeric alpha-hemolysin on the cell surface, supporting a role for this molecule in blockade of the lytic pore. An examination of IB201 in murine S. aureus pneumonia demonstrated that administration of this compound prevents and treats disease, protecting against mortality. Consistent with the vital importance of alpha-hemolysin in pneumonia caused by methicillin-sensitive and highly virulent methicillin-resistant S. aureus strains, IB201 protects against lethal challenge with both types of isolates. These observations, coupled with a favorable safety profile of beta-cyclodextrin compounds, provide a novel strategy that may be developed to combat S. aureus pneumonia.
Collapse
|
47
|
Meesters C, Brack A, Hellmann N, Decker H. Structural characterization of the alpha-hemolysin monomer from Staphylococcus aureus. Proteins 2009; 75:118-26. [PMID: 18798569 DOI: 10.1002/prot.22227] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Alpha-hemolysin from Staphylococcus aureus is secreted as a water-soluble monomer and assembles on membranes to oligomerize into a homo-heptameric, water-filled pore. These pores lead to lysis and cell death. Although the structure of the heptameric pore is solved by means of X-ray crystallography, structures of intermediate states-from the soluble monomer to all potential "pre-pore" structures-are yet unknown. Here, we propose a model of the monomeric alpha-hemolysin in solution based on molecular modeling, verified by small angle X-ray scattering data. This structure reveals details of the monomeric conformation of the alpha-hemolysin, for example inherent flexibility, along with definite differences in comparison to the structures used as templates.
Collapse
|
48
|
Anti-alpha-hemolysin monoclonal antibodies mediate protection against Staphylococcus aureus pneumonia. Infect Immun 2009; 77:2712-8. [PMID: 19380475 DOI: 10.1128/iai.00115-09] [Citation(s) in RCA: 162] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Staphylococcus aureus pneumonia is one of the most common invasive diseases caused by this human pathogen. S. aureus alpha-hemolysin, a pore-forming cytotoxin, is an essential virulence factor in the pathogenesis of pneumonia. Vaccine-based targeting of this toxin provides protection against lethal staphylococcal pneumonia in a murine model system, suggesting that a monoclonal antibody-based therapy may likewise prove to be efficacious for prevention and treatment of this disease. We report the generation of two distinct anti-alpha-hemolysin monoclonal antibodies that antagonize toxin activity, preventing human lung cell injury in vitro and protecting experimental animals against lethal S. aureus pneumonia. Each of these two monoclonal antibodies recognized an epitope within the first 50 amino acid residues of the mature toxin and blocked the formation of a stable alpha-hemolysin oligomer on the target cell surface. Active immunization with the first 50 amino acids of the toxin also conferred protection against S. aureus pneumonia. Together, these data reveal passive and active immunization strategies for prevention or therapy of staphylococcal pneumonia and highlight the potential role that a critical epitope may play in defining human susceptibility to this deadly disease.
Collapse
|
49
|
Löhner S, Walev I, Boukhallouk F, Palmer M, Bhakdi S, Angela V. Pore formation by
Vibrio cholerae
cytolysin follows the same archetypical mode as β‐barrel toxins from gram‐positive organisms. FASEB J 2009; 23:2521-8. [DOI: 10.1096/fj.08-127688] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Sabine Löhner
- Institute of Medical Microbiology and HygieneUniversity of MainzMainzGermany
| | - Iwan Walev
- Institute of Medical Microbiology and HygieneUniversity of MainzMainzGermany
| | - Fatima Boukhallouk
- Institute of Medical Microbiology and HygieneUniversity of MainzMainzGermany
| | - Michael Palmer
- Department of ChemistryUniversity of WaterlooWaterlooOntarioCanada
| | - Sucharit Bhakdi
- Institute of Medical Microbiology and HygieneUniversity of MainzMainzGermany
| | - Valeva Angela
- Institute of Medical Microbiology and HygieneUniversity of MainzMainzGermany
| |
Collapse
|
50
|
Poring over pores: alpha-hemolysin and Panton-Valentine leukocidin in Staphylococcus aureus pneumonia. Nat Med 2008; 13:1405-6. [PMID: 18064027 DOI: 10.1038/nm1207-1405] [Citation(s) in RCA: 385] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|