1
|
Yixuan L, Nakata M, Migita H, Matsumoto A, Oogai Y, Takebe K, Yamaguchi M, Okahashi N, Sumitomo T, Kawabata S. Identification of PilX, pilus component of Streptococcus sanguinis. J Oral Biosci 2025:100664. [PMID: 40254127 DOI: 10.1016/j.job.2025.100664] [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: 03/26/2025] [Revised: 04/15/2025] [Accepted: 04/17/2025] [Indexed: 04/22/2025]
Abstract
OBJECTIVES Streptococcus sanguinis is an oral commensal bacterium that promotes dental biofilm formation and causes infective endocarditis. S. sanguinis strain SK36 produces pili comprising PilA, PilB, and PilC. This study determined whether the ssa1635 gene adjacent to the pilus-related gene locus encodes a pilus component and its roles in biofilm formation and eukaryotic cell adhesion. METHODS Using a series of mutant strains and antisera against PilA, PilB, PilC, and SSA1635, immunoblot analyses and immunoprecipitation assays were performed for SSA1635 characterization. Both the involvement of the deduced pilus-specific transpeptidase SrtC in pilus assembly and SSA1635 localization were examined by immunoblot analysis of various mutant strains. Furthermore, biofilm formation assays on saliva-coated surfaces and adhesion to HeLa cells were performed to assess their functions. RESULTS SSA1635, designated as PilX, formed complexes with PilA, PilB, and PilC. PilX was identified as a tip pilin incorporated into the pilus structure by SrtC. Notably, the deletion of pilX impaired the polymerization of other pilins. Furthermore, a pilX deletion mutant exhibited decreased biofilm formation compared with the wild-type and revertant strains and comparable rates of adherence to HeLa cells. CONCLUSIONS PilX is a potential pilin tip that may aid in facilitating the polymerization of other pilins. PilX contributes to biofilm formation, although it appears to be dispensable for adhesion to HeLa cells. Further characterization of PilX-binding specificities will provide valuable insights into the colonization mechanisms of S. sanguinis.
Collapse
Affiliation(s)
- Li Yixuan
- Department of Microbiology, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Masanobu Nakata
- Department of Microbiology, Osaka University Graduate School of Dentistry, Osaka, Japan; Department of Oral Microbiology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan.
| | - Hirono Migita
- Department of Oral Microbiology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan; Department of Oral and Maxillofacial Surgery, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Airi Matsumoto
- Department of Oral Microbiology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Yuichi Oogai
- Department of Oral Microbiology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Katsuki Takebe
- Department of Microbiology, Osaka University Graduate School of Dentistry, Osaka, Japan; Department of Dental Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Japan
| | - Masaya Yamaguchi
- Department of Microbiology, Osaka University Graduate School of Dentistry, Osaka, Japan; Laboratory of Microbial Informatics, Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Nobuo Okahashi
- Department of Oral Microbiology and Immunology, Graduate School of Dentistry, Showa University, Tokyo, Japan
| | - Tomoko Sumitomo
- Department of Microbiology, Osaka University Graduate School of Dentistry, Osaka, Japan; Department of Oral Microbiology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Shigetada Kawabata
- Department of Microbiology, Osaka University Graduate School of Dentistry, Osaka, Japan; Center for Infectious Diseases Education and Research, Osaka University, Japan
| |
Collapse
|
2
|
Barua N, Chan BCL, Lau CBS, Leung PC, Fung KP, Ip M. Antivirulence Properties of Kuraridin Against Methicillin-Resistant Staphylococcus aureus (MRSA). Biomedicines 2025; 13:564. [PMID: 40149540 PMCID: PMC11940505 DOI: 10.3390/biomedicines13030564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 02/15/2025] [Accepted: 02/22/2025] [Indexed: 03/29/2025] Open
Abstract
Background: Methicillin-resistant Staphylococcus aureus (MRSA) is a major human opportunistic pathogen that causes a wide range of infections. The vast arsenal of virulence factors expressed remains the biggest challenge in treating MRSA with conventional antibiotic therapy. Methods: We investigated the effects of Kuraridin at subinhibitory minimum inhibition concentrations (MICs) of 1/8, 1/16, and 1/32 (concentrations that did not inhibit bacterial growth) on adhesion to fibrinogen, adhesion, internalization into HaCaT cells, and biofilm production in three MRSA strains representing the clonal types USA300, ST30, and ST239. Results: All three MRSA strains exhibited a significant decrease (p < 0.001) in adhesion to fibrinogen upon treatment with 1/8 and 1/16 MICs of Kuraridin. The adhesion and internalization of all the MRSA strains to HaCaT cells were decreased significantly (p < 0.001) upon treatment with the three subinhibitory concentrations of Kuraridin. The biofilm formation of USA300 (p < 0.001), ST30 (p < 0.001), and ST239 (p < 0.01) was significantly reduced at a 1/8 MIC. A significant decrease in biofilm formation at a 1/16 MIC was observed for USA300 (p < 0.001) and ST30 (p < 0.05). Confocal laser scanning microscopy (CSLM) analysis of the biofilms revealed a reduction in biofilm formation in the MRSA strain when treated with Kuraridin. In the in vivo Caenorhabditis elegans model, Kuraridin offered a sizable degree of protection against MRSA infection without being toxic to the nematode. Conclusions: Our findings reveal that Kuraridin has the potential to be an alternative antivirulence option for reducing MRSA pathogenicity.
Collapse
Affiliation(s)
- Nilakshi Barua
- Department of Microbiology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, N.T, Hong Kong 999077, China
| | - Ben Chung Lap Chan
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Shatin, N.T, Hong Kong 999077, China; (B.C.L.C.)
| | - Clara Bik-San Lau
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Shatin, N.T, Hong Kong 999077, China; (B.C.L.C.)
| | - Ping-Chung Leung
- Institute of Chinese Medicine and State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Shatin, N.T, Hong Kong 999077, China; (B.C.L.C.)
| | - Kwok Pui Fung
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T, Hong Kong 999077, China
| | - Margaret Ip
- Department of Microbiology, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, N.T, Hong Kong 999077, China
| |
Collapse
|
3
|
Giormezis N, Rechenioti A, Doumanas K, Sotiropoulos C, Paliogianni F, Kolonitsiou F. Bacteriophage Resistance, Adhesin's and Toxin's Genes Profile of Staphylococcus aureus Causing Infections in Children and Adolescents. Microorganisms 2025; 13:484. [PMID: 40142376 PMCID: PMC11946024 DOI: 10.3390/microorganisms13030484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 02/17/2025] [Accepted: 02/19/2025] [Indexed: 03/28/2025] Open
Abstract
Staphylococcus aureus is a common pathogen, often recovered from children's infections. Βiofilm formation, antimicrobial resistance and production of adhesins and toxins contribute to its virulence. As resistance to antimicrobials rises worldwide, alternative therapies like bacteriophages (among them the well-studied Bacteriophage K) can be helpful. The aim of this study was to determine the bacteriophage and antimicrobial susceptibility and the presence of virulence genes among S. aureus from infections in children and adolescents. Eighty S. aureus isolates were tested for biofilm formation and antimicrobial susceptibility. The presence of two genes of the ica operon (icaA, icaD), adhesin's (fnbA, fnbB, sasG) and toxin's genes (PVL, tst, eta, etb) was tested by PCRs. Susceptibility to Bacteriophage K was determined using a spot assay. Thirteen isolates were methicillin-resistant (MRSA) and 41 were multi-resistant. Twenty-five S. aureus (31.3%) were resistant to Bacteriophage K, mostly from ocular and ear infections. Twelve S. aureus (15%) were PVL-positive, seven (8.8%) positive for tst, 18 (22.5%) were eta-positive and 46 were (57.5%) etb-positive. A total of 66 (82.5%) isolates carried fnbA, 16 (20%) fnbB and 26 (32.5%) sasG. PVL, tst and sasG carriage were more frequent in MRSA. Bacteriophage-susceptible isolates carried more frequently eta (32.7%) and etb (69.1%) compared to phage-resistant S. aureus (0% and 32%, respectively). Although mainly methicillin-sensitive, S. aureus from pediatric infections exhibited high antimicrobial resistance and carriage of virulence genes (especially for exfoliative toxins and fnbA). MRSA was associated with PVL, tst and sasG carriage, whereas Bacteriophage susceptibility was associated with eta and etb. The high level of Bacteriophage K susceptibility highlights its potential use against staphylococcal infections.
Collapse
Affiliation(s)
- Nikolaos Giormezis
- Department of Microbiology, School of Medicine, University of Patras, 26504 Patras, Greece
| | - Assimina Rechenioti
- Department of Microbiology, School of Medicine, University of Patras, 26504 Patras, Greece
| | - Konstantinos Doumanas
- Department of Microbiology, School of Medicine, University of Patras, 26504 Patras, Greece
| | | | - Fotini Paliogianni
- Department of Microbiology, School of Medicine, University of Patras, 26504 Patras, Greece
| | - Fevronia Kolonitsiou
- Department of Microbiology, School of Medicine, University of Patras, 26504 Patras, Greece
| |
Collapse
|
4
|
Liu H, Wei X, Peng H, Yang Y, Hu Z, Rao Y, Wang Z, Dou J, Huang X, Hu Q, Tan L, Wang Y, Chen J, Liu L, Yang Y, Wu J, Hu X, Lu S, Shang W, Rao X. LysSYL-Loaded pH-Switchable Self-Assembling Peptide Hydrogels Promote Methicillin-Resistant Staphylococcus Aureus Elimination and Wound Healing. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2412154. [PMID: 39548922 DOI: 10.1002/adma.202412154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 10/26/2024] [Indexed: 11/18/2024]
Abstract
Staphylococcus aureus (S. aureus), especially methicillin-resistant S. aureus (MRSA), causes wound infections, whose treatment remains a clinical challenge. Bacterium-infected wounds often create acidic niches with a pH 4.5-6.5. Endolysin LysSYL, which is derived from phage SYL, shows promise as an antistaphylococcal agent. However, endolysins generally exhibit instability and possess low bioavailability in acidic microenvironments. Here, an array of self-assembling peptides is designed, and peptide L5 is screened out based on its gel formation property and bioavailability. L5 exerted a pH-switchable antimicrobial effect (pH 5.5) and formed biocompatible hydrogels at neutral pH (pH 7.4). The LysSYL-loaded L5 can assemble L5@LysSYL hydrogels, increase thermal stability, and exhibit the slow-release effect of LysSYL. Effective elimination of S. aureus is achieved by L5@LysSYL through bacterial membrane disruption and cell separation inhibition. Moreover, L5@LysSYL hydrogels exhibit great potential in promoting wound healing in a mouse wound model infected by MRSA. Furthermore, L5@LysSYL hydrogels are safe and can decrease the cytokine levels and increase the number of key factors for vessel formation, which contribute to wound healing. Overall, the self-assembling L5@LysSYL can effectively clean MRSA and promote wound healing, which suggests its potential as a pH-sensitive wound dressing for the management of wound infections.
Collapse
Affiliation(s)
- He Liu
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University, Key Laboratory of Microbial Engineering under the Educational Committee in Chongqing, Chongqing, 400038, China
| | - Xuemei Wei
- Institute of Biomedical Research, Southwest University, Chongqing, 400037, China
| | - Huagang Peng
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University, Key Laboratory of Microbial Engineering under the Educational Committee in Chongqing, Chongqing, 400038, China
| | - Yi Yang
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University, Key Laboratory of Microbial Engineering under the Educational Committee in Chongqing, Chongqing, 400038, China
| | - Zhen Hu
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University, Key Laboratory of Microbial Engineering under the Educational Committee in Chongqing, Chongqing, 400038, China
| | - Yifan Rao
- Department of Emergency Medicine, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Zhefen Wang
- Department of Neurology, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Jianxiong Dou
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University, Key Laboratory of Microbial Engineering under the Educational Committee in Chongqing, Chongqing, 400038, China
| | - Xiaonan Huang
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University, Key Laboratory of Microbial Engineering under the Educational Committee in Chongqing, Chongqing, 400038, China
| | - Qiwen Hu
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University, Key Laboratory of Microbial Engineering under the Educational Committee in Chongqing, Chongqing, 400038, China
| | - Li Tan
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University, Key Laboratory of Microbial Engineering under the Educational Committee in Chongqing, Chongqing, 400038, China
| | - Yuting Wang
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University, Key Laboratory of Microbial Engineering under the Educational Committee in Chongqing, Chongqing, 400038, China
| | - Juan Chen
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University, Key Laboratory of Microbial Engineering under the Educational Committee in Chongqing, Chongqing, 400038, China
| | - Lu Liu
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University, Key Laboratory of Microbial Engineering under the Educational Committee in Chongqing, Chongqing, 400038, China
| | - Yuhua Yang
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University, Key Laboratory of Microbial Engineering under the Educational Committee in Chongqing, Chongqing, 400038, China
| | - Jianghong Wu
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University, Key Laboratory of Microbial Engineering under the Educational Committee in Chongqing, Chongqing, 400038, China
| | - Xiaomei Hu
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University, Key Laboratory of Microbial Engineering under the Educational Committee in Chongqing, Chongqing, 400038, China
| | - Shuguang Lu
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University, Key Laboratory of Microbial Engineering under the Educational Committee in Chongqing, Chongqing, 400038, China
| | - Weilong Shang
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University, Key Laboratory of Microbial Engineering under the Educational Committee in Chongqing, Chongqing, 400038, China
| | - Xiancai Rao
- Department of Microbiology, College of Basic Medical Sciences, Army Medical University, Key Laboratory of Microbial Engineering under the Educational Committee in Chongqing, Chongqing, 400038, China
- Institute of Biomedical Research, Southwest University, Chongqing, 400037, China
| |
Collapse
|
5
|
Keim K, Bhattacharya M, Crosby HA, Jenul C, Mills K, Schurr M, Horswill A. Polymicrobial interactions between Staphylococcus aureus and Pseudomonas aeruginosa promote biofilm formation and persistence in chronic wound infections. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.04.621402. [PMID: 39574578 PMCID: PMC11580920 DOI: 10.1101/2024.11.04.621402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2024]
Abstract
Chronic, non-healing wounds are a leading cause of prolonged patient morbidity and mortality due to biofilm- associated, polymicrobial infections. Staphylococcus aureus and Pseudomonas aeruginosa are the most frequently co-isolated pathogens from chronic wound infections. Competitive interactions between these pathogens contribute to enhanced virulence, persistence, and antimicrobial tolerance. P. aeruginosa utilizes the extracellular proteases LasB, LasA, and AprA to degrade S. aureus surface structures, disrupt cellular physiology, and induce cell lysis, gaining a competitive advantage during co-infection. S. aureus evades P. aeruginosa by employing aggregation mechanisms to form biofilms. The cell wall protein SasG is implicated in S. aureus biofilm formation by facilitating intercellular aggregation upon cleavage by an extracellular protease. We have previously shown that proteolysis by a host protease can induce aggregation. In this study, we report that P. aeruginosa proteases LasA, LasB, and AprA cleave SasG to induce S. aureus aggregation. We demonstrate that SasG contributes to S. aureus biofilm formation in response to interactions with P. aeruginosa proteases by quantifying aggregation, SasG degradation, and proteolytic kinetics. Additionally, we assess the role of SasG in influencing S. aureus biofilm architecture during co-infection in vivo, chronic wound co-infections. This work provides further knowledge of some of the principal interactions that contribute to S. aureus persistence within chronic wounds co-infected with P. aeruginosa, and their impact on healing and infection outcomes.
Collapse
Affiliation(s)
- Klara Keim
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, United States of America
| | - Mohini Bhattacharya
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, United States of America
| | - Heidi A Crosby
- New England Biolabs, Ipswich, MA, United States of America
| | - Christian Jenul
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, United States of America
- Department of Genetics and Genome Biology, University of Leicester, Leicester, United Kingdom
| | - Krista Mills
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, United States of America
- Alphabet Health, New York, NY, United States of America
| | - Michael Schurr
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, United States of America
| | - Alexander Horswill
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO, United States of America
| |
Collapse
|
6
|
Rimal B, Chang JD, Liu C, Kim H, Aderotoye O, Zechmann B, Kim SJ. Scanning Electron Microscopy and Energy-Dispersive X-ray Spectroscopy of Staphylococcus aureus Biofilms. ACS OMEGA 2024; 9:37610-37620. [PMID: 39281927 PMCID: PMC11391442 DOI: 10.1021/acsomega.4c01168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 06/06/2024] [Accepted: 07/01/2024] [Indexed: 09/18/2024]
Abstract
Understanding the dynamics of biofilm formation and its elemental composition is crucial for developing effective strategies against biofilm-associated infections. In this study, we employed scanning electron microscopy (SEM) and energy-dispersive X-ray spectroscopy (EDS) to investigate the morphological changes and elemental compositions of Staphylococcus aureus biofilms. SEM images revealed distinct stages of biofilm development, from initial aggregation to the formation of mature and aged biofilms. EDS analysis consistently showed elevated levels of sodium (Na), oxygen (O), and phosphorus (P) in the biofilm matrix, indicating its high negative charge and the presence of anionic biopolymers. The incorporation of extracellular DNA (eDNA) into the biofilm matrix, leading to significant retention of sodium ions, underscored the importance of electrostatic interactions in biofilm formation and stability. Our findings highlight the potential of EDS analysis in quantifying elemental compositions and elucidating the role of anionic biopolymers in biofilm development.
Collapse
Affiliation(s)
- Binayak Rimal
- Institute of Biomedical Studies, Baylor University, Waco, Texas 76798, United States
| | - James D Chang
- Department of Chemistry and Biochemistry, Baylor University, Waco, Texas 76798, United States
| | - Chengyin Liu
- Department of Chemistry, Howard University, Washington, District of Columbia 20059, United States
| | - Haley Kim
- Department of Chemistry, University of Maryland, College Park, Maryland 20742, United States
| | - Oluwatobi Aderotoye
- Department of Chemistry, Howard University, Washington, District of Columbia 20059, United States
| | - Bernd Zechmann
- Center for Microscopy and Imaging, Baylor University, Waco, Texas 76798, United States
| | - Sung Joon Kim
- Department of Chemistry, Howard University, Washington, District of Columbia 20059, United States
| |
Collapse
|
7
|
Tang C, Jing W, Han K, Yang Z, Zhang S, Liu M, Zhang J, Zhao X, Liu Y, Shi C, Chai Q, Li Z, Han M, Wang Y, Fu Z, Zheng Z, Zhao K, Sun P, Zhu D, Chen C, Zhang D, Li D, Ni S, Li T, Cui J, Jiang X. mRNA-Laden Lipid-Nanoparticle-Enabled in Situ CAR-Macrophage Engineering for the Eradication of Multidrug-Resistant Bacteria in a Sepsis Mouse Model. ACS NANO 2024; 18:2261-2278. [PMID: 38207332 DOI: 10.1021/acsnano.3c10109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2024]
Abstract
Sepsis, which is the most severe clinical manifestation of acute infection and has a mortality rate higher than that of cancer, represents a significant global public health burden. Persistent methicillin-resistant Staphylococcus aureus (MRSA) infection and further host immune paralysis are the leading causes of sepsis-associated death, but limited clinical interventions that target sepsis have failed to effectively restore immune homeostasis to enable complete eradication of MRSA. To restimulate anti-MRSA innate immunity, we developed CRV peptide-modified lipid nanoparticles (CRV/LNP-RNAs) for transient in situ programming of macrophages (MΦs). The CRV/LNP-RNAs enabled the delivery of MRSA-targeted chimeric antigen receptor (CAR) mRNA (SasA-CAR mRNA) and CASP11 (a key MRSA intracellular evasion target) siRNA to MΦs in situ, yielding CAR-MΦs with boosted bactericidal potency. Specifically, our results demonstrated that the engineered MΦs could efficiently phagocytose and digest MRSA intracellularly, preventing immune evasion by the "superbug" MRSA. Our findings highlight the potential of nanoparticle-enabled in vivo generation of CAR-MΦs as a therapeutic platform for multidrug-resistant (MDR) bacterial infections and should be confirmed in clinical trials.
Collapse
Affiliation(s)
- Chunwei Tang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province 250012, China
| | - Weiqiang Jing
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Cultural West Road, Jinan, Shandong Province 250012, China
| | - Kun Han
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province 250012, China
| | - Zhenmei Yang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province 250012, China
| | - Shengchang Zhang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province 250012, China
| | - Miaoyan Liu
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province 250012, China
| | - Jing Zhang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province 250012, China
| | - Xiaotian Zhao
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province 250012, China
| | - Ying Liu
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province 250012, China
| | - Chongdeng Shi
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province 250012, China
| | - Qihao Chai
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province 250012, China
| | - Ziyang Li
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province 250012, China
| | - Maosen Han
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province 250012, China
| | - Yan Wang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province 250012, China
| | - Zhipeng Fu
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province 250012, China
| | - Zuolin Zheng
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province 250012, China
| | - Kun Zhao
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province 250012, China
| | - Peng Sun
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province 250355, China
| | - Danqing Zhu
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, 4572A Academic Building, Clear Water Bay, Kowloon, Hong Kong 999077, China
| | - Chen Chen
- Key Laboratory for Experimental Teratology of Ministry of Education, Key Laboratory of Infection and Immunity of Shandong Province and Department of Immunology, School of Basic Medical Sciences, Cheeloo Medical College of Shandong University, Jinan, Shandong Province 250012, China
| | - Daizhou Zhang
- Shandong Academy of Pharmaceutical Sciences, Jinan, Shandong Province 250101, China
| | - Dawei Li
- Shandong Academy of Pharmaceutical Sciences, Jinan, Shandong Province 250101, China
| | - Shilei Ni
- Department of Neurosurgery, Qilu Hospital and Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, 107 Cultural West Road, Jinan, Shandong Province 250012, China
| | - Tao Li
- Department of General Surgery, Qilu Hospital, Shandong University, 107 Cultural West Road, Jinan, Shandong Province 250012, China
| | - Jiwei Cui
- Key Laboratory of Colloid and Interface Chemistry of the Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan, Shandong Province 250100, China
| | - Xinyi Jiang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province 250012, China
| |
Collapse
|
8
|
Li Z, Zhang S, Fu Z, Liu Y, Man Z, Shi C, Tang C, Chen C, Chai Q, Yang Z, Zhang J, Zhao X, Xu H, Han M, Wang Y, Liao Z, Yu G, Shi B, Zhao K, Li W, Jiang X. Surficial nano-deposition locoregionally yielding bactericidal super CAR-macrophages expedites periprosthetic osseointegration. SCIENCE ADVANCES 2023; 9:eadg3365. [PMID: 37256944 PMCID: PMC10413653 DOI: 10.1126/sciadv.adg3365] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 04/20/2023] [Indexed: 06/02/2023]
Abstract
Tracking and eradicating Staphylococcus aureus in the periprosthetic microenvironment are critical for preventing periprosthetic joint infection (PJI), yet effective strategies remain elusive. Here, we report an implant nanoparticle coating that locoregionally yields bactericidal super chimeric antigen receptor macrophages (CAR-MΦs) to prevent PJI. We demonstrate that the plasmid-laden nanoparticle from the coating can introduce S. aureus-targeted CAR genes and caspase-11 short hairpin RNA (CASP11 shRNA) into macrophage nuclei to generate super CAR-MΦs in mouse models. CASP11 shRNA allowed mitochondria to be recruited around phagosomes containing phagocytosed bacteria to deliver mitochondria-generated bactericidal reactive oxygen species. These super CAR-MΦs targeted and eradicated S. aureus and conferred robust bactericidal immunologic activity at the bone-implant interface. Furthermore, the coating biodegradability precisely matched the bone regeneration process, achieving satisfactory osteogenesis. Overall, our work establishes a locoregional treatment strategy for priming macrophage-specific bactericidal immunity with broad application in patients suffering from multidrug-resistant bacterial infection.
Collapse
Affiliation(s)
- Ziyang Li
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province 250012, China
- Shandong Provincial Hospital, Shandong University, Jinan, Shandong Province 250021, China
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong Province 250021, China
| | - Shengchang Zhang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province 250012, China
| | - Zhipeng Fu
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province 250012, China
| | - Ying Liu
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province 250012, China
| | - Zhentao Man
- Shandong Provincial Hospital, Shandong University, Jinan, Shandong Province 250021, China
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong Province 250021, China
| | - Chongdeng Shi
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province 250012, China
| | - Chunwei Tang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province 250012, China
| | - Chen Chen
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province 250012, China
| | - Qihao Chai
- Shandong Provincial Hospital, Shandong University, Jinan, Shandong Province 250021, China
| | - Zhenmei Yang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province 250012, China
| | - Jing Zhang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province 250012, China
| | - Xiaotian Zhao
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province 250012, China
| | - Hailun Xu
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong Province 250021, China
| | - Maosen Han
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province 250012, China
| | - Yan Wang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province 250012, China
| | - Ziyang Liao
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province 250012, China
| | - Gongchang Yu
- Neck-Shoulder and Lumbocrural Pain Hospital of Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Bin Shi
- Neck-Shoulder and Lumbocrural Pain Hospital of Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Kun Zhao
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province 250012, China
| | - Wei Li
- Shandong Provincial Hospital, Shandong University, Jinan, Shandong Province 250021, China
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong Province 250021, China
| | - Xinyi Jiang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Cultural West Road, Jinan, Shandong Province 250012, China
| |
Collapse
|
9
|
Collagen-like Motifs of SasG: A Novel Fold for Protein Mechanical Strength. J Mol Biol 2023; 435:167980. [PMID: 36708761 DOI: 10.1016/j.jmb.2023.167980] [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: 10/18/2022] [Revised: 01/11/2023] [Accepted: 01/19/2023] [Indexed: 01/26/2023]
Abstract
The Staphylococcus aureus surface protein G (SasG) is associated with host colonisation and biofilm formation. As colonisation occurs at the liquid-substrate interface bacteria are subject to a myriad of external forces and, presumably as a consequence, SasG displays extreme mechanical strength. This mechanical phenotype arises from the B-domain; a repetitive region composed of alternating E and G5 subdomains. These subdomains have an unusual structure comprising collagen-like regions capped by triple-stranded β-sheets. To identify the determinants of SasG mechanical strength, we characterised the mechanical phenotype and thermodynamic stability of 18 single substitution variants of a pseudo-wildtype protein. Visualising the mechanically-induced transition state at a residue-level by ϕ-value analysis reveals that the main force-bearing regions are the N- and C-terminal 'Mechanical Clamps' and their side-chain interactions. This is tailored by contacts at the pseudo-hydrophobic core interface. We also describe a novel mechanical motif - the collagen-like region and show that glycine to alanine substitutions, analogous to those found in Osteogenesis Imperfecta (brittle bone disease), result in a significantly reduced mechanical strength.
Collapse
|
10
|
Malik A, Shoombuatong W, Kim CB, Manavalan B. GPApred: The first computational predictor for identifying proteins with LPXTG-like motif using sequence-based optimal features. Int J Biol Macromol 2023; 229:529-538. [PMID: 36596370 DOI: 10.1016/j.ijbiomac.2022.12.315] [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: 10/19/2022] [Revised: 12/19/2022] [Accepted: 12/28/2022] [Indexed: 01/02/2023]
Abstract
The cell surface proteins of gram-positive bacteria are involved in many important biological functions, including the infection of host cells. Owing to their virulent nature, these proteins are also considered strong candidates for potential drug or vaccine targets. Among the various cell surface proteins of gram-positive bacteria, LPXTG-like proteins form a major class. These proteins have a highly conserved C-terminal cell wall sorting signal, which consists of an LPXTG sequence motif, a hydrophobic domain, and a positively charged tail. These surface proteins are targeted to the cell envelope by a sortase enzyme via transpeptidation. A variety of LPXTG-like proteins have been experimentally characterized; however, their number in public databases has increased owing to extensive bacterial genome sequencing without proper annotation. In the absence of experimental characterization, identifying and annotating these sequences is extremely challenging. Therefore, in this study, we developed the first machine learning-based predictor called GPApred, which can identify LPXTG-like proteins from their primary sequences. Using a newly constructed benchmark dataset, we explored different classifiers and five feature encodings and their hybrids. Optimal features were derived using the recursive feature elimination method, and these features were then trained using a support vector machine algorithm. The performance of different models was evaluated using independent datasets, and a final model (GPApred) was selected based on consistency during cross-validation and independent assessment. GPApred can be an effective tool for predicting LPXTG-like sequences and can be further employed for functional characterization or drug targeting. Availability: https://procarb.org/gpapred/.
Collapse
Affiliation(s)
- Adeel Malik
- Institute of Intelligence Informatics Technology, Sangmyung University, Seoul 03016, Republic of Korea
| | - Watshara Shoombuatong
- Center of Data Mining and Biomedical Informatics, Faculty of Medical Technology, Mahidol University, Bangkok 10700, Thailand
| | - Chang-Bae Kim
- Department of Biotechnology, Sangmyung University, Seoul 03016, Republic of Korea.
| | - Balachandran Manavalan
- Computational Biology and Bioinformatics Laboratory, Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon 16419, Gyeonggi-do, Republic of Korea.
| |
Collapse
|
11
|
Maciag JJ, Chantraine C, Mills KB, Yadav R, Yarawsky AE, Chaton CT, Vinod D, Fitzkee NC, Mathelié-Guinlet M, Dufrêne YF, Fey PD, Horswill AR, Herr AB. Mechanistic basis of staphylococcal interspecies competition for skin colonization. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.26.525635. [PMID: 36747832 PMCID: PMC9900903 DOI: 10.1101/2023.01.26.525635] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Staphylococci, whether beneficial commensals or pathogens, often colonize human skin, potentially leading to competition for the same niche. In this multidisciplinary study we investigate the structure, binding specificity, and mechanism of adhesion of the Aap lectin domain required for Staphylococcus epidermidis skin colonization and compare its characteristics to the lectin domain from the orthologous Staphylococcus aureus adhesin SasG. The Aap structure reveals a legume lectin-like fold with atypical architecture, showing specificity for N-acetyllactosamine and sialyllactosamine. Bacterial adhesion assays using human corneocytes confirmed the biological relevance of these Aap-glycan interactions. Single-cell force spectroscopy experiments measured individual binding events between Aap and corneocytes, revealing an extraordinarily tight adhesion force of nearly 900 nN and a high density of receptors at the corneocyte surface. The SasG lectin domain shares similar structural features, glycan specificity, and corneocyte adhesion behavior. We observe cross-inhibition of Aap-and SasG-mediated staphylococcal adhesion to corneocytes. Together, these data provide insights into staphylococcal interspecies competition for skin colonization and suggest potential avenues for inhibition of S. aureus colonization.
Collapse
Affiliation(s)
- Joseph J. Maciag
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Constance Chantraine
- Louvain Institute of Biomolecular Science and Technology, UCLouvain, Louvain-la-Neuve, Belgium
| | - Krista B. Mills
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO
| | - Rahul Yadav
- Department of Chemistry, Mississippi State University, Mississippi State, MS
| | - Alexander E. Yarawsky
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Catherine T. Chaton
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Divya Vinod
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- Medical Sciences Undergraduate Program, University of Cincinnati, Cincinnati, OH
| | - Nicholas C. Fitzkee
- Department of Chemistry, Mississippi State University, Mississippi State, MS
| | - Marion Mathelié-Guinlet
- Institut de Chimie et Biologie des Membranes et des Nano-Objets, CNRS UMR 5248, University of Bordeaux, Pessac, France
| | - Yves F. Dufrêne
- Louvain Institute of Biomolecular Science and Technology, UCLouvain, Louvain-la-Neuve, Belgium
| | - Paul D. Fey
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE
| | - Alexander R. Horswill
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO
| | - Andrew B. Herr
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- Division of Infectious Diseases, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
| |
Collapse
|
12
|
Su X, Yu H, Wang X, Zhang C, Wang H, Kong X, Qu Y, Luan Y, Meng Y, Guan J, Song G, Wang L, Song W, Zhao Y. Cyanidin chloride protects mice from methicillin-resistant Staphylococcus aureus-induced pneumonia by targeting Sortase A. Virulence 2022; 13:1434-1445. [PMID: 35983964 PMCID: PMC9397467 DOI: 10.1080/21505594.2022.2112831] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) has been developing rapidly in recent years. It poses a severe peril to global health care, and the new strategies to against the MRSA is urgently needed. Sortase A (SrtA) regulates the anchoring of many surface proteins. Compounds repress Staphylococcus aureus (S. aureus) cysteine transpeptidase SrtA are considered adequate potent virulence inhibitors. Then, we describe the identification of an effective SrtA inhibitor, cyanidin chloride, a bioflavonoid compound isolated from various plants. It has a reversible inhibitory effect on SrtA activity at an IC50 of 21.91 μg/mL. As a SrtA inhibitor, cyanidin chloride antagonizes SrtA-related virulence phenotypes due to its breadth and specificity, including fibrinogen adhesion, A549 cell invasion, biofilm formation, and surface protein (SpA) anchoring. Subsequently, molecular docking and fluorescence quenching revealed that SrtA and cyanidin chloride had robust mutual affinity. Further mechanistic studies revealed that Arg-197, Gly-167, and Sep-116 were the key-binding sites mediating the interaction between SrtA and cyanidin chloride. Notably, a significant therapeutic effect of cyanidin chloride in vivo was also observed on the mouse pneumonia model induced by MRSA. In conclusion, our study indicates that cyanidin chloride potentially represents a new candidate SrtA inhibitor for S. aureus and potentially be developed as a new antivirulence agent.
Collapse
Affiliation(s)
- Xin Su
- College of Clinical Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Hangqian Yu
- Center for Pathogen Biology and Infectious Diseases, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, China
| | - Xingye Wang
- College of Clinical Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Chi Zhang
- College of Clinical Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Heming Wang
- Department of Gastroenterology, Zhongshan Hospital of Fudan University, Shanghai, China
| | - Xiangri Kong
- College of Clinical Medicine, Changchun University of Chinese Medicine, Changchun, China,College of Animal Science, Jilin University, Changchun, China
| | - Yishen Qu
- Endocrinology Department, Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Yanhe Luan
- College of Animal Science, Jilin University, Changchun, China
| | - Ying Meng
- College of Clinical Medicine, Changchun University of Chinese Medicine, Changchun, China
| | - Jiyu Guan
- Department of Anesthesiology, Peking University Third Hospital, Beijing, China
| | - Guangqi Song
- Department of Gastroenterology, Zhongshan Hospital of Fudan University, Shanghai, China
| | - Li Wang
- College of Clinical Medicine, Changchun University of Chinese Medicine, Changchun, China,CONTACT Li Wang
| | - Wu Song
- College of Clinical Medicine, Changchun University of Chinese Medicine, Changchun, China,Wu Song
| | - Yicheng Zhao
- College of Clinical Medicine, Changchun University of Chinese Medicine, Changchun, China,Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China,Yicheng Zhao
| |
Collapse
|
13
|
Grousd JA, Dresden BP, Riesmeyer AM, Cooper VS, Bomberger JM, Richardson AR, Alcorn JF. Novel Requirement for Staphylococcal Cell Wall-Anchored Protein SasD in Pulmonary Infection. Microbiol Spectr 2022; 10:e0164522. [PMID: 36040164 PMCID: PMC9603976 DOI: 10.1128/spectrum.01645-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 08/16/2022] [Indexed: 02/04/2023] Open
Abstract
Staphylococcus aureus can complicate preceding viral infections, including influenza virus. A bacterial infection combined with a preceding viral infection, known as superinfection, leads to worse outcomes than a single infection. Most of the pulmonary infection literature focuses on the changes in immune responses to bacteria between homeostatic and virally infected lungs. However, it is unclear how much of an influence bacterial virulence factors have in single or superinfection. Staphylococcal species express a broad range of cell wall-anchored proteins (CWAs) that have roles in host adhesion, nutrient acquisition, and immune evasion. We screened the importance of these CWAs using mutants lacking individual CWAs in vivo in both bacterial pneumonia and influenza superinfection. In bacterial pneumonia, the lack of individual CWAs leads to various decreases in bacterial burden, lung damage, and immune infiltration into the lung. However, the presence of a preceding influenza infection partially abrogates the requirement for CWAs. In the screen, we found that the uncharacterized CWA S. aureus surface protein D (SasD) induced changes in both inflammatory and homeostatic lung markers. We further characterized a SasD mutant (sasD A50.1) in the context of pneumonia. Mice infected with sasD A50.1 have decreased bacterial burden, inflammatory responses, and mortality compared to wild-type S. aureus. Mice also have reduced levels of interleukin-1β (IL-1β), likely derived from macrophages. Reductions in IL-1β transcript levels as well as increased macrophage viability point at differences in cell death pathways. These data identify a novel virulence factor for S. aureus that influences inflammatory signaling within the lung. IMPORTANCE Staphylococcus aureus is a common commensal bacterium that can cause severe infections, such as pneumonia. In the lung, viral infections increase the risk of staphylococcal pneumonia, leading to combined infections known as superinfections. The most common virus associated with S. aureus pneumonia is influenza, and superinfections lead to worse patient outcomes than either infection alone. While there is much known about how the immune system differs between healthy and virally infected lungs, the role of bacterial virulence factors in single and superinfection is less understood. The significance of our research is identifying bacterial components that play a role in the initiation of lung injury, which could lead to future therapies to prevent pulmonary single or superinfection with S. aureus.
Collapse
Affiliation(s)
- Jennifer A. Grousd
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Pediatrics, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Brooke P. Dresden
- Department of Pediatrics, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Abigail M. Riesmeyer
- Department of Pediatrics, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Vaughn S. Cooper
- Department of Microbiology & Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Jennifer M. Bomberger
- Department of Microbiology & Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Anthony R. Richardson
- Department of Microbiology & Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - John F. Alcorn
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Pediatrics, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
14
|
The long and the short of Periscope Proteins. Biochem Soc Trans 2022; 50:1293-1302. [PMID: 36196877 DOI: 10.1042/bst20220194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 09/10/2022] [Accepted: 09/13/2022] [Indexed: 11/17/2022]
Abstract
Bacteria sense, interact with, and modify their environmental niche by deploying a molecular ensemble at the cell surface. The changeability of this exposed interface, combined with extreme changes in the functional repertoire associated with lifestyle switches from planktonic to adherent and biofilm states necessitate dynamic variability. Dynamic surface changes include chemical modifications to the cell wall; export of diverse extracellular biofilm components; and modulation of expression of cell surface proteins for adhesion, co-aggregation and virulence. Local enrichment for highly repetitive proteins with high tandem repeat identity has been an enigmatic phenomenon observed in diverse bacterial species. Preliminary observations over decades of research suggested these repeat regions were hypervariable, as highly related strains appeared to express homologues with diverse molecular mass. Long-read sequencing data have been interrogated to reveal variation in repeat number; in combination with structural, biophysical and molecular dynamics approaches, the Periscope Protein class has been defined for cell surface attached proteins that dynamically expand and contract tandem repeat tracts at the population level. Here, I review the diverse high-stability protein folds and coherent interdomain linkages culminating in the formation of highly anisotropic linear repeat arrays, so-called rod-like protein 'stalks', supporting roles in bacterial adhesion, biofilm formation, cell surface spatial competition, and immune system modulation. An understanding of the functional impacts of dynamic changes in repeat arrays and broader characterisation of the unusual protein folds underpinning this variability will help with the design of immunisation strategies, and contribute to synthetic biology approaches including protein engineering and microbial consortia construction.
Collapse
|
15
|
The C-Terminal Domain of Staphylococcus aureus Zinc Transport Protein AdcA Binds Plasminogen and Factor H In Vitro. Pathogens 2022; 11:pathogens11020240. [PMID: 35215183 PMCID: PMC8878332 DOI: 10.3390/pathogens11020240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 02/08/2022] [Accepted: 02/09/2022] [Indexed: 11/17/2022] Open
Abstract
Bacterial acquisition of metals from a host is an essential attribute to facilitate survival and colonization within an infected organism. Staphylococcus aureus, a bacterial pathogen of medical importance, has evolved its strategies to acquire multiple metals, including iron, manganese, and zinc. Other important strategies for the colonization and infection of the host have been reported for staphylococci and include the expression of adhesins on the bacterial surface, as well as the acquisition of host plasminogen and complement regulatory proteins. Here we assess the ability of the zinc transport protein AdcA from Staphylococcus aureus, first characterized elsewhere as a zinc-binding protein of the ABC (ATP-binding cassette) transporters, to bind to host molecules. Like other staphylococcus ion-scavenging proteins, such as MntC, a manganese-binding protein, AdcA interacts with human plasminogen. Once activated, plasmin bound to AdcA cleaves fibrinogen and vitronectin. In addition, AdcA interacts with the human negative complement regulator factor H (FH). Plasminogen and FH have been shown to bind to distinct sites on the AdcA C-terminal portion. In conclusion, our in vitro data pave the way for future studies addressing the relevance of AdcA interactions with host molecules in vivo.
Collapse
|
16
|
Antimicrobial characterization of a titanium coating derived from mussel-glue and Bothrops asper snake venom for the prevention of implant-associated infections caused by Staphylococcus. ELECTRON J BIOTECHN 2022. [DOI: 10.1016/j.ejbt.2022.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
17
|
Araujo-Alves AV, Kraychete GB, Gilmore MS, Barros EM, Giambiagi-deMarval M. shsA: A novel orthologous of sasX/sesI virulence genes is detected in Staphylococcus haemolyticus Brazilian strains. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2022; 97:105189. [PMID: 34920103 DOI: 10.1016/j.meegid.2021.105189] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 12/06/2021] [Accepted: 12/12/2021] [Indexed: 06/14/2023]
Abstract
The surface protein SasX, has a key role in methicillin-resistant Staphylococcus aureus (MRSA) colonization and pathogenesis, and has been associated with the epidemic success of some MRSA clones. To date, only one SasX homologous protein, named SesI, has been described in Staphylococcus epidermidis. In this work, we analyze the occurrence of the sasX gene and its genetic environment in Staphylococcus haemolyticus S. haemolyticus clinical strains (n = 62) were screened for the presence of the sasX gene and its carrier, the prophage Φ SPβ-like. A deep characterization was done in one strain (MD43), through which we determined the complete nucleotide sequence for the S. haemolitycus sasX-like gene. Whole genome sequencing of strain MD43 was performed, and the gene, termed here because of its unique attributes, shsA, was mapped to the Φ SPβ-like prophage sequence. The shsA gene was detected in 33 out of 62 strains showing an average identity of 92 and 96% with the sasX and sesI genes and at the amino acid level, 88% identity with SasX and 92% identity with SesI. The ~124Kb Φ SPβ-like prophage sequence showed a largely intact prophage compared to its counterpart in S. epidermidis strain RP62A, including the sesI insertion site. In conclusion, we identified a new sasX ortholog in S. haemolyticus (shsA). Its horizontal spread from this reservoir could represent an emergent threat in healthcare facilities since so far, no S. aureus sasX+ strains have been reported in Brazil.
Collapse
Affiliation(s)
- Amanda V Araujo-Alves
- Departamento de Microbiologia Médica, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Gabriela B Kraychete
- Departamento de Microbiologia Médica, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Michael S Gilmore
- Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Boston, MA 02114, USA; Department of Microbiology, Harvard Medical School, Boston, MA 02114, USA; Infectious Disease and Microbiome Program, Broad Institute, Cambridge, MA 02142, USA
| | - Elaine M Barros
- Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Boston, MA 02114, USA
| | - Marcia Giambiagi-deMarval
- Departamento de Microbiologia Médica, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil..
| |
Collapse
|
18
|
Carrera-Salinas A, González-Díaz A, Vázquez-Sánchez DA, Camoez M, Niubó J, Càmara J, Ardanuy C, Martí S, Domínguez MÁ, Garcia M, Marco F, Chaves F, Cercenado E, Tapiol J, Xercavins M, Fontanals D, Loza E, Rodríguez-López F, Olarte I, Mirelis B, Ruiz de Gopegui E, Lepe J, Larrosa N. Staphylococcus aureus surface protein G (sasG) allelic variants: correlation between biofilm formation and their prevalence in methicillin-resistant S. aureus (MRSA) clones. Res Microbiol 2022; 173:103921. [DOI: 10.1016/j.resmic.2022.103921] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 01/12/2022] [Accepted: 01/14/2022] [Indexed: 01/02/2023]
|
19
|
Bai Q, Ma J, Zhang Z, Zhong X, Pan Z, Zhu Y, Zhang Y, Wu Z, Liu G, Yao H. YSIRK-G/S-directed translocation is required for Streptococcus suis to deliver diverse cell wall anchoring effectors contributing to bacterial pathogenicity. Virulence 2021; 11:1539-1556. [PMID: 33138686 PMCID: PMC7644249 DOI: 10.1080/21505594.2020.1838740] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The Streptococcus suis serotype 2 (SS2) is a significant zoonotic pathogen that is responsible for various swine diseases, even causing cytokine storms of Streptococcal toxic shock-like syndromes amongst human. Cell wall anchoring proteins with a C-terminal LPxTG are considered to play vital roles during SS2 infection; however, their exporting mechanism across cytoplasmic membranes has remained vague. This study found that YSIRK-G/S was involved in the exportation of LPxTG-anchoring virulence factors MRP and SspA in virulent SS2 strain ZY05719. The whole-genome analysis indicated that diverse LPxTG proteins fused with an N-terminal YSIRK-G/S motif are encoded in strain ZY05719. Two novel LPxTG proteins SspB and YzpA were verified to be exported via a putative transport system that was dependent on the YSIRK-G/S directed translocation, and portrayed vital functions during the infection of SS2 strain ZY05719. Instead of exhibiting an inactivation of C5a peptidase in SspB, another LPxTG protein with an N-terminal YSIRK-G/S motif from Streptococcus agalactiae was depicted to cleave the C5a component of the host complement. The consequent domain-architecture retrieval determined more than 10,000 SspB/YzpA like proteins that are extensively distributed in the Gram-positive bacteria, and most of them harbor diverse glycosyl hydrolase or peptidase domains within their middle regions, thus presenting their capability to interact with host cells. The said findings provide compelling evidence that LPxTG proteins with an N-terminal YSIRK-G/S motif are polymorphic effectors secreted by Gram-positive bacteria, which can be further proposed to define as cell wall anchoring effectors in a new subset.
Collapse
Affiliation(s)
- Qiankun Bai
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University , Nanjing, China.,Key Lab of Animal Bacteriology, Ministry of Agriculture , Nanjing, China.,Department of pathogenic diagnosis, OIE Reference Lab for Swine Streptococcosis , Nanjing, China
| | - Jiale Ma
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University , Nanjing, China.,Key Lab of Animal Bacteriology, Ministry of Agriculture , Nanjing, China.,Department of pathogenic diagnosis, OIE Reference Lab for Swine Streptococcosis , Nanjing, China
| | - Ze Zhang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University , Nanjing, China.,Key Lab of Animal Bacteriology, Ministry of Agriculture , Nanjing, China.,Department of pathogenic diagnosis, OIE Reference Lab for Swine Streptococcosis , Nanjing, China
| | - Xiaojun Zhong
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University , Nanjing, China.,Key Lab of Animal Bacteriology, Ministry of Agriculture , Nanjing, China.,Department of pathogenic diagnosis, OIE Reference Lab for Swine Streptococcosis , Nanjing, China
| | - Zihao Pan
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University , Nanjing, China.,Key Lab of Animal Bacteriology, Ministry of Agriculture , Nanjing, China.,Department of pathogenic diagnosis, OIE Reference Lab for Swine Streptococcosis , Nanjing, China
| | - Yinchu Zhu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University , Nanjing, China.,Key Lab of Animal Bacteriology, Ministry of Agriculture , Nanjing, China.,Department of pathogenic diagnosis, OIE Reference Lab for Swine Streptococcosis , Nanjing, China
| | - Yue Zhang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University , Nanjing, China.,Key Lab of Animal Bacteriology, Ministry of Agriculture , Nanjing, China.,Department of pathogenic diagnosis, OIE Reference Lab for Swine Streptococcosis , Nanjing, China
| | - Zongfu Wu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University , Nanjing, China.,Key Lab of Animal Bacteriology, Ministry of Agriculture , Nanjing, China.,Department of pathogenic diagnosis, OIE Reference Lab for Swine Streptococcosis , Nanjing, China
| | - Guangjin Liu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University , Nanjing, China.,Key Lab of Animal Bacteriology, Ministry of Agriculture , Nanjing, China.,Department of pathogenic diagnosis, OIE Reference Lab for Swine Streptococcosis , Nanjing, China
| | - Huochun Yao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University , Nanjing, China.,Key Lab of Animal Bacteriology, Ministry of Agriculture , Nanjing, China.,Department of pathogenic diagnosis, OIE Reference Lab for Swine Streptococcosis , Nanjing, China
| |
Collapse
|
20
|
Plota M, Sazakli E, Giormezis N, Gkartziou F, Kolonitsiou F, Leotsinidis M, Antimisiaris SG, Spiliopoulou I. In Vitro Anti-Biofilm Activity of Bacteriophage K (ATCC 19685-B1) and Daptomycin against Staphylococci. Microorganisms 2021; 9:1853. [PMID: 34576751 PMCID: PMC8468654 DOI: 10.3390/microorganisms9091853] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 08/24/2021] [Accepted: 08/27/2021] [Indexed: 11/17/2022] Open
Abstract
The purpose of the present study was to investigate anti-staphylococcal activity of daptomycin and bacteriophage K, alone or in combination, against biofilm-producers and non-producers S. aureus and S. epidermidis strains, under biofilm forming and cells' proliferation conditions. Daptomycin and bacteriophage K (ATCC 19685B1), in different concentrations, were tested against 10 Staphylococcus aureus and 10 S. epidermidis, characterized by phenotypes and genotypes. The quantitative microtiter plate (crystal violet, CV), methylthiazoltetrazolium (MTT), and growth curve (GC) assays were performed. No statistically significant difference was found between species, whereas daptomycin alone performed better using medium and high concentrations of the drug and bacteriophage K was more active against strains with higher susceptibility, by CV and MTT assays. Best results were achieved using both agents combined in high concentrations. Bacteriophage K was effective within 3.8 and 2.4 h, depending on the concentration used, by the GC assay. Combination of daptomycin with bacteriophage K was more effective against staphylococci, depending on the concentrations used and strains' susceptibility. Further studies are needed to evaluate if this approach might be a choice for prevention or therapy of biofilm-associated infections.
Collapse
Affiliation(s)
- Maria Plota
- Department of Microbiology, School of Medicine, University of Patras, 26504 Patras, Greece; (M.P.); (F.K.)
- National Reference Centre for Staphylococci, School of Medicine, University of Patras, 26504 Patras, Greece;
| | - Eleni Sazakli
- Laboratory of Public Health, School of Medicine, University of Patras, 26504 Patras, Greece; (E.S.); (M.L.)
| | - Nikolaos Giormezis
- National Reference Centre for Staphylococci, School of Medicine, University of Patras, 26504 Patras, Greece;
| | - Foteini Gkartziou
- Institute of Chemical Engineering Sciences, FORTH/ICE-HT, Platani, 26504 Patras, Greece; (F.G.); (S.G.A.)
| | - Fevronia Kolonitsiou
- Department of Microbiology, School of Medicine, University of Patras, 26504 Patras, Greece; (M.P.); (F.K.)
- National Reference Centre for Staphylococci, School of Medicine, University of Patras, 26504 Patras, Greece;
| | - Michalis Leotsinidis
- Laboratory of Public Health, School of Medicine, University of Patras, 26504 Patras, Greece; (E.S.); (M.L.)
| | - Sophia G. Antimisiaris
- Institute of Chemical Engineering Sciences, FORTH/ICE-HT, Platani, 26504 Patras, Greece; (F.G.); (S.G.A.)
- Department of Pharmacy, School of Health Sciences, University of Patras, 26504 Patras, Greece
| | - Iris Spiliopoulou
- National Reference Centre for Staphylococci, School of Medicine, University of Patras, 26504 Patras, Greece;
| |
Collapse
|
21
|
Cardoso Guimarães L, Marques de Souza B, de Oliveira Whitaker C, Abreu F, Barreto Rocha Ferreira R, Dos Santos KRN. Increased biofilm formation by Staphylococcus aureus clinical isolates on surfaces covered with plasma proteins. J Med Microbiol 2021; 70. [PMID: 34338626 DOI: 10.1099/jmm.0.001389] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Introduction. Biofilm formation is a major virulence factor associated with Staphylococcus aureus infections. However, the influence of plasma proteins on biofilm formation of clinical isolates in vitro remains unclear.Hypotheses. We hypothesized that coating surfaces with plasma proteins might induce biofilm formation by S. aureus of different clonal lineages.Aim. To evaluate biofilm production by clinical S. aureus isolates of different clonal lineages isolated in Rio de Janeiro hospitals and investigated the presence of biofilm-associated genes.Methodology. This study assessed biofilm production of 60 S. aureus isolates in polystyrene microtitre plates with and without fibrinogen or fibronectin. The biochemical composition of the biofilm matrices was determined and the biofilm formation on fibrinogen-coated surfaces was also evaluated by confocal laser scanning microscopy. The presence of biofilm-related genes was detected by PCR, and the typing and functionality of agr operon was also evaluated.Results. Most of the isolates (45 %) were weak biofilm producers or non-producers. However, most of them presented a significant increase in biofilm production on plates covered with plasma proteins. There was no significant difference in biofilm formation between methicillin-resistant and -susceptible S. aureus isolates, or between different clonal lineages, except for ST30-IV (weak producers) and ST239-III (strong producers). The fnbB gene was associated with higher biofilm production.Conclusion. An increase in biofilm production in the presence of plasma proteins highlights the importance of investigating biofilm formation by S. aureus clinical isolates under different conditions since this virulence factor contributes to persistent infections and increased resistance to antimicrobials.
Collapse
Affiliation(s)
- Lorrayne Cardoso Guimarães
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Bruna Marques de Souza
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | | | - Fernanda Abreu
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | | | | |
Collapse
|
22
|
Periscope Proteins are variable-length regulators of bacterial cell surface interactions. Proc Natl Acad Sci U S A 2021; 118:2101349118. [PMID: 34074781 PMCID: PMC8201768 DOI: 10.1073/pnas.2101349118] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
The structure of single and tandem SHIRT domains from the streptococcal surface protein Sgo_0707 were determined. In conjunction with biophysics and molecular dynamics simulations, the results show that the observed gene length variation would result in differential projection of the host ligand binding domain on the bacterial cell surface. An analysis of long-read DNA sequence data reveals many other repetitive bacterial surface proteins that appear to undergo gene length variation. We propose that these variable-length “Periscope Proteins” represent an important mechanism of bacterial cell surface modification with potential roles in infection and immune evasion. Changes at the cell surface enable bacteria to survive in dynamic environments, such as diverse niches of the human host. Here, we reveal “Periscope Proteins” as a widespread mechanism of bacterial surface alteration mediated through protein length variation. Tandem arrays of highly similar folded domains can form an elongated rod-like structure; thus, variation in the number of domains determines how far an N-terminal host ligand binding domain projects from the cell surface. Supported by newly available long-read genome sequencing data, we propose that this class could contain over 50 distinct proteins, including those implicated in host colonization and biofilm formation by human pathogens. In large multidomain proteins, sequence divergence between adjacent domains appears to reduce interdomain misfolding. Periscope Proteins break this “rule,” suggesting that their length variability plays an important role in regulating bacterial interactions with host surfaces, other bacteria, and the immune system.
Collapse
|
23
|
Thappeta KRV, Zhao LN, Nge CE, Crasta S, Leong CY, Ng V, Kanagasundaram Y, Fan H, Ng SB. In-Silico Identified New Natural Sortase A Inhibitors Disrupt S. aureus Biofilm Formation. Int J Mol Sci 2020; 21:ijms21228601. [PMID: 33202690 PMCID: PMC7696255 DOI: 10.3390/ijms21228601] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 11/09/2020] [Accepted: 11/12/2020] [Indexed: 12/12/2022] Open
Abstract
Sortase A (SrtA) is a membrane-associated enzyme that anchors surface-exposed proteins to the cell wall envelope of Gram-positive bacteria such as Staphylococcus aureus. As SrtA is essential for Gram-positive bacterial pathogenesis but dispensable for microbial growth or viability, SrtA is considered a favorable target for the enhancement of novel anti-infective drugs that aim to interfere with key bacterial virulence mechanisms, such as biofilm formation, without developing drug resistance. Here, we used virtual screening to search an in-house natural compound library and identified two natural compounds, N1287 (Skyrin) and N2576 ((4,5-dichloro-1H-pyrrol-2-yl)-[2,4-dihydroxy-3-(4-methyl-pentyl)-phenyl]-methanone) that inhibited the enzymatic activity of SrtA. These compounds also significantly reduced the growth of S. aureus but possessed moderate mammalian toxicity. Furthermore, S. aureus strains treated with these compounds exhibited reduction in adherence to host fibrinogen, as well as biofilm formation. Hence, these compounds may represent an anti-infective therapy without the side effects of antibiotics.
Collapse
Affiliation(s)
- Kishore Reddy Venkata Thappeta
- Singapore Institute of Food and Biotechnology Innovation (SIFBI), Agency for Science, Technology and Research (A*STAR), 31 Biopolis Way, #01-02 Nanos, Singapore 138669, Singapore; (K.R.V.T.); (C.E.N.); (S.C.); (C.Y.L.); (V.N.)
| | - Li Na Zhao
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), 30 Biopolis Street, #07-01 Matrix, Singapore 138671, Singapore;
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, #3-09 Proteos, Singapore 138673, Singapore
| | - Choy Eng Nge
- Singapore Institute of Food and Biotechnology Innovation (SIFBI), Agency for Science, Technology and Research (A*STAR), 31 Biopolis Way, #01-02 Nanos, Singapore 138669, Singapore; (K.R.V.T.); (C.E.N.); (S.C.); (C.Y.L.); (V.N.)
| | - Sharon Crasta
- Singapore Institute of Food and Biotechnology Innovation (SIFBI), Agency for Science, Technology and Research (A*STAR), 31 Biopolis Way, #01-02 Nanos, Singapore 138669, Singapore; (K.R.V.T.); (C.E.N.); (S.C.); (C.Y.L.); (V.N.)
| | - Chung Yan Leong
- Singapore Institute of Food and Biotechnology Innovation (SIFBI), Agency for Science, Technology and Research (A*STAR), 31 Biopolis Way, #01-02 Nanos, Singapore 138669, Singapore; (K.R.V.T.); (C.E.N.); (S.C.); (C.Y.L.); (V.N.)
| | - Veronica Ng
- Singapore Institute of Food and Biotechnology Innovation (SIFBI), Agency for Science, Technology and Research (A*STAR), 31 Biopolis Way, #01-02 Nanos, Singapore 138669, Singapore; (K.R.V.T.); (C.E.N.); (S.C.); (C.Y.L.); (V.N.)
| | - Yoganathan Kanagasundaram
- Singapore Institute of Food and Biotechnology Innovation (SIFBI), Agency for Science, Technology and Research (A*STAR), 31 Biopolis Way, #01-02 Nanos, Singapore 138669, Singapore; (K.R.V.T.); (C.E.N.); (S.C.); (C.Y.L.); (V.N.)
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), 30 Biopolis Street, #07-01 Matrix, Singapore 138671, Singapore;
- Correspondence: (Y.K.); (H.F.); (S.B.N.); Tel.: +65-6586-9508 (Y.K.); +65-6478-8500 (H.F.); +65-6478-8513 (S.B.N.)
| | - Hao Fan
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), 30 Biopolis Street, #07-01 Matrix, Singapore 138671, Singapore;
- Correspondence: (Y.K.); (H.F.); (S.B.N.); Tel.: +65-6586-9508 (Y.K.); +65-6478-8500 (H.F.); +65-6478-8513 (S.B.N.)
| | - Siew Bee Ng
- Singapore Institute of Food and Biotechnology Innovation (SIFBI), Agency for Science, Technology and Research (A*STAR), 31 Biopolis Way, #01-02 Nanos, Singapore 138669, Singapore; (K.R.V.T.); (C.E.N.); (S.C.); (C.Y.L.); (V.N.)
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), 30 Biopolis Street, #07-01 Matrix, Singapore 138671, Singapore;
- Correspondence: (Y.K.); (H.F.); (S.B.N.); Tel.: +65-6586-9508 (Y.K.); +65-6478-8500 (H.F.); +65-6478-8513 (S.B.N.)
| |
Collapse
|
24
|
Ménard G, Bonnaure-Mallet M, Donnio PY. Adhesion of Staphylococcus aureus to epithelial cells: an in vitro approach to study interactions within the nasal microbiota. J Med Microbiol 2020; 69:1253-1261. [PMID: 32909934 DOI: 10.1099/jmm.0.001248] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Introduction. Staphylococcus aureus is a skin and mucous commensal bacterium of warm-blooded animals. In humans, the nose is the main ecological niche of S. aureus, and nasal carriage is a risk factor for developing an endogenous infection. S. aureus nasal colonization is a multifactorial process, involving inter-species interactions among the nasal microbiota.Aims. The objectives of this study were to characterize the microbiota of carriers and non-carriers of S. aureus and to demonstrate the importance of inter-species relationships in the adhesion of S. aureus, a key step in nasal colonization.Methodology. First, we characterized the nasal microbiota from 30 S. aureus carriers and non-carriers by a culturomic approach. We then evaluated the adhesion of S. aureus, first alone and then along with other bacteria of the nasal microbiota. To do that, we used an in vitro model to measure the interactions among bacteria in the presence of epithelial cells.Results. Analysis of the nasal microbiota of the carriers and non-carriers of S. aureus made it possible to observe that each microbiota has specific features in terms of composition. However, this composition differs significantly between carriers and non-carriers mainly through two bacterial groups: coagulase-negative staphylococci and corynebacteria. In a second part, adhesion of S. aureus to epithelial cells showed competition between S. aureus and these bacteria, suggesting a limitation of nasal colonization by S. aureus.Conclusion. These findings demonstrate the existence of a negative correlation between S. aureus and other species which inhibits adhesion and could limit nasal colonization.
Collapse
Affiliation(s)
- Guillaume Ménard
- Univ Rennes, CHU Rennes, Inserm BRM UMR 1230, F-35000 Rennes, France
| | | | | |
Collapse
|
25
|
Adhesin genes and biofilm formation among pediatric Staphylococcus aureus isolates from implant-associated infections. PLoS One 2020. [PMID: 32569268 DOI: 10.1371/journalpone0235115] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023] Open
Abstract
BACKGROUND Microbial surface component recognizing adhesive matrix molecules (MSCRAMMs) facilitate Staphylococcus aureus adherence to host tissue. We hypothesized that S. aureus isolates from implant-associated infections (IAIs) would differ in MSCRAMM profile and biofilm formation in vitro compared to skin and soft tissue infection (SSTI) isolates. METHODS Pediatric patients and their isolates were identified retrospectively. IAI and SSTI isolates were matched (1:4). Pulsed field gel electrophoresis was performed to group isolates as USA300 vs. non-USA300. Whole genome sequencing was performed and raw sequence data were interrogated for presence of MSCRAMMs (clfA, clfB, cna, ebh, efb, fnbpA, fnbpB, isdA, isdB, sdrC, sdrD, sdrE), biofilm-associated (icaA,D,B,C), and Panton-Valentine leukocidin (lukSF-PV) genes, accessory gene regulator group, and multilocus sequence types. In vitro biofilm formation was assessed for 47 IAI and 47 SSTI isolates using a microtiter plate assay. Conditional logistic regression was performed for analysis of matched data (STATA11, College Station, TX). RESULTS Forty-seven IAI and 188 SSTI isolates were studied. IAI isolates were more often methicillin susceptible S. aureus and non-USA300 vs. SSTI isolates [34 (72%) vs. 79 (42%), p = 0.001 and 38 (81%) vs. 57 (30%) p <0.001, respectively]. Greater than 98% of isolates carried clfA, clfB, efb, isdA, isdB, and icaA,D,B,C while cna was more frequently found among IAI vs. SSTI isolates (p = 0.003). Most isolates were strong biofilm producers. CONCLUSIONS S. aureus IAI isolates were significantly more likely to be MSSA and non-USA300 than SSTI isolates. Carriage of MSCRAMMs and biofilm formation did not differ significantly between isolates. Evaluation of genetic polymorphisms and gene expression profiles are needed to further delineate the role of adhesins in the pathogenesis of IAIs.
Collapse
|
26
|
Foster CE, Kok M, Flores AR, Minard CG, Luna RA, Lamberth LB, Kaplan SL, Hulten KG. Adhesin genes and biofilm formation among pediatric Staphylococcus aureus isolates from implant-associated infections. PLoS One 2020; 15:e0235115. [PMID: 32569268 PMCID: PMC7307771 DOI: 10.1371/journal.pone.0235115] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 06/09/2020] [Indexed: 02/01/2023] Open
Abstract
Background Microbial surface component recognizing adhesive matrix molecules (MSCRAMMs) facilitate Staphylococcus aureus adherence to host tissue. We hypothesized that S. aureus isolates from implant-associated infections (IAIs) would differ in MSCRAMM profile and biofilm formation in vitro compared to skin and soft tissue infection (SSTI) isolates. Methods Pediatric patients and their isolates were identified retrospectively. IAI and SSTI isolates were matched (1:4). Pulsed field gel electrophoresis was performed to group isolates as USA300 vs. non-USA300. Whole genome sequencing was performed and raw sequence data were interrogated for presence of MSCRAMMs (clfA, clfB, cna, ebh, efb, fnbpA, fnbpB, isdA, isdB, sdrC, sdrD, sdrE), biofilm-associated (icaA,D,B,C), and Panton-Valentine leukocidin (lukSF-PV) genes, accessory gene regulator group, and multilocus sequence types. In vitro biofilm formation was assessed for 47 IAI and 47 SSTI isolates using a microtiter plate assay. Conditional logistic regression was performed for analysis of matched data (STATA11, College Station, TX). Results Forty-seven IAI and 188 SSTI isolates were studied. IAI isolates were more often methicillin susceptible S. aureus and non-USA300 vs. SSTI isolates [34 (72%) vs. 79 (42%), p = 0.001 and 38 (81%) vs. 57 (30%) p <0.001, respectively]. Greater than 98% of isolates carried clfA, clfB, efb, isdA, isdB, and icaA,D,B,C while cna was more frequently found among IAI vs. SSTI isolates (p = 0.003). Most isolates were strong biofilm producers. Conclusions S. aureus IAI isolates were significantly more likely to be MSSA and non-USA300 than SSTI isolates. Carriage of MSCRAMMs and biofilm formation did not differ significantly between isolates. Evaluation of genetic polymorphisms and gene expression profiles are needed to further delineate the role of adhesins in the pathogenesis of IAIs.
Collapse
Affiliation(s)
- Catherine E. Foster
- Department of Pediatrics, Section of Infectious Diseases, Baylor College of Medicine and Texas Children’s Hospital, Houston, TX, United States of America
- * E-mail:
| | - Melissa Kok
- Department of Pediatrics, Section of Infectious Diseases, Baylor College of Medicine and Texas Children’s Hospital, Houston, TX, United States of America
| | - Anthony R. Flores
- Division of Infectious Diseases, Department of Pediatrics, Center for Antimicrobial Resistance and Microbial Genomics, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States of America
| | - Charles G. Minard
- Institute for Clinical and Translational Research, Baylor College of Medicine and Texas Children’s Hospital, Houston, TX, United States of America
| | - Ruth A. Luna
- Department of Pediatrics, Section of Pathology and Immunology, Baylor College of Medicine and Texas Children’s Hospital, Houston, TX, United States of America
| | - Linda B. Lamberth
- Department of Pediatrics, Section of Infectious Diseases, Baylor College of Medicine and Texas Children’s Hospital, Houston, TX, United States of America
| | - Sheldon L. Kaplan
- Department of Pediatrics, Section of Infectious Diseases, Baylor College of Medicine and Texas Children’s Hospital, Houston, TX, United States of America
| | - Kristina G. Hulten
- Department of Pediatrics, Section of Infectious Diseases, Baylor College of Medicine and Texas Children’s Hospital, Houston, TX, United States of America
| |
Collapse
|
27
|
Lange J, Heidenreich K, Higelin K, Dyck K, Marx V, Reichel C, van Wamel W, den Reijer M, Görlich D, Kahl BC. Staphylococcus aureus Pathogenicity in Cystic Fibrosis Patients-Results from an Observational Prospective Multicenter Study Concerning Virulence Genes, Phylogeny, and Gene Plasticity. Toxins (Basel) 2020; 12:toxins12050279. [PMID: 32357453 PMCID: PMC7290773 DOI: 10.3390/toxins12050279] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 04/16/2020] [Accepted: 04/21/2020] [Indexed: 12/04/2022] Open
Abstract
Staphylococcus aureus and cystic fibrosis (CF) are closely interlinked. To date, however, the impact of S. aureus culture in CF airways on lung function and disease progression has only been elucidated to a limited degree. This analysis aims to identify bacterial factors associated to clinical deterioration. Data were collected during an observational prospective multi-center study following 195 patients from 17 centers. The average follow-up time was 80 weeks. S. aureus isolates (n = 3180) were scanned for the presence of 25 virulence genes and agr-types using single and multiplex PCR. The presence of specific virulence genes was not associated to clinical deterioration. For the agr-types 1 and 4, however, a link to the subjects’ clinical status became evident. Furthermore, a significant longitudinal decrease in the virulence gene quantity was observed. Analyses of the plasticity of the virulence genes revealed significantly increased plasticity rates in the presence of environmental stress. The results suggest that the phylogenetic background defines S. aureus pathogenicity rather than specific virulence genes. The longitudinal loss of virulence genes most likely reflects the adaptation process directed towards a persistent and colonizing rather than infecting lifestyle.
Collapse
Affiliation(s)
- Jonas Lange
- Institute of Medical Microbiology, University Hospital Münster, 48149 Münster, Germany; (J.L.); (K.H.); (K.H.); (K.D.); (V.M.); (C.R.)
| | - Kathrin Heidenreich
- Institute of Medical Microbiology, University Hospital Münster, 48149 Münster, Germany; (J.L.); (K.H.); (K.H.); (K.D.); (V.M.); (C.R.)
| | - Katharina Higelin
- Institute of Medical Microbiology, University Hospital Münster, 48149 Münster, Germany; (J.L.); (K.H.); (K.H.); (K.D.); (V.M.); (C.R.)
| | - Kristina Dyck
- Institute of Medical Microbiology, University Hospital Münster, 48149 Münster, Germany; (J.L.); (K.H.); (K.H.); (K.D.); (V.M.); (C.R.)
| | - Vanessa Marx
- Institute of Medical Microbiology, University Hospital Münster, 48149 Münster, Germany; (J.L.); (K.H.); (K.H.); (K.D.); (V.M.); (C.R.)
| | - Christian Reichel
- Institute of Medical Microbiology, University Hospital Münster, 48149 Münster, Germany; (J.L.); (K.H.); (K.H.); (K.D.); (V.M.); (C.R.)
| | - Willem van Wamel
- Department of Medical Microbiology and Infectious Diseases, Erasmus Medical Center Rotterdam, 3015 CN Rotterdam, The Netherlands; (W.v.W.); (M.d.R.)
| | - Martijn den Reijer
- Department of Medical Microbiology and Infectious Diseases, Erasmus Medical Center Rotterdam, 3015 CN Rotterdam, The Netherlands; (W.v.W.); (M.d.R.)
| | - Dennis Görlich
- Institute of Biostatistics and Clinical Research, University Hospital Münster, 48149 Münster, Germany;
| | - Barbara C. Kahl
- Institute of Medical Microbiology, University Hospital Münster, 48149 Münster, Germany; (J.L.); (K.H.); (K.H.); (K.D.); (V.M.); (C.R.)
- Correspondence: ; Tel.: +49-251-8355358
| |
Collapse
|
28
|
Parastan R, Kargar M, Solhjoo K, Kafilzadeh F. A synergistic association between adhesion-related genes and multidrug resistance patterns of Staphylococcus aureus isolates from different patients and healthy individuals. J Glob Antimicrob Resist 2020; 22:379-385. [PMID: 32169685 DOI: 10.1016/j.jgar.2020.02.025] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 01/16/2020] [Accepted: 02/26/2020] [Indexed: 11/16/2022] Open
Abstract
OBJECTIVES Biofilm -forming capacity of Staphylococcus aureus (S. aureus) as a commensal opportunistic bacterial species induce a growth in antibiotic resistance in chronic diseases. Since expression of biofilm- related genes and antibiotic resistance function are interdependent, the present study was an attempt to inquire biofilm formation and its relationship with antibiotic resistance in clinical isolates. METHODS 208 S. aureus clinical isolates from four major provinces of Iran were investigated in terms of presence of adhesion genes (icaA, icaD, icaB, icaC, fnbpA, fnbpB, clfA, clfB, cna, sasC, sasG and bap) using PCR. In addition, microtiter plate (Mtp) assay was performed to examine quantitative biofilm formation of the isolates and their antibiotic resistance patterns against 16 antibiotics determined upon CLSI criteria. RESULTS The results revealed high prevalence rate (almost 100%) of icaADBC and MSCRAMMs genes in the isolates. Moreover, bap gene was not detected in any of the tested clinical isolates. Based on phenotypic method 169 isolates (81.25%) were also found to have biofilm formation ability. Among 208 isolates, 98 (47.12%) isolates were multidrug resistant (MDR). Vancomycin, linezolid, nitrofurantoin and quinupristin/dalfopristin were the most effective drugs against MDR strains. Furthermore, the findings demonstrated a significant relationship between MDR and biofilm forming capacity. CONCLUSION Prevalence rate of adhesion- related genes was high in S. aureus from isolates in Iran ;so these genes might be expressed under certain conditions and cause emergence of MDR strains. Therefore, further investigations are necessary to prevent initial attachment based on new candidate adhesion genes for vaccine design.
Collapse
Affiliation(s)
- Raziey Parastan
- Department of Microbiology, Jahrom Branch, Islamic Azad University, Jahrom, Iran; Department of Microbiology, Jahrom University of Medical Sciences, Jahrom, Iran.
| | - Mohammad Kargar
- Department of Microbiology, Jahrom Branch, Islamic Azad University, Jahrom, Iran.
| | - Kavous Solhjoo
- Department of Parasitology, School of Medicine, Jahrom University of Medical Sciences, Jahrom, Iran.
| | - Farshid Kafilzadeh
- Department of Microbiology, Jahrom Branch, Islamic Azad University, Jahrom, Iran.
| |
Collapse
|
29
|
Tørresen OK, Star B, Mier P, Andrade-Navarro MA, Bateman A, Jarnot P, Gruca A, Grynberg M, Kajava AV, Promponas VJ, Anisimova M, Jakobsen KS, Linke D. Tandem repeats lead to sequence assembly errors and impose multi-level challenges for genome and protein databases. Nucleic Acids Res 2019; 47:10994-11006. [PMID: 31584084 PMCID: PMC6868369 DOI: 10.1093/nar/gkz841] [Citation(s) in RCA: 183] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 09/03/2019] [Accepted: 10/01/2019] [Indexed: 12/13/2022] Open
Abstract
The widespread occurrence of repetitive stretches of DNA in genomes of organisms across the tree of life imposes fundamental challenges for sequencing, genome assembly, and automated annotation of genes and proteins. This multi-level problem can lead to errors in genome and protein databases that are often not recognized or acknowledged. As a consequence, end users working with sequences with repetitive regions are faced with 'ready-to-use' deposited data whose trustworthiness is difficult to determine, let alone to quantify. Here, we provide a review of the problems associated with tandem repeat sequences that originate from different stages during the sequencing-assembly-annotation-deposition workflow, and that may proliferate in public database repositories affecting all downstream analyses. As a case study, we provide examples of the Atlantic cod genome, whose sequencing and assembly were hindered by a particularly high prevalence of tandem repeats. We complement this case study with examples from other species, where mis-annotations and sequencing errors have propagated into protein databases. With this review, we aim to raise the awareness level within the community of database users, and alert scientists working in the underlying workflow of database creation that the data they omit or improperly assemble may well contain important biological information valuable to others.
Collapse
Affiliation(s)
- Ole K Tørresen
- Centre for Ecological and Evolutionary Synthesis, Department of Biosciences, University of Oslo, NO-0316 Oslo, Norway
| | - Bastiaan Star
- Centre for Ecological and Evolutionary Synthesis, Department of Biosciences, University of Oslo, NO-0316 Oslo, Norway
| | - Pablo Mier
- Faculty of Biology, Johannes Gutenberg University Mainz, Hans-Dieter-Husch-Weg 15, 55128 Mainz, Germany
| | - Miguel A Andrade-Navarro
- Faculty of Biology, Johannes Gutenberg University Mainz, Hans-Dieter-Husch-Weg 15, 55128 Mainz, Germany
| | - Alex Bateman
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton. CB10 1SD, UK
| | - Patryk Jarnot
- Institute of Informatics, Silesian University of Technology, Akademicka 16, 44-100 Gliwice, Poland
| | - Aleksandra Gruca
- Institute of Informatics, Silesian University of Technology, Akademicka 16, 44-100 Gliwice, Poland
| | - Marcin Grynberg
- Institute of Biochemistry and Biophysics PAS, Pawińskiego 5A, 02-106 Warsaw, Poland
| | - Andrey V Kajava
- Centre de Recherche en Biologie cellulaire de Montpellier, UMR 5237 CNRS, Universite Montpellier 1919 Route de Mende, CEDEX 5, 34293 Montpellier, France
- Institut de Biologie Computationnelle, 34095 Montpellier, France
| | - Vasilis J Promponas
- Bioinformatics Research Laboratory, Department of Biological Sciences, University of Cyprus, PO Box 20537, CY 1678 Nicosia, Cyprus
| | - Maria Anisimova
- Institute of Applied Simulations, School of Life Sciences and Facility Management, Zurich University of Applied Sciences (ZHAW), Wädenswil, Switzerland
- Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | - Kjetill S Jakobsen
- Centre for Ecological and Evolutionary Synthesis, Department of Biosciences, University of Oslo, NO-0316 Oslo, Norway
| | - Dirk Linke
- Section for Genetics and Evolutionary Biology, Department of Biosciences, University of Oslo, NO-0316 Oslo, Norway
| |
Collapse
|
30
|
Ferraro A, Buonocore SM, Auquier P, Nicolas I, Wallemacq H, Boutriau D, van der Most RG. Role and plasticity of Th1 and Th17 responses in immunity to Staphylococcus aureus. Hum Vaccin Immunother 2019; 15:2980-2992. [PMID: 31149870 PMCID: PMC6930085 DOI: 10.1080/21645515.2019.1613126] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The human commensal Staphylococcus aureus (SA) is a leading cause of skin/soft tissue and surgical-site infections, and bacteremia. Functional antibodies and T-cell-mediated immunity, particularly Th1/Th17 responses, are thought to mediate protection. Vaccine development may be hindered by modulation of vaccine-induced T cells by pathogen-activated immunoregulatory responses, e.g., via IL-10.We screened SA proteins for CD4+ T-cell-activating and IL-10/IL-17-inducing capacities using healthy donor-derived PBMCs. Responses were characterized (Th1/Th17/Th22/immunosuppressive IL-10-producing cells) using intracellular cytokine staining and flow cytometry. Phenotypic plasticity of Th1/Th17 cells was evaluated under pro- or anti-inflammatory conditions using modulatory cytokines. The impact of vaccination on SA-specific memory responses was assessed using samples from a clinical trial evaluating AS03-adjuvanted and non-adjuvanted multicomponent (CPS5/CPS8/α-toxin/ClfA) vaccines (NCT01160172).The donors exhibited SA-specific memory T-cell responses, indicative of pre-existing immunity to SA. We identified effective activators of Th1 responses (EbhA/IsaA/SdrE/MntC/Aaa/α-toxin), and Th17 and Th1/Th17 responses (EbhA/IsaA/SdrE and, to a lesser extent, α-toxin), but not of Th22 responses or IL-10 production. MRPII, IsdA, and ClfA were inefficient CD4+ T-cell activators in our assays. IL-10, likely produced by innate immune cells, influenced mainly Th1 cells by suppressing IFN-γ production. The memory CD4+ T-cells observed after long-term stimulation with α-toxin and ClfA indicated that vaccination with these proteins had induced expansion of pre-existing Th1 but not Th17 responses, without apparent adjuvant effect, confirming the trial data. The Th1/Th17-driving proteins (EbhA/IsaA/SdrE) shared low IL-10-promoting abilities and restricted phenotypic plasticity under pro- and anti-inflammatory conditions.Given the complex immunopathology and multiple virulence factors, identification of Th1/Th17-driving antigens, adjuvants and administration routes, and delineation of the role of memory responses, may advance vaccine development.
Collapse
|
31
|
Suphatpahirapol C, Nguyen TH, Tansiri Y, Yingchutrakul Y, Roytrakul S, Nitipan S, Wajjwalku W, Haltrich D, Prapong S, Keawsompong S. Expression of a leptospiral leucine-rich repeat protein using a food-grade vector in Lactobacillus plantarum, as a strategy for vaccine delivery. 3 Biotech 2019; 9:324. [PMID: 31406646 DOI: 10.1007/s13205-019-1856-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 08/01/2019] [Indexed: 01/05/2023] Open
Abstract
In this study, a first food-grade mucosal vaccine against leptospirosis was developed without the use of antibiotic resistance gene. This expression system is based on a food-grade host/vector system of Lactobacillus plantarum and a new vaccine candidate antigen, a leucine-rich repeat (LRR) protein of Leptospira borgpetersenii. The LRR of interest from serovar Sejroe is encoded by two overlapping genes and these genes were fused together by site-directed mutagenesis. The mutant gene thus obtained could be successfully expressed in this system as was shown by western blot analysis and liquid chromatography-mass spectrometry (LC-MS/MS) analysis. In addition, this analysis showed that the mutant LRR protein fused to a homologous signal peptide of L. plantarum could be exported to the cell surface as a result of the native LPXAG motif of the heterologous LRR protein, which presumably is responsible for anchoring the protein to the cell wall of L. plantarum. This new strategy could be an essential tool for further studies of leptospirosis mucosal vaccine delivery.
Collapse
Affiliation(s)
- Chattip Suphatpahirapol
- 1Interdisciplinary Graduate Program in Genetic Engineering, Graduate School, Kasetsart University, Bangkok, Thailand
- 2Department of Physiology, Faculty of Veterinary Medicine, Kasetsart University, Bangkok, Thailand
| | - Thu-Ha Nguyen
- 3Department of Food Sciences and Technology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Yada Tansiri
- 2Department of Physiology, Faculty of Veterinary Medicine, Kasetsart University, Bangkok, Thailand
- 4Center for Advanced Studies for Agriculture and Food (CASAF), Institute for Advanced Studies, Kasetsart University, Bangkok, Thailand
| | - Yodying Yingchutrakul
- 5National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency, Pathum Thani, Thailand
| | - Sittiruk Roytrakul
- 1Interdisciplinary Graduate Program in Genetic Engineering, Graduate School, Kasetsart University, Bangkok, Thailand
- 5National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency, Pathum Thani, Thailand
| | - Supachai Nitipan
- 6Department of Biology, Faculty of Sciences, Thaksin University, Phatthalung Campus, Phatthalung, Thailand
| | - Worawidh Wajjwalku
- 1Interdisciplinary Graduate Program in Genetic Engineering, Graduate School, Kasetsart University, Bangkok, Thailand
| | - Dietmar Haltrich
- 1Interdisciplinary Graduate Program in Genetic Engineering, Graduate School, Kasetsart University, Bangkok, Thailand
- 3Department of Food Sciences and Technology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Siriwan Prapong
- 1Interdisciplinary Graduate Program in Genetic Engineering, Graduate School, Kasetsart University, Bangkok, Thailand
- 2Department of Physiology, Faculty of Veterinary Medicine, Kasetsart University, Bangkok, Thailand
- 4Center for Advanced Studies for Agriculture and Food (CASAF), Institute for Advanced Studies, Kasetsart University, Bangkok, Thailand
| | - Suttipun Keawsompong
- 1Interdisciplinary Graduate Program in Genetic Engineering, Graduate School, Kasetsart University, Bangkok, Thailand
- 4Center for Advanced Studies for Agriculture and Food (CASAF), Institute for Advanced Studies, Kasetsart University, Bangkok, Thailand
- 7Department of Biotechnology, Faculty of Agro-Industry, Kasetsart University, Bangkok, Thailand
| |
Collapse
|
32
|
Foster TJ. Surface Proteins of Staphylococcus aureus. Microbiol Spectr 2019; 7:10.1128/microbiolspec.gpp3-0046-2018. [PMID: 31267926 PMCID: PMC10957221 DOI: 10.1128/microbiolspec.gpp3-0046-2018] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Indexed: 12/20/2022] Open
Abstract
The surface of Staphylococcus aureus is decorated with over 20 proteins that are covalently anchored to peptidoglycan by the action of sortase A. These cell wall-anchored (CWA) proteins can be classified into several structural and functional groups. The largest is the MSCRAMM family, which is characterized by tandemly repeated IgG-like folded domains that bind peptide ligands by the dock lock latch mechanism or the collagen triple helix by the collagen hug. Several CWA proteins comprise modules that have different functions, and some individual domains can bind different ligands, sometimes by different mechanisms. For example, the N-terminus of the fibronectin binding proteins comprises an MSCRAMM domain which binds several ligands, while the C-terminus is composed of tandem fibronectin binding repeats. Surface proteins promote adhesion to host cells and tissue, including components of the extracellular matrix, contribute to biofilm formation by stimulating attachment to the host or indwelling medical devices followed by cell-cell accumulation via homophilic interactions between proteins on neighboring cells, help bacteria evade host innate immune responses, participate in iron acquisition from host hemoglobin, and trigger invasion of bacteria into cells that are not normally phagocytic. The study of genetically manipulated strains using animal infection models has shown that many CWA proteins contribute to pathogenesis. Fragments of CWA proteins have the potential to be used in multicomponent vaccines to prevent S. aureus infections.
Collapse
|
33
|
Abstract
We developed a new approach that couples Southwestern blotting and mass spectrometry to discover proteins that bind extracellular DNA (eDNA) in bacterial biofilms. Using Staphylococcus aureus as a model pathogen, we identified proteins with known DNA-binding activity and uncovered a series of lipoproteins with previously unrecognized DNA-binding activity. We demonstrated that expression of these lipoproteins results in an eDNA-dependent biofilm enhancement. Additionally, we found that while deletion of lipoproteins had a minimal impact on biofilm accumulation, these lipoprotein mutations increased biofilm porosity, suggesting that lipoproteins and their associated interactions contribute to biofilm structure. For one of the lipoproteins, SaeP, we showed that the biofilm phenotype requires the lipoprotein to be anchored to the outside of the cellular membrane, and we further showed that increased SaeP expression correlates with more retention of high-molecular-weight DNA on the bacterial cell surface. SaeP is a known auxiliary protein of the SaeRS system, and we also demonstrated that the levels of SaeP correlate with nuclease production, which can further impact biofilm development. It has been reported that S. aureus biofilms are stabilized by positively charged cytoplasmic proteins that are released into the extracellular environment, where they make favorable electrostatic interactions with the negatively charged cell surface and eDNA. In this work we extend this electrostatic net model to include secreted eDNA-binding proteins and membrane-attached lipoproteins that can function as anchor points between eDNA in the biofilm matrix and the bacterial cell surface.IMPORTANCE Many bacteria are capable of forming biofilms encased in a matrix of self-produced extracellular polymeric substances (EPS) that protects them from chemotherapies and the host defenses. As a result of these inherent resistance mechanisms, bacterial biofilms are extremely difficult to eradicate and are associated with chronic wounds, orthopedic and surgical wound infections, and invasive infections, such as infective endocarditis and osteomyelitis. It is therefore important to understand the nature of the interactions between the bacterial cell surface and EPS that stabilize biofilms. Extracellular DNA (eDNA) has been recognized as an EPS constituent for many bacterial species and has been shown to be important in promoting biofilm formation. Using Staphylococcus aureus biofilms, we show that membrane-attached lipoproteins can interact with the eDNA in the biofilm matrix and promote biofilm formation, which suggests that lipoproteins are potential targets for novel therapies aimed at disrupting bacterial biofilms.
Collapse
|
34
|
Redundant and Distinct Roles of Secreted Protein Eap and Cell Wall-Anchored Protein SasG in Biofilm Formation and Pathogenicity of Staphylococcus aureus. Infect Immun 2019. [PMID: 30670553 DOI: 10.1128/iai00894-18] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023] Open
Abstract
Chronic and fatal infections caused by Staphylococcus aureus are sometimes associated with biofilm formation. Secreted proteins and cell wall-anchored proteins (CWAPs) are important for the development of polysaccharide-independent biofilms, but functional relationships between these proteins are unclear. In the present study, we report the roles of the extracellular adherence protein Eap and the surface CWAP SasG in S. aureus MR23, a clinical methicillin-resistant isolate that forms a robust protein-dependent biofilm and accumulates a large amount of Eap in the extracellular matrix. Double deletion of eap and sasG, but not single eap or sasG deletion, reduced the biomass of the formed biofilm. Mutational analysis demonstrated that cell wall anchorage is essential for the role of SasG in biofilm formation. Confocal laser scanning microscopy revealed that MR23 formed a rugged and thick biofilm; deletion of both eap and sasG reduced biofilm ruggedness and thickness. Although sasG deletion did not affect either of these features, eap deletion reduced the ruggedness but not the thickness of the biofilm. This indicated that Eap contributes to the rough irregular surface structure of the MR23 biofilm and that both Eap and SasG play roles in biofilm thickness. The level of pathogenicity of the Δeap ΔsasG strain in a silkworm larval infection model was significantly lower (P < 0.05) than those of the wild type and single-deletion mutants. Collectively, these findings highlight the redundant and distinct roles of a secreted protein and a CWAP in biofilm formation and pathogenicity of S. aureus and may inform new strategies to control staphylococcal biofilm infections.
Collapse
|
35
|
Redundant and Distinct Roles of Secreted Protein Eap and Cell Wall-Anchored Protein SasG in Biofilm Formation and Pathogenicity of Staphylococcus aureus. Infect Immun 2019; 87:IAI.00894-18. [PMID: 30670553 DOI: 10.1128/iai.00894-18] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 01/12/2019] [Indexed: 01/19/2023] Open
Abstract
Chronic and fatal infections caused by Staphylococcus aureus are sometimes associated with biofilm formation. Secreted proteins and cell wall-anchored proteins (CWAPs) are important for the development of polysaccharide-independent biofilms, but functional relationships between these proteins are unclear. In the present study, we report the roles of the extracellular adherence protein Eap and the surface CWAP SasG in S. aureus MR23, a clinical methicillin-resistant isolate that forms a robust protein-dependent biofilm and accumulates a large amount of Eap in the extracellular matrix. Double deletion of eap and sasG, but not single eap or sasG deletion, reduced the biomass of the formed biofilm. Mutational analysis demonstrated that cell wall anchorage is essential for the role of SasG in biofilm formation. Confocal laser scanning microscopy revealed that MR23 formed a rugged and thick biofilm; deletion of both eap and sasG reduced biofilm ruggedness and thickness. Although sasG deletion did not affect either of these features, eap deletion reduced the ruggedness but not the thickness of the biofilm. This indicated that Eap contributes to the rough irregular surface structure of the MR23 biofilm and that both Eap and SasG play roles in biofilm thickness. The level of pathogenicity of the Δeap ΔsasG strain in a silkworm larval infection model was significantly lower (P < 0.05) than those of the wild type and single-deletion mutants. Collectively, these findings highlight the redundant and distinct roles of a secreted protein and a CWAP in biofilm formation and pathogenicity of S. aureus and may inform new strategies to control staphylococcal biofilm infections.
Collapse
|
36
|
Bonar EA, Bukowski M, Hydzik M, Jankowska U, Kedracka-Krok S, Groborz M, Dubin G, Akkerboom V, Miedzobrodzki J, Sabat AJ, Friedrich AW, Wladyka B. Joint Genomic and Proteomic Analysis Identifies Meta-Trait Characteristics of Virulent and Non-virulent Staphylococcus aureus Strains. Front Cell Infect Microbiol 2018; 8:313. [PMID: 30237986 PMCID: PMC6136393 DOI: 10.3389/fcimb.2018.00313] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 08/16/2018] [Indexed: 12/18/2022] Open
Abstract
Staphylococcus aureus is an opportunistic pathogen of humans and warm-blooded animals and presents a growing threat in terms of multi-drug resistance. Despite numerous studies, the basis of staphylococcal virulence and switching between commensal and pathogenic phenotypes is not fully understood. Using genomics, we show here that S. aureus strains exhibiting virulent (VIR) and non-virulent (NVIR) phenotypes in a chicken embryo infection model genetically fall into two separate groups, with the VIR group being much more cohesive than the NVIR group. Significantly, the genes encoding known staphylococcal virulence factors, such as clumping factors, are either found in different allelic variants in the genomes of NVIR strains (compared to VIR strains) or are inactive pseudogenes. Moreover, the pyruvate carboxylase and gamma-aminobutyrate permease genes, which were previously linked with virulence, are pseudogenized in NVIR strain ch22. Further, we use comprehensive proteomics tools to characterize strains that show opposing phenotypes in a chicken embryo virulence model. VIR strain CH21 had an elevated level of diapolycopene oxygenase involved in staphyloxanthin production (protection against free radicals) and expressed a higher level of immunoglobulin-binding protein Sbi on its surface compared to NVIR strain ch22. Furthermore, joint genomic and proteomic approaches linked the elevated production of superoxide dismutase and DNA-binding protein by NVIR strain ch22 with gene duplications.
Collapse
Affiliation(s)
- Emilia A Bonar
- Department of Analytical Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Michal Bukowski
- Department of Analytical Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Marcin Hydzik
- Department of Analytical Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Urszula Jankowska
- Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
| | - Sylwia Kedracka-Krok
- Department of Physical Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Magdalena Groborz
- Department of Analytical Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Grzegorz Dubin
- Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland.,Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Viktoria Akkerboom
- Department of Medical Microbiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Jacek Miedzobrodzki
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Artur J Sabat
- Department of Medical Microbiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Alexander W Friedrich
- Department of Medical Microbiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Benedykt Wladyka
- Department of Analytical Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| |
Collapse
|
37
|
Frisch MB, Castillo-Ramírez S, Petit RA, Farley MM, Ray SM, Albrecht VS, Limbago BM, Hernandez J, See I, Satola SW, Read TD. Invasive Methicillin-Resistant Staphylococcus aureus USA500 Strains from the U.S. Emerging Infections Program Constitute Three Geographically Distinct Lineages. mSphere 2018; 3:e00571-17. [PMID: 29720528 PMCID: PMC5932375 DOI: 10.1128/msphere.00571-17] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 04/09/2018] [Indexed: 01/09/2023] Open
Abstract
USA500 isolates are clonal complex 8 (CC8) Staphylococcus aureus strains closely related to the prominent community- and hospital-associated USA300 group. Despite being relatively understudied, USA500 strains cause a significant burden of disease and are the third most common methicillin-resistant S. aureus (MRSA) strains identified in the U.S. Emerging Infections Program (EIP) invasive S. aureus surveillance. To better understand the genetic relationships of the strains, we sequenced the genomes of 539 USA500 MRSA isolates from sterile site infections collected through the EIP between 2005 and 2013 in the United States. USA500 isolates fell into three major clades principally separated by their distribution across different U.S. regions. Clade C1 strains, found principally in the Northeast, were associated with multiple IS256 insertion elements in their genomes and higher levels of antibiotic resistance. C2 was associated with Southern states, and E1 was associated with Western states. C1 and C2 strains all shared a frameshift in the gene encoding AdsA surface-attached surface protein. We propose that the term "USA500" should be used for CC8 strains sharing a recent common ancestor with the C1, C2, and E1 strains but not in the USA300 group.IMPORTANCE In this work, we have removed some of the confusion surrounding the use of the name "USA500," placed USA500 strains in the context of the CC8 group, and developed a strategy for assignment to subclades based on genome sequence. Our new phylogeny of USA300/USA500 will be a reference point for understanding the genetic adaptations that have allowed multiple highly virulent clonal strains to emerge from within CC8 over the past 50 years.
Collapse
Affiliation(s)
- M B Frisch
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | | | - R A Petit
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - M M Farley
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
- Atlanta VA Medical Center, Decatur, Georgia, USA
| | - S M Ray
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - V S Albrecht
- Division of Healthcare Quality Promotion, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - B M Limbago
- Division of Healthcare Quality Promotion, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - J Hernandez
- Division of Healthcare Quality Promotion, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - I See
- Division of Healthcare Quality Promotion, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - S W Satola
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - T D Read
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
38
|
Belyi Y, Rybolovlev I, Polyakov N, Chernikova A, Tabakova I, Gintsburg A. Staphylococcus Aureus Surface Protein G is An Immunodominant Protein and a Possible Target in An Anti-Biofilm Drug Development. Open Microbiol J 2018; 12:94-106. [PMID: 29785216 PMCID: PMC5944129 DOI: 10.2174/1874285801812010094] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 04/08/2018] [Accepted: 04/11/2018] [Indexed: 12/18/2022] Open
Abstract
Background Staphylococcus aureus is a Gram-positive bacterium that causes severe illnesses in the human population. The capacity of S. aureus strains to form biofilms on biotic and abiotic surfaces creates serious problems for treatment of hospital infections and has stimulated efforts to develop new means of specific protection or immunotherapy. Material and Methods We found that rabbit serum raised against crude concentrated S. aureus liquid culture significantly decreased the development of staphylococcal biofilm in vitro. To discover the corresponding staphylococcal antigen, we used mass-spectrometry and molecular cloning and identified three major immunodominant proteins. They included α-haemolysin, serine proteinase SplB and S. aureus surface protein G, known as adhesin SasG. Results Although according to literature data, all these proteins represent virulence factors of S. aureus and play diverse and important roles in the pathogenesis of staphylococcal diseases, only SasG can be directly implicated into the biofilm formation because of its surface location on a staphylococcal cell. Indeed, rabbit serum directed against purified recombinant SasG, similar to serum against crude staphylococcal liquid culture, prevented the formation of a biofilm. Conclusion SasG can be considered as a target in an anti-biofilm drug development and a component of the vaccine or immunotherapeutic preparations directed against staphylococcal infections in humans.
Collapse
Affiliation(s)
- Yury Belyi
- Gamaleya Research Centre for Epidemiology and Microbiology, Moscow, Russia
| | - Ivan Rybolovlev
- Gamaleya Research Centre for Epidemiology and Microbiology, Moscow, Russia
| | - Nikita Polyakov
- Gamaleya Research Centre for Epidemiology and Microbiology, Moscow, Russia.,Vernadsky Institute of Geochemistry and Analytical Chemistry, Moscow, Russia
| | | | - Irina Tabakova
- Gamaleya Research Centre for Epidemiology and Microbiology, Moscow, Russia
| | | |
Collapse
|
39
|
Methicillin-Resistant Staphylococcus aureus Sequence Type (ST) 5 Isolates from Health Care and Agricultural Sources Adhere Equivalently to Human Keratinocytes. Appl Environ Microbiol 2018; 84:AEM.02073-17. [PMID: 29101193 DOI: 10.1128/aem.02073-17] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 10/28/2017] [Indexed: 11/20/2022] Open
Abstract
Staphylococcus aureus is part of the nasal microbiome of many humans and has become a significant public health burden due to infections with antibiotic-resistant strains, including methicillin-resistant S. aureus (MRSA) strains. Several lineages of S. aureus, including MRSA, are found in livestock species and can be acquired by humans through contact with animals. These livestock-associated MRSA (LA-MRSA) isolates raise public health concerns because of the potential for livestock to act as reservoirs for MRSA outside the hospital setting. In the United States, swine harbor a mixed population of LA-MRSA isolates, with the sequence type 398 (ST398), ST9, and ST5 lineages being detected. LA-MRSA ST5 isolates are particularly concerning to the public health community because, unlike the isolates in the ST398 and ST9 lineages, isolates in the ST5 lineage are a significant cause of human disease in both the hospital and community settings globally. The ability of swine-associated LA-MRSA ST5 isolates to adhere to human keratinocytes in vitro was investigated, and the adherence genes harbored by these isolates were evaluated and compared to those in clinical MRSA ST5 isolates from humans with no swine contact. The two subsets of isolates adhered equivalently to human keratinocytes in vitro and contained an indistinguishable complement of adherence genes that possessed a high degree of sequence identity. Collectively, our data indicate that, unlike LA-MRSA ST398 isolates, LA-MRSA ST5 isolates do not exhibit a reduced genotypic or phenotypic capacity to adhere to human keratinocytes.IMPORTANCE Our data indicate that swine-associated livestock-associated methicillin-resistant Staphylococcus aureus (LA-MRSA) ST5 isolates are as capable of adhering to human skin and have the same genetic potential to adhere as clinical MRSA ST5 isolates from humans. This suggests that humans in contact with livestock have the potential to become colonized with LA-MRSA ST5 isolates; however, the genes that contribute to the persistence of S. aureus on human skin were absent in LA-MRSA ST5 isolates. The data presented here are important evidence in evaluating the potential risks that LA-MRSA ST5 isolates pose to humans who come into contact with livestock.
Collapse
|
40
|
Surface proteins and the formation of biofilms by Staphylococcus aureus. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1860:749-756. [PMID: 29229527 DOI: 10.1016/j.bbamem.2017.12.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 12/01/2017] [Accepted: 12/04/2017] [Indexed: 11/21/2022]
Abstract
Staphylococcus aureus biofilms pose a serious clinical threat as reservoirs for persistent infections. Despite this clinical significance, the composition and mechanism of formation of S. aureus biofilms are unknown. To address these problems, we used solid-state NMR to examine S. aureus (SA113), a strong biofilm-forming strain. We labeled whole cells and cell walls of planktonic cells, young biofilms formed for 12-24h after stationary phase, and more mature biofilms formed for up to 60h after stationary phase. All samples were labeled either by (i) [15N]glycine and l-[1-13C]threonine, or in separate experiments, by (ii) l-[2-13C,15N]leucine. We then measured 13C-15N direct bonds by C{N} rotational-echo double resonance (REDOR). The increase in peptidoglycan stems that have bridges connected to a surface protein was determined directly by a cell-wall double difference (biofilm REDOR difference minus planktonic REDOR difference). This procedure eliminates errors arising from differences in 15N isotopic enrichments and from the routing of 13C label from threonine degradation to glycine. For both planktonic cells and the mature biofilm, 20% of pentaglycyl bridges are not cross-linked and are potential surface-protein attachment sites. None of these sites has a surface protein attached in the planktonic cells, but one-fourth have a surface protein attached in the mature biofilm. Moreover, the leucine-label shows that the concentration of β-strands in leucine-rich regions doubles in the mature biofilm. Thus, a primary event in establishing a S. aureus biofilm is extensive decoration of the cell surface with surface proteins that are linked covalently to the cell wall and promote cell-cell adhesion.
Collapse
|
41
|
Proteomics of Staphylococcus aureus biofilm matrix in a rat model of orthopedic implant-associated infection. PLoS One 2017; 12:e0187981. [PMID: 29121106 PMCID: PMC5679556 DOI: 10.1371/journal.pone.0187981] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 10/30/2017] [Indexed: 01/09/2023] Open
Abstract
The matrix proteins of Staphylococcus aureus biofilm have not been well defined. Previous efforts to identify these proteins were performed using in vitro systems. Here we use a proteomic approach to identify biofilm matrix proteins directly from infected bone implants using a rat model of orthopedic implant-associated S. aureus infection. Despite heavy presence of host proteins, a total of 28 and 105 S. aureus proteins were identified during acute infection and chronic infection, respectively. Our results show that biofilm matrix contains mostly intracellular cytoplasmic proteins and, to a much less extent, extracellular and cell surface-associated proteins. Significantly, leukocidins were identified in the biofilm matrix during chronic infection, suggesting S. aureus is actively attacking the host immune system even though they are protected within the biofilm. The presence of two surface-associated proteins, Ebh and SasF, in the infected bone tissue during acute infection was confirmed by immunohistochemistry. In addition, a large number of host proteins were found differentially expressed in response to S. aureus biofilm formed on bone implants.
Collapse
|
42
|
Catheterization alters bladder ecology to potentiate Staphylococcus aureus infection of the urinary tract. Proc Natl Acad Sci U S A 2017; 114:E8721-E8730. [PMID: 28973850 DOI: 10.1073/pnas.1707572114] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is an emerging cause of catheter-associated urinary tract infection (CAUTI), which frequently progresses to more serious invasive infections. We adapted a mouse model of CAUTI to investigate how catheterization increases an individual's susceptibility to MRSA UTI. This analysis revealed that catheterization was required for MRSA to achieve high-level, persistent infection in the bladder. As shown previously, catheter placement induced an inflammatory response resulting in the release of the host protein fibrinogen (Fg), which coated the bladder and implant. Following infection, we showed that MRSA attached to the urothelium and implant in patterns that colocalized with deposited Fg. Furthermore, MRSA exacerbated the host inflammatory response to stimulate the additional release and accumulation of Fg in the urinary tract, which facilitated MRSA colonization. Consistent with this model, analysis of catheters from patients with S. aureus-positive cultures revealed colocalization of Fg, which was deposited on the catheter, with S. aureus Clumping Factors A and B (ClfA and ClfB) have been shown to contribute to MRSA-Fg interactions in other models of disease. We found that mutants in clfA had significantly greater Fg-binding defects than mutants in clfB in several in vitro assays. Paradoxically, only the ClfB- strain was significantly attenuated in the CAUTI model. Together, these data suggest that catheterization alters the urinary tract environment to promote MRSA CAUTI pathogenesis by inducing the release of Fg, which the pathogen enhances to persist in the urinary tract despite the host's robust immune response.
Collapse
|
43
|
Zhang B, Teng Z, Li X, Lu G, Deng X, Niu X, Wang J. Chalcone Attenuates Staphylococcus aureus Virulence by Targeting Sortase A and Alpha-Hemolysin. Front Microbiol 2017; 8:1715. [PMID: 28932220 PMCID: PMC5592744 DOI: 10.3389/fmicb.2017.01715] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 08/24/2017] [Indexed: 12/21/2022] Open
Abstract
Staphylococcus aureus (S.aureus) resistance, considered a dilemma for the clinical treatment of this bacterial infection, is becoming increasingly intractable. Novel anti-virulence strategies will undoubtedly provide a path forward in combating these resistant bacterial infections. Sortase A (SrtA), an enzyme responsible for anchoring virulence-related surface proteins, and alpha-hemolysin (Hla), a pore-forming cytotoxin, have aroused great scientific interest, as they have been regarded as targets for promising agents against S. aureus infection. In this study, we discovered that chalcone, a natural small compound with little anti-S. aureus activity, could significantly inhibit SrtA activity with an IC50 of 53.15 μM and Hla hemolysis activity with an IC50 of 17.63 μM using a fluorescence resonance energy transfer (FRET) assay and a hemolysis assay, respectively. In addition, chalcone was proven to reduce protein A (SpA) display in intact bacteria, binding to fibronectin, formation of biofilm and S. aureus invasion. Chalcone could down-regulate the transcriptional levels of the hla gene and the agrA gene, thus leading to a reduction in the expression of Hla and significant protection against Hla-mediated A549 cell injury; more importantly, chalcone could also reduce mortality in infected mice. Additionally, molecular dynamics simulations and mutagenesis assays were used to identify the mechanism of chalcone against SrtA, which implied that the inhibitory activity lies in the bond between chalcone and SrtA residues Val168, Ile182, and Arg197. Taken together, the in vivo and in vitro experiments suggest that chalcone is a potential novel therapeutic compound for S. aureus infection via targeting SrtA and Hla.
Collapse
Affiliation(s)
- Bing Zhang
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin UniversityChangchun, China.,Center of Infection and Immunity, The First Hospital, Jilin UniversityChangchun, China
| | - Zihao Teng
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin UniversityChangchun, China
| | - Xianhe Li
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin UniversityChangchun, China
| | - Gejin Lu
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin UniversityChangchun, China
| | - Xuming Deng
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin UniversityChangchun, China.,Center of Infection and Immunity, The First Hospital, Jilin UniversityChangchun, China
| | - Xiaodi Niu
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin UniversityChangchun, China
| | - Jianfeng Wang
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin UniversityChangchun, China.,Center of Infection and Immunity, The First Hospital, Jilin UniversityChangchun, China
| |
Collapse
|
44
|
Balachandran M, Giannone RJ, Bemis DA, Kania SA. Molecular basis of surface anchored protein A deficiency in the Staphylococcus aureus strain Wood 46. PLoS One 2017; 12:e0183913. [PMID: 28859130 PMCID: PMC5578664 DOI: 10.1371/journal.pone.0183913] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 08/14/2017] [Indexed: 11/18/2022] Open
Abstract
Protein A in Staphylococcus aureus is encoded by the spa (staphylococcal protein A) gene and binds to immunoglobulin (Ig). The S. aureus strain Wood 46 has been variously reported as protein A-deficient and/or spa negative and used as a control in animal models of staphylococcal infections. The results of this study indicate that Wood 46 has normal spa expression but transcribes very low levels of the srtA gene which encodes the sortase A (SrtA) enzyme. This is consistent with unique mutations in the srtA promoter. In this study, a low level of sortase A explains deficient anchoring of proteins with an LPXTG motif, such as protein A, fibrinogen-binding protein and fibronectin-binding proteins A and B on to the peptidoglycan cell wall. The activity of secreted protein A is an important consideration for use of Wood 46 in functional experiments and animal models.
Collapse
Affiliation(s)
- Manasi Balachandran
- Department of Biomedical and Diagnostic Sciences, The University of Tennessee, Knoxville, Tennessee, United States of America
| | - Richard J. Giannone
- Chemical Sciences Division, Mass Spectrometry and Laser Spectrometry, Oakridge National Laboratories, Oakridge, Tennessee, United States of America
| | - David A. Bemis
- Department of Biomedical and Diagnostic Sciences, The University of Tennessee, Knoxville, Tennessee, United States of America
| | - Stephen A. Kania
- Department of Biomedical and Diagnostic Sciences, The University of Tennessee, Knoxville, Tennessee, United States of America
- * E-mail:
| |
Collapse
|
45
|
Abstract
The staphylococci comprise a diverse genus of Gram-positive, nonmotile commensal organisms that inhabit the skin and mucous membranes of humans and other mammals. In general, staphylococci are benign members of the natural flora, but many species have the capacity to be opportunistic pathogens, mainly infecting individuals who have medical device implants or are otherwise immunocompromised. Staphylococcus aureus and Staphylococcus epidermidis are major sources of hospital-acquired infections and are the most common causes of surgical site infections and medical device-associated bloodstream infections. The ability of staphylococci to form biofilms in vivo makes them highly resistant to chemotherapeutics and leads to chronic diseases. These biofilm infections include osteomyelitis, endocarditis, medical device infections, and persistence in the cystic fibrosis lung. Here, we provide a comprehensive analysis of our current understanding of staphylococcal biofilm formation, with an emphasis on adhesins and regulation, while also addressing how staphylococcal biofilms interact with the immune system. On the whole, this review will provide a thorough picture of biofilm formation of the staphylococcus genus and how this mode of growth impacts the host.
Collapse
|
46
|
Claes J, Liesenborghs L, Peetermans M, Veloso TR, Missiakas D, Schneewind O, Mancini S, Entenza JM, Hoylaerts MF, Heying R, Verhamme P, Vanassche T. Clumping factor A, von Willebrand factor-binding protein and von Willebrand factor anchor Staphylococcus aureus to the vessel wall. J Thromb Haemost 2017; 15:1009-1019. [PMID: 28182324 PMCID: PMC6232194 DOI: 10.1111/jth.13653] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Indexed: 01/10/2023]
Abstract
Essentials Staphylococcus aureus (S. aureus) binds to endothelium via von Willebrand factor (VWF). Secreted VWF-binding protein (vWbp) mediates S. aureus adhesion to VWF under shear stress. vWbp interacts with VWF and the Sortase A-dependent surface protein Clumping factor A (ClfA). VWF-vWbp-ClfA anchor S. aureus to vascular endothelium under shear stress. SUMMARY Objective When establishing endovascular infections, Staphylococcus aureus (S. aureus) overcomes shear forces of flowing blood by binding to von Willebrand factor (VWF). Staphylococcal VWF-binding protein (vWbp) interacts with VWF, but it is unknown how this secreted protein binds to the bacterial cell wall. We hypothesized that vWbp interacts with a staphylococcal surface protein, mediating the adhesion of S. aureus to VWF and vascular endothelium under shear stress. Methods We studied the binding of S. aureus to vWbp, VWF and endothelial cells in a micro-parallel flow chamber using various mutants deficient in Sortase A (SrtA) and SrtA-dependent surface proteins, and Lactococcus lactis expressing single staphylococcal surface proteins. In vivo adhesion of bacteria was evaluated in the murine mesenteric circulation using real-time intravital vascular microscopy. Results vWbp bridges the bacterial cell wall and VWF, allowing shear-resistant binding of S. aureus to inflamed or damaged endothelium. Absence of SrtA and Clumping factor A (ClfA) reduced adhesion of S. aureus to vWbp, VWF and activated endothelial cells. ADAMTS-13 and an anti-VWF A1 domain antibody, when combined, reduced S. aureus adhesion to activated endothelial cells by 90%. Selective overexpression of ClfA in the membrane of Lactococcus lactis enabled these bacteria to bind to VWF and activated endothelial cells but only in the presence of vWbp. Absence of ClfA abolished bacterial adhesion to the activated murine vessel wall. Conclusions vWbp interacts with VWF and with the SrtA-dependent staphylococcal surface protein ClfA. The complex formed by VWF, secreted vWbp and bacterial ClfA anchors S. aureus to vascular endothelium under shear stress.
Collapse
Affiliation(s)
- J Claes
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
- Cardiovascular Developmental Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - L Liesenborghs
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - M Peetermans
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - T R Veloso
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
- Cardiovascular Developmental Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - D Missiakas
- Department of Microbiology, University of Chicago, Chicago, IL, USA
| | - O Schneewind
- Department of Microbiology, University of Chicago, Chicago, IL, USA
| | - S Mancini
- Department of Fundamental Microbiology, University of Lausanne, Lausanne, Switzerland
| | - J M Entenza
- Department of Fundamental Microbiology, University of Lausanne, Lausanne, Switzerland
| | - M F Hoylaerts
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - R Heying
- Cardiovascular Developmental Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - P Verhamme
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - T Vanassche
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| |
Collapse
|
47
|
Yang Y, Yu R, Yang X, Liu S, Fang T, Song X, Hou L, Yu C, Xu J, Fu L, Yi S, Chen W. Protection against Staphylococcus aureus and tetanus infections by a combined vaccine containing SasA and TeNT‑Hc in mice. Mol Med Rep 2017; 15:2369-2373. [PMID: 28259925 DOI: 10.3892/mmr.2017.6227] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 12/19/2016] [Indexed: 11/06/2022] Open
Abstract
In developing countries, trauma patients and neonates are vulnerable to Staphylococcus aureus (S. aureus) and Clostridium tetani infections. It has been suggested that a combined vaccine against the two infections may be a reliable and cost‑effective strategy. Previous studies have indicated that the S. aureus surface protein A (SasA) and the C fragment of tetanus neurotoxin (TeNT‑Hc) may be suitable candidates for a vaccine against S. aureus and tetanus infections, respectively. In the present study, mice were immunized with a combined vaccine containing SasA and TeNT‑Hc, which induced a robust immune response to both antigens, and mutual interference between SasA and TeNT‑Hc was not observed. In the S.aureus challenge model, the combined vaccine fully protected BALB/c mice against lethal intraperitoneal challenges with 3x109 colony‑forming units of a methicillin‑resistant S. aureus USA300 strain. In the TeNT challenge model, the combined vaccine conferred complete protection against a lethal dose of (2x103) xLD50 tetanus toxin. These results implied that SasA and TeNT‑Hc promising components for a combined vaccine against S. aureus and tetanus infections.
Collapse
Affiliation(s)
- Yilong Yang
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Fengtai, Beijing 100071, P.R. China
| | - Rui Yu
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Fengtai, Beijing 100071, P.R. China
| | - Xiuxu Yang
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Fengtai, Beijing 100071, P.R. China
| | - Shuling Liu
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Fengtai, Beijing 100071, P.R. China
| | - Ting Fang
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Fengtai, Beijing 100071, P.R. China
| | - Xiaohong Song
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Fengtai, Beijing 100071, P.R. China
| | - Lihua Hou
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Fengtai, Beijing 100071, P.R. China
| | - Changming Yu
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Fengtai, Beijing 100071, P.R. China
| | - Junjie Xu
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Fengtai, Beijing 100071, P.R. China
| | - Ling Fu
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Fengtai, Beijing 100071, P.R. China
| | - Shaoqiong Yi
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Fengtai, Beijing 100071, P.R. China
| | - Wei Chen
- Laboratory of Vaccine and Antibody Engineering, Beijing Institute of Biotechnology, Fengtai, Beijing 100071, P.R. China
| |
Collapse
|
48
|
Bleiziffer I, Eikmeier J, Pohlentz G, McAulay K, Xia G, Hussain M, Peschel A, Foster S, Peters G, Heilmann C. The Plasmin-Sensitive Protein Pls in Methicillin-Resistant Staphylococcus aureus (MRSA) Is a Glycoprotein. PLoS Pathog 2017; 13:e1006110. [PMID: 28081265 PMCID: PMC5230774 DOI: 10.1371/journal.ppat.1006110] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 12/02/2016] [Indexed: 01/16/2023] Open
Abstract
Most bacterial glycoproteins identified to date are virulence factors of pathogenic bacteria, i.e. adhesins and invasins. However, the impact of protein glycosylation on the major human pathogen Staphylococcus aureus remains incompletely understood. To study protein glycosylation in staphylococci, we analyzed lysostaphin lysates of methicillin-resistant Staphylococcus aureus (MRSA) strains by SDS-PAGE and subsequent periodic acid-Schiff’s staining. We detected four (>300, ∼250, ∼165, and ∼120 kDa) and two (>300 and ∼175 kDa) glycosylated surface proteins with strain COL and strain 1061, respectively. The ∼250, ∼165, and ∼175 kDa proteins were identified as plasmin-sensitive protein (Pls) by mass spectrometry. Previously, Pls has been demonstrated to be a virulence factor in a mouse septic arthritis model. The pls gene is encoded by the staphylococcal cassette chromosome (SCC)mec type I in MRSA that also encodes the methicillin resistance-conferring mecA and further genes. In a search for glycosyltransferases, we identified two open reading frames encoded downstream of pls on the SCCmec element, which we termed gtfC and gtfD. Expression and deletion analysis revealed that both gtfC and gtfD mediate glycosylation of Pls. Additionally, the recently reported glycosyltransferases SdgA and SdgB are involved in Pls glycosylation. Glycosylation occurs at serine residues in the Pls SD-repeat region and modifying carbohydrates are N-acetylhexosaminyl residues. Functional characterization revealed that Pls can confer increased biofilm formation, which seems to involve two distinct mechanisms. The first mechanism depends on glycosylation of the SD-repeat region by GtfC/GtfD and probably also involves eDNA, while the second seems to be independent of glycosylation as well as eDNA and may involve the centrally located G5 domains. Other previously known Pls properties are not related to the sugar modifications. In conclusion, Pls is a glycoprotein and Pls glycosyl residues can stimulate biofilm formation. Thus, sugar modifications may represent promising new targets for novel therapeutic or prophylactic measures against life-threatening S. aureus infections. Staphylococcus aureus is a serious pathogen that causes life-threatening infections due to its ability to attach to surfaces, form biofilms, and persist inside the host. One of previously identified virulence factors in S. aureus pathogenesis is the plasmin-sensitive surface protein Pls. We here identified Pls as a posttranslationally modified glycoprotein and characterized the domain within Pls that becomes glycosylated as well as the modifying sugars. Moreover, we found that the glycosyltransferases GtfC and GtfD carry out the glycosylation reactions. In a search for a role for the modifying sugars, we found that Pls can stimulate biofilm formation apparently via two distinct mechanisms, one being dependent on glycosylation by GtfC and GtfD the other being independent of glycosylation as well as eDNA. Moreover, we found that none of the already known Pls functions is mediated by the sugar moieties. Thus, we conclude that GtfC/GtfD-glycosylated Pls may contribute to MRSA pathogenicity via stimulation of biofilm formation and may serve as future target to combat or prevent infections with this serious pathogen.
Collapse
Affiliation(s)
- Isabelle Bleiziffer
- Institute of Medical Microbiology, University of Münster, Münster, Germany
- Interdisciplinary Center for Clinical Research (IZKF), University of Münster, Münster, Germany
| | - Julian Eikmeier
- Institute of Medical Microbiology, University of Münster, Münster, Germany
- Interdisciplinary Center for Clinical Research (IZKF), University of Münster, Münster, Germany
| | | | - Kathryn McAulay
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, United Kingdom
| | - Guoqing Xia
- Division of Infection, Immunity and Respiratory Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Muzaffar Hussain
- Institute of Medical Microbiology, University of Münster, Münster, Germany
| | - Andreas Peschel
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany
- German Center for Infection Research (DZIF), partner site Tübingen, University of Tübingen, Tübingen, Germany
| | - Simon Foster
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, United Kingdom
| | - Georg Peters
- Institute of Medical Microbiology, University of Münster, Münster, Germany
- Interdisciplinary Center for Clinical Research (IZKF), University of Münster, Münster, Germany
- Cluster of Excellence EXC 1003, Cells in Motion, University of Münster, Münster, Germany
| | - Christine Heilmann
- Institute of Medical Microbiology, University of Münster, Münster, Germany
- Interdisciplinary Center for Clinical Research (IZKF), University of Münster, Münster, Germany
- * E-mail:
| |
Collapse
|
49
|
Foster TJ. The remarkably multifunctional fibronectin binding proteins of Staphylococcus aureus. Eur J Clin Microbiol Infect Dis 2016; 35:1923-1931. [PMID: 27604831 DOI: 10.1007/s10096-016-2763-0] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 08/16/2016] [Indexed: 12/01/2022]
Abstract
Staphylococcus aureus expresses two distinct but closely related multifunctional cell wall-anchored (CWA) proteins that bind to the host glycoprotein fibronectin. The fibronectin binding proteins FnBPA and FnBPB comprise two distinct domains. The C-terminal domain comprises a tandem array of repeats that bind to the N-terminal type I modules of fibronectin by the tandem β-zipper mechanism. This causes allosteric activation of a cryptic integrin binding domain, allowing fibronectin to act as a bridge between bacterial cells and the α5β1 integrin on host cells, triggering bacterial uptake by endocytosis. Variants of FnBPA with polymorphisms in fibronectin binding repeats (FnBRs) that increase affinity for the ligand are associated with strains that infect cardiac devices and cause endocarditis, suggesting that binding affinity is particularly important in intravascular infections. The N-terminal A domains of FnBPA and FnBPB have diverged into seven antigenically distinct isoforms. Each binds fibrinogen by the 'dock, lock and latch' mechanism characteristic of clumping factor A. However, FnBPs can also bind to elastin, which is probably important in adhesion to connective tissue in vivo. In addition, they can capture plasminogen from plasma, which can be activated to plasmin by host and bacterial plasminogen activators. The bacterial cells become armed with a host protease which destroys opsonins, contributing to immune evasion and promotes spreading during skin infection. Finally, some methicillin-resistant S. aureus (MRSA) strains form biofilm that depends on the elaboration of FnBPs rather than polysaccharide. The A domains of the FnBPs can interact homophilically, allowing cells to bind together as the biofilm accumulates.
Collapse
Affiliation(s)
- T J Foster
- Microbiology Department, Trinity College, Dublin 2, Ireland.
| |
Collapse
|
50
|
Versatility of Biofilm Matrix Molecules in Staphylococcus epidermidis Clinical Isolates and Importance of Polysaccharide Intercellular Adhesin Expression during High Shear Stress. mSphere 2016; 1:mSphere00165-16. [PMID: 27747298 PMCID: PMC5064449 DOI: 10.1128/msphere.00165-16] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 09/20/2016] [Indexed: 01/13/2023] Open
Abstract
Staphylococcus epidermidis is a leading cause of infections related to biomaterials, mostly due to their ability to form biofilm. Biofilm accumulation mechanisms vary, including those that are dependent on specific proteins, environmental DNA (eDNA), or polysaccharide intercellular adhesin (PIA). We found that those isolates obtained from high-shear environments, such as the lumen of a catheter, are more likely to produce PIA-mediated biofilms than those isolates obtained from a low-shear biomaterial-related infection. This suggests that PIA functions as a mechanism that is protective against shear flow. Finally, we performed selection experiments documenting the heterogeneity of biofilm accumulation molecules that function in the absence of PIA, further documenting the biofilm-forming potential of S. epidermidis. Staphylococcus epidermidis is a leading cause of hospital-associated infections, including those of intravascular catheters, cerebrospinal fluid shunts, and orthopedic implants. Multiple biofilm matrix molecules with heterogeneous characteristics have been identified, including proteinaceous, polysaccharide, and nucleic acid factors. Two of the best-studied components in S. epidermidis include accumulation-associated protein (Aap) and polysaccharide intercellular adhesin (PIA), produced by the enzymatic products of the icaADBC operon. Biofilm composition varies by strain as well as environmental conditions, and strains producing PIA-mediated biofilms are more robust. Clinically, biofilm-mediated infections occur in a variety of anatomical sites with diverse physiological properties. To test the hypothesis that matrix composition exhibits niche specificity, biofilm-related genetic and physical properties were compared between S. epidermidis strains isolated from high-shear and low-shear environments. Among a collection of 105 clinical strains, significantly more isolates from high-shear environments carried the icaADBC operon than did those from low-shear settings (43.9% versus 22.9%, P < 0.05), while there was no significant difference in the presence of aap (77.2% versus 75.0%, P > 0.05). Additionally, a significantly greater number of high-shear isolates were capable of forming biofilm in vitro in a microtiter assay (82.5% versus 45.8%, P < 0.0001). However, even among high-shear clinical isolates, less than half contained the icaADBC locus; therefore, we selected for ica-negative variants with increased attachment to abiotic surfaces to examine PIA-independent biofilm mechanisms. Sequencing of selected variants identified substitutions capable of enhancing biofilm formation in multiple genes, further highlighting the heterogeneity of S. epidermidis biofilm molecules and mechanisms. IMPORTANCEStaphylococcus epidermidis is a leading cause of infections related to biomaterials, mostly due to their ability to form biofilm. Biofilm accumulation mechanisms vary, including those that are dependent on specific proteins, environmental DNA (eDNA), or polysaccharide intercellular adhesin (PIA). We found that those isolates obtained from high-shear environments, such as the lumen of a catheter, are more likely to produce PIA-mediated biofilms than those isolates obtained from a low-shear biomaterial-related infection. This suggests that PIA functions as a mechanism that is protective against shear flow. Finally, we performed selection experiments documenting the heterogeneity of biofilm accumulation molecules that function in the absence of PIA, further documenting the biofilm-forming potential of S. epidermidis.
Collapse
|