1
|
Jia Y, Yang Y, Zhang H. Comparative Study on the Lubrication Mechanism and Performance of Two Representative Ionic and Nonionic Self-Adhesive Polymer Coatings. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:8271-8283. [PMID: 38557053 DOI: 10.1021/acs.langmuir.4c00539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Surface modification of lubricating coatings on biomedical devices is a pivotal strategy to improve the overall performance and clinical efficacy, significantly reducing friction between devices and human tissues and mitigating tissue damage during intervention and long-term implantation. Recently, various hydrophilic polymeric materials have been used for achieving surface functionalization, endowing the biomedical device with excellent superlubrication performance. N-Vinylpyrrolidone (NVP) and 2-methacryloyloxyethyl phosphorylcholine (MPC) are two typical representatives of nonionic and zwitterionic materials. However, there is still a research gap in a comparative study of the lubrication mechanisms and properties between them. In this study, a bioinspired and dopamine-assisted codeposition technique was used to fabricate biomimetic hydrophilic coatings, including P(DMA-NVP) and P(DMA-MPC), on polyurethane. To achieve a thorough comparative analysis of the self-adhesive coating performance, 3 M ratios of the copolymers were synthesized and comprehensive material evaluations were conducted. Additionally, surface morphology, hydrophilicity, and lubrication at both the microscale and macroscale were performed. It was found that both hydrophilic coatings exhibited good stability. The P(DMA-MPC) coating, due to the ability to attract and bind a large number of water molecules, demonstrated superior lubrication effects compared to the P(DMA-NVP) coating. The study provides an in-depth understanding of the lubrication behavior of the self-adhesive coatings to enhance the functionality and application in biomedical engineering.
Collapse
Affiliation(s)
- Yiran Jia
- Joint Diseases Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China
- State Key Laboratory of Tribology in Advanced Equipment, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, China
| | - Yinuo Yang
- State Key Laboratory of Tribology in Advanced Equipment, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, China
| | - Hongyu Zhang
- Joint Diseases Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China
- State Key Laboratory of Tribology in Advanced Equipment, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, China
| |
Collapse
|
2
|
Mahmood Janlou MA, Sahebjamee H, Yazdani M, Fozouni L. Structure-based virtual screening and molecular dynamics approaches to identify new inhibitors of Staphylococcus aureus sortase A. J Biomol Struct Dyn 2024; 42:1157-1169. [PMID: 37184111 DOI: 10.1080/07391102.2023.2201863] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 03/28/2023] [Indexed: 05/16/2023]
Abstract
Staphylococcus aureus is a prevalent Gram-positive bacteria leading cause of a wide range of human pathologies. Moreover, antibiotic résistance of pathogenesis bacteria is one of the worldwide health problems. In Gram-positive bacteria, the enzyme of SrtA, is responsible for the anchoring of surface-exposed proteins to the cell wall peptidoglycan. Because of its critical role in Gram-positive bacterial pathogenesis, SrtA is an attractive target for anti-virulence during drug development. To date, some SrtA inhibitors have been discovered most of them being derived from flavonoid compounds, like Myricetin. In order to provide potential hit molecules against SrtA for clinical use, we obtained a total of 293 compounds by performing in silico shape-based screening of compound libraries against Myristin as a reference structure. Employing molecular docking and scoring functions, the top 3 compounds Apigenin, Efloxate, and Compound 8261032 were screened by comparing their docking scores with Myricetin. Furthermore, MD simulations and MM-PBSA binding energy calculation studies revealed that only Compound 8261032 strongly binds to the catalytic core of the SrtA enzyme than Myricetin, and stable behavior was consistently observed in the docking complex. Compound 8261032 showed a good number of hydrogen bonds with SrtA and higher MM-PBSA binding energy when compared to all three molecules. Also, it makes strength interactions with Arg139 and His62, which are critical for SrtA biological activity. This study showed that the development of this inhibitor could be a fundamental strategy against resistant bacteria, but further studies in vitro are needed to confirm this claim.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Mehr Ali Mahmood Janlou
- Department of Biophysics, Faculty of Biological Sciences, Gorgan Branch, Islamic Azad University, Gorgan, Iran
| | - Hassan Sahebjamee
- Department of Biophysics, Faculty of Biological Science, Varamin-Pishva Branch, Islamic Azad University, Varamin, Iran
| | - Mohsen Yazdani
- Laboratory of Bioinformatics and Drug Design, Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Leila Fozouni
- Department of Microbiology, Faculty of Biological Sciences, Gorgan Branch, Islamic Azad University, Gorgan, Iran
| |
Collapse
|
3
|
Alves LA, Naveed H, Franco EM, Garcia MT, Freitas VA, Junqueira JC, Bastos DC, Araujo TLS, Chen T, Mattos-Graner RO. PepO and CppA modulate Streptococcus sanguinis susceptibility to complement immunity and virulence. Virulence 2023; 14:2239519. [PMID: 37563831 PMCID: PMC10424592 DOI: 10.1080/21505594.2023.2239519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 07/12/2023] [Accepted: 07/15/2023] [Indexed: 08/12/2023] Open
Abstract
Streptococcus sanguinis is a ubiquitous commensal species of the oral cavity commonly involved as an opportunistic pathogen in cardiovascular infections. In this study, we investigated the functions of endopeptidase O (PepO) and a C3-degrading protease (CppA) in the systemic virulence of S. sanguinis. Isogenic mutants of pepO and cppA obtained in strain SK36 showed increased susceptibility to C3b deposition and to opsonophagocytosis by human polymorphonuclear neutrophils (PMN). These mutants differ, however, in their profiles of binding to serum amyloid P component (SAP) and C1q, whereas both showed reduced interaction with C4b-binding protein (C4BP) and/or factor H (FH) regulators as compared to SK36. The two mutants showed defects in ex vivo persistence in human blood, serum-mediated invasion of HCAEC endothelial cells, and virulence in a Galleria mellonella infection model. The transcriptional activities of pepO and cppA, assessed by RT-qPCR in nine wild-type strains, further indicated strain-specific profiles of pepO/cppA expression. Moreover, non-conserved amino acid substitutions were detected among the strains, mostly in CppA. Phylogenetic comparisons with homologues of streptococcal species of the oral and oropharyngeal sites suggested that S. sanguinis PepO and CppA have independent ancestralities. Thus, this study showed that PepO and CppA are complement evasion proteins expressed by S. sanguinis in a strain-specific manner, which are required for multiple functions associated with cardiovascular virulence.
Collapse
Affiliation(s)
- Lívia A. Alves
- Department of Oral Diagnosis, Piracicaba Dental School, State University of Campinas (UNICAMP), Piracicaba, SP, Brazil
| | - Hassan Naveed
- Department of Oral Diagnosis, Piracicaba Dental School, State University of Campinas (UNICAMP), Piracicaba, SP, Brazil
| | - Eduardo M. Franco
- Department of Oral Diagnosis, Piracicaba Dental School, State University of Campinas (UNICAMP), Piracicaba, SP, Brazil
| | - Maíra Terra Garcia
- Department of Biosciences and Oral Diagnosis, Institute of Science and Technology, São Paulo State University (UNESP), São José dos Campos, SP, Brazil
| | - Victor A. Freitas
- Department of Oral Diagnosis, Piracicaba Dental School, State University of Campinas (UNICAMP), Piracicaba, SP, Brazil
| | - Juliana C. Junqueira
- Department of Biosciences and Oral Diagnosis, Institute of Science and Technology, São Paulo State University (UNESP), São José dos Campos, SP, Brazil
| | - Débora C. Bastos
- Department of Biosciences, Piracicaba Dental School, State University of Campinas (UNICAMP), Piracicaba, SP, Brazil
- Department of Cell Biology, São Leopoldo Mandic Medical School, Campinas, SP, Brazil
| | - Thaís L. S. Araujo
- Department of Biochemistry, Institute of Chemistry, University of São Paulo (USP), São Paulo, SP, Brazil
| | - Tsute Chen
- Department of Microbiology, The Forsyth Institute, Cambridge, MA, USA
| | - Renata O. Mattos-Graner
- Department of Oral Diagnosis, Piracicaba Dental School, State University of Campinas (UNICAMP), Piracicaba, SP, Brazil
| |
Collapse
|
4
|
Franco EM, Alves LA, Naveed H, Freitas VAA, Bastos DC, Mattos-Graner RO. Amyloid Fibrils Produced by Streptococcus sanguinis Contribute to Biofilm Formation and Immune Evasion. Int J Mol Sci 2023; 24:15686. [PMID: 37958670 PMCID: PMC10647432 DOI: 10.3390/ijms242115686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 10/20/2023] [Indexed: 11/15/2023] Open
Abstract
Bacterial surface proteins assembled into amyloids contribute to biofilm formation and host immune evasion. Streptococcus sanguinis, a pioneer colonizer of teeth commonly involved in cardiovascular infections, expresses about thirty-three proteins anchored to the cell wall by sortase A. Here, we characterized the production of amyloid in S. sanguinis strains differing in biofilm and immune evasion phenotypes and investigated the role of sortase A in amyloidogenesis. Amyloid was identified in biofilms formed by nine strains, using Congo red (CR) staining and cross-polarized light microscopy. Additionally, EGCG, an amyloid inhibitor, impaired biofilm maturation in a strain-specific fashion. The amounts of amyloid-like components quantified in culture fluids of nine strains using thioflavin T and fluorimetry negatively correlated with bacterial binding to complement-activating proteins (SAP, C1q), C3b deposition and rates of opsonophagocytosis in PMNs, implying amyloid production in immune evasion. The deletion of the sortase A gene (srtA) in strain SK36 compromised amyloid production and sucrose-independent biofilm maturation. The srtA mutant further showed increased susceptibility to C3b deposition and altered interactions with PMNs as well as reduced persistence in human blood. These findings highlight the contribution of amyloids to biofilm formation and host immune evasion in S. sanguinis strains, further indicating the participation of sortase A substrates in amyloidogenesis.
Collapse
Affiliation(s)
- Eduardo M. Franco
- Department of Oral Diagnosis, Piracicaba Dental School, State University of Campinas, Piracicaba 13414-903, SP, Brazil; (E.M.F.); (L.A.A.); (H.N.); (V.A.A.F.)
| | - Lívia A. Alves
- Department of Oral Diagnosis, Piracicaba Dental School, State University of Campinas, Piracicaba 13414-903, SP, Brazil; (E.M.F.); (L.A.A.); (H.N.); (V.A.A.F.)
- School of Dentistry, Cruzeiro do Sul University (UNICSUL), São Paulo 01506-000, SP, Brazil
| | - Hassan Naveed
- Department of Oral Diagnosis, Piracicaba Dental School, State University of Campinas, Piracicaba 13414-903, SP, Brazil; (E.M.F.); (L.A.A.); (H.N.); (V.A.A.F.)
| | - Victor A. A. Freitas
- Department of Oral Diagnosis, Piracicaba Dental School, State University of Campinas, Piracicaba 13414-903, SP, Brazil; (E.M.F.); (L.A.A.); (H.N.); (V.A.A.F.)
| | - Débora C. Bastos
- Department of Biosciences, Piracicaba Dental School, State University of Campinas, Piracicaba 13414-903, SP, Brazil;
- São Leopoldo Mandic Medical School, Campinas 13045-755, SP, Brazil
| | - Renata O. Mattos-Graner
- Department of Oral Diagnosis, Piracicaba Dental School, State University of Campinas, Piracicaba 13414-903, SP, Brazil; (E.M.F.); (L.A.A.); (H.N.); (V.A.A.F.)
| |
Collapse
|
5
|
Wang L, Wang H, Zhang H, Wu H. Formation of a biofilm matrix network shapes polymicrobial interactions. THE ISME JOURNAL 2023; 17:467-477. [PMID: 36639539 PMCID: PMC9938193 DOI: 10.1038/s41396-023-01362-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 12/30/2022] [Accepted: 01/09/2023] [Indexed: 01/15/2023]
Abstract
Staphylococcus aureus colonizes the same ecological niche as many commensals. However, little is known about how such commensals modulate staphylococcal fitness and persistence. Here we report a new mechanism that mediates dynamic interactions between a commensal streptococcus and S. aureus. Commensal Streptococcus parasanguinis significantly increased the staphylococcal biofilm formation in vitro and enhanced its colonization in vivo. A streptococcal biofilm-associated protein BapA1, not fimbriae-associated protein Fap1, is essential for dual-species biofilm formation. On the other side, three staphylococcal virulence determinants responsible for the BapA1-dependent dual-species biofilm formation were identified by screening a staphylococcal transposon mutant library. The corresponding staphylococcal mutants lacked binding to recombinant BapA1 (rBapA1) due to lower amounts of eDNA in their culture supernatants and were defective in biofilm formation with streptococcus. The rBapA1 selectively colocalized with eDNA within the dual-species biofilm and bound to eDNA in vitro, highlighting the contributions of the biofilm matrix formed between streptococcal BapA1 and staphylococcal eDNA to dual-species biofilm formation. These findings have revealed an additional new mechanism through which an interspecies biofilm matrix network mediates polymicrobial interactions.
Collapse
Affiliation(s)
- Lijun Wang
- Departments of Pediatric Dentistry and Microbiology, University of Alabama at Birmingham Schools of Dentistry and Medicine, Birmingham, Alabama, 35294, USA
- Department of Laboratory Medicine, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, 102218, Beijing, China
| | - Hongxia Wang
- Departments of Pediatric Dentistry and Microbiology, University of Alabama at Birmingham Schools of Dentistry and Medicine, Birmingham, Alabama, 35294, USA
| | - Hua Zhang
- Departments of Pediatric Dentistry and Microbiology, University of Alabama at Birmingham Schools of Dentistry and Medicine, Birmingham, Alabama, 35294, USA
- Department of Integrative Biomedical and Diagnostic Sciences, Oregon Health and Science University School of Dentistry, Portland, OR, 97239, USA
| | - Hui Wu
- Departments of Pediatric Dentistry and Microbiology, University of Alabama at Birmingham Schools of Dentistry and Medicine, Birmingham, Alabama, 35294, USA.
- Department of Integrative Biomedical and Diagnostic Sciences, Oregon Health and Science University School of Dentistry, Portland, OR, 97239, USA.
| |
Collapse
|
6
|
Antibacterial activity of chitosan-polyethylene oxide nanofibers containing silver nanoparticles against aerobic and anaerobic bacteria. J Mol Struct 2023. [DOI: 10.1016/j.molstruc.2022.134304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
7
|
Alves LA, Salvatierra GC, Freitas VA, Höfling JF, Bastos DC, Araujo TLS, Mattos-Graner RO. Diversity in Phenotypes Associated With Host Persistence and Systemic Virulence in Streptococcus sanguinis Strains. Front Microbiol 2022; 13:875581. [PMID: 35509310 PMCID: PMC9058168 DOI: 10.3389/fmicb.2022.875581] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 03/08/2022] [Indexed: 11/13/2022] Open
Abstract
Streptococcus sanguinis is a pioneer commensal species of dental biofilms, abundant in different oral sites and commonly associated with opportunist cardiovascular infections. In this study, we addressed intra-species functional diversity to better understand the S. sanguinis commensal and pathogenic lifestyles. Multiple phenotypes were screened in nine strains isolated from dental biofilms or from the bloodstream to identify conserved and strain-specific functions involved in biofilm formation and/or persistence in oral and cardiovascular tissues. Strain phenotypes of biofilm maturation were independent of biofilm initiation phenotypes, and significantly influenced by human saliva and by aggregation mediated by sucrose-derived exopolysaccharides (EPS). The production of H2O2 was conserved in most strains, and consistent with variations in extracellular DNA (eDNA) production observed in few strains. The diversity in complement C3b deposition correlated with the rates of opsonophagocytosis by human PMN and was influenced by culture medium and sucrose-derived EPS in a strain-specific fashion. Differences in C3b deposition correlated with strain binding to recognition proteins of the classical pathway, C1q and serum amyloid protein (SAP). Importantly, differences in strain invasiveness into primary human coronary artery endothelial cells (HCAEC) were significantly associated with C3b binding, and in a lesser extent, with binding to host glycoproteins (such as fibrinogen, plasminogen, fibronectin, and collagen). Thus, by identifying conserved and strain-specific phenotypes involved in host persistence and systemic virulence, this study indicates potential new functions involved in systemic virulence and highlights the need of including a wider panel of strains in molecular studies to understand S. sanguinis biology.
Collapse
Affiliation(s)
- Livia A. Alves
- Department of Oral Diagnosis, Piracicaba Dental School, State University of Campinas, Piracicaba, Brazil
| | - Geovanny C. Salvatierra
- Department of Oral Diagnosis, Piracicaba Dental School, State University of Campinas, Piracicaba, Brazil
| | - Victor A. Freitas
- Department of Oral Diagnosis, Piracicaba Dental School, State University of Campinas, Piracicaba, Brazil
| | - José F. Höfling
- Department of Oral Diagnosis, Piracicaba Dental School, State University of Campinas, Piracicaba, Brazil
| | - Débora C. Bastos
- Department of Biosciences, Piracicaba Dental School, State University of Campinas, Piracicaba, Brazil
- São Leopoldo Mandic Medical School, Campinas, Brazil
| | - Thaís L. S. Araujo
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Renata O. Mattos-Graner
- Department of Oral Diagnosis, Piracicaba Dental School, State University of Campinas, Piracicaba, Brazil
- *Correspondence: Renata O. Mattos-Graner,
| |
Collapse
|
8
|
Katsumata T, Nguyen-Tra Le M, Kawada-Matsuo M, Taniguchi Y, Ouhara K, Oogai Y, Nakata M, Mizuno N, Nishitani Y, Komatsuzawa H. KATSUMATA et al.Comprehensive characterization of sortase A-dependent surface proteins in Streptococcus mutansComprehensive characterization of sortase A-dependent surface proteins in Streptococcus mutans. Microbiol Immunol 2021; 66:145-156. [PMID: 34888908 DOI: 10.1111/1348-0421.12958] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 11/30/2021] [Accepted: 12/01/2021] [Indexed: 11/29/2022]
Abstract
Streptococcus mutans, a cariogenic pathogen, adheres to the tooth surface and forms a biofilm. Bacterial cell surface proteins are associated with adherence to substrates. Sortase A (SrtA) mediates the localization of proteins with an LPXTG motif-containing proteins to the cell surface by covalent binding to peptidoglycan. In S. mutans UA159, 6 SrtA-dependent proteins, SpaP, WapA, WapE, DexA, FruA, and GbpC, were identified. Although some of these proteins were characterized, a comprehensive analysis of the 6 proteins has not been reported. In this study, we constructed mutants deficient in each of these proteins and the SrtA-deficient mutant. The SrtA-deficient mutant showed drastically decreased binding to salivary components, biofilm formation, bacterial coaggregation activity, hydrophobicity, and cellular matrix binding (collagen type I, fibronectin, and laminin). The SpaP-deficient mutant showed significantly reduced binding to salivary components and partially increased coaggregation with Porphyromonas gingivalis, and decreased hydrophobicity, and collagen binding. The WapA-deficient mutant showed slightly decreased coaggregation with Fusobacterium nucleatum. Although the SrtA-deficient mutant showed drastically altered phenotypes, all SrtA-dependent protein-deficient mutants, except the SpaP-deficient mutant, did not show considerable alterations in binding to salivary components. These results indicate that the 6 proteins may coordinately contribute to these activities. In addition, using genomic data of 125 S. mutans strains, we compared the amino acid sequences of each surface protein and found many variations among strains, which may affect the phenotype of cell surface proteins in S. mutans. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Tamaki Katsumata
- Department of Restorative Dentistry and Endodontology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Mi Nguyen-Tra Le
- Department of Bacteriology, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Miki Kawada-Matsuo
- Department of Bacteriology, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Yuri Taniguchi
- Department of Periodontal Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Kazuhisa Ouhara
- Department of Periodontal Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Yuichi Oogai
- Department of Oral Microbiology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Masanobu Nakata
- Department of Oral Microbiology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Noriyoshi Mizuno
- Department of Periodontal Medicine, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Yoshihiro Nishitani
- Department of Restorative Dentistry and Endodontology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Hitoshi Komatsuzawa
- Department of Bacteriology, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| |
Collapse
|
9
|
Han Y, Zhao W, Zheng Y, Wang H, Sun Y, Zhang Y, Luo J, Zhang H. Self-adhesive lubricated coating for enhanced bacterial resistance. Bioact Mater 2021; 6:2535-2545. [PMID: 33615044 PMCID: PMC7868611 DOI: 10.1016/j.bioactmat.2021.01.028] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 01/06/2021] [Accepted: 01/22/2021] [Indexed: 12/11/2022] Open
Abstract
Limited surface lubrication and bacterial biofilm formation pose great challenges to biomedical implants. Although hydrophilic lubricated coatings and bacterial resistance coatings have been reported, the harsh and tedious synthesis greatly compromises their application, and more importantly, the bacterial resistance property has seldom been investigated in combination with the lubrication property. In this study, bioinspired by the performances of mussel and articular cartilage, we successfully synthesized self-adhesive lubricated coating and simultaneously achieved optimal lubrication and bacterial resistance properties. Additionally, we reported the mechanism of bacterial resistance on the nanoscale by studying the adhesion interactions between biomimetic coating and hydrophilic/hydrophobic tip or living bacteria via atomic force microscopy. In summary, the self-adhesive lubricated coating can effectively enhance lubrication and bacterial resistance performances based on hydration lubrication and hydration repulsion, and represent a universal and facial strategy for surface functionalization of biomedical implants.
Collapse
Affiliation(s)
- Ying Han
- State Key Laboratory of Tribology, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
| | - Weiwei Zhao
- State Key Laboratory of Tribology, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
| | - Yiwei Zheng
- State Key Laboratory of Tribology, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
| | - Haimang Wang
- State Key Laboratory of Tribology, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
| | - Yulong Sun
- State Key Laboratory of Tribology, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
| | - Yifei Zhang
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, 100081, China
| | - Jing Luo
- Beijing Research Institute of Automation for Machinery Industry Co., Ltd, Beijing, 100120, China
| | - Hongyu Zhang
- State Key Laboratory of Tribology, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
| |
Collapse
|
10
|
Álvarez S, Leiva-Sabadini C, Schuh CMAP, Aguayo S. Bacterial adhesion to collagens: implications for biofilm formation and disease progression in the oral cavity. Crit Rev Microbiol 2021; 48:83-95. [PMID: 34270375 DOI: 10.1080/1040841x.2021.1944054] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Collagen is the most abundant structural protein in the body and the main component of the extracellular matrix of most tissues, including dentine and periodontal tissues. Despite the well-characterized role of collagen and specifically type-I collagen, as a ligand for host cells, its role as a substrate for bacterial adhesion and biofilm formation is less explored. Therefore, the purpose of this review is to discuss recent findings regarding the adhesion of oral bacteria to collagen surfaces and its role in the progression and severity of oral and systemic diseases. Initial oral colonizers such as streptococci have evolved collagen-binding proteins (cbp) that are important for the colonization of dentine and periodontal tissues. Also, periodontal pathogens such as Porphyromonas gingivalis and Tannerella forsythia utilise cbps for tissue sensing and subsequent invasion. The implications of bacteria-collagen coupling in the context of collagen biomaterials and regenerative dentistry approaches are also addressed. Furthermore, the importance of interdisciplinary techniques such as atomic force microscopy for the nanocharacterization of bacteria-collagen interactions is also considered. Overall, understanding the process of oral bacterial adhesion onto collagen is important for developing future therapeutic approaches against oral and systemic diseases, by modulating the early stages of biofilm formation.
Collapse
Affiliation(s)
- Simón Álvarez
- Centro de Medicina Regenerativa, Facultad de Medicina Clínica Alemana-Universidad del Desarrollo, Santiago, Chile.,Dentistry School, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Camila Leiva-Sabadini
- Dentistry School, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Christina M A P Schuh
- Centro de Medicina Regenerativa, Facultad de Medicina Clínica Alemana-Universidad del Desarrollo, Santiago, Chile
| | - Sebastian Aguayo
- Dentistry School, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile.,Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
11
|
Noda M, Sugihara N, Sugimoto Y, Hayashi I, Sugimoto S, Danshiitsoodol N, Sugiyama M. Lactobacillus reuteri BM53-1 Produces a Compound That Inhibits Sticky Glucan Synthesis by Streptococcus mutans. Microorganisms 2021; 9:microorganisms9071390. [PMID: 34199080 PMCID: PMC8307965 DOI: 10.3390/microorganisms9071390] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 06/17/2021] [Accepted: 06/23/2021] [Indexed: 11/21/2022] Open
Abstract
Cariogenic bacteria, such as Streptococcus (S.) mutans and S. sobrinus, produce insoluble and sticky glucans as a biofilm material. The present study demonstrates that a lactic acid bacterium (LAB) named BM53-1 produces a substance that inhibits the sticky glucan synthesis. The BM53-1 strain was isolated from a flower of Actinidia polygama and identified as Lactobacillus reuteri. The substance that inhibits sticky glucan synthesis does not exhibit antibacterial activity against S. mutans. The cariogenic S. mutans produces glucans under the control of three glucosyltransferase (GTF) enzymes, named GtfB, GtfC, and GtfD. Although GtfB and GtfC produce insoluble glucans, GtfD forms soluble glucans. Through quantitative reverse-transcriptional (qRT)-PCR analysis, it was revealed that the BM53-1-derived glucan-production inhibitor (GI) enhances the transcriptions of gtfB and gtfC genes 2- to 7-fold at the early stage of cultivation. However, that of gtfD was not enhanced in the presence of the GI, indicating that the glucan stickiness produced by S. mutans was significantly weaker in the presence of the GI. Our result demonstrates that Lb. reuteri BM53-1 is useful to prevent dental caries.
Collapse
Affiliation(s)
- Masafumi Noda
- Department of Probiotic Science for Preventive Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan; (M.N.); (N.S.); (Y.S.); (N.D.)
| | - Naho Sugihara
- Department of Probiotic Science for Preventive Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan; (M.N.); (N.S.); (Y.S.); (N.D.)
| | - Yoshimi Sugimoto
- Department of Probiotic Science for Preventive Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan; (M.N.); (N.S.); (Y.S.); (N.D.)
| | - Ikue Hayashi
- Central Research Laboratory, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan;
| | - Sachiko Sugimoto
- Department of Pharmacognosy, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan;
| | - Narandalai Danshiitsoodol
- Department of Probiotic Science for Preventive Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan; (M.N.); (N.S.); (Y.S.); (N.D.)
| | - Masanori Sugiyama
- Department of Probiotic Science for Preventive Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan; (M.N.); (N.S.); (Y.S.); (N.D.)
- Correspondence: ; Tel.: +81-(82)-257-5280
| |
Collapse
|
12
|
Susmitha A, Bajaj H, Madhavan Nampoothiri K. The divergent roles of sortase in the biology of Gram-positive bacteria. ACTA ACUST UNITED AC 2021; 7:100055. [PMID: 34195501 PMCID: PMC8225981 DOI: 10.1016/j.tcsw.2021.100055] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 06/11/2021] [Accepted: 06/11/2021] [Indexed: 12/16/2022]
Abstract
The bacterial cell wall contains numerous surface-exposed proteins, which are covalently anchored and assembled by a sortase family of transpeptidase enzymes. The sortase are cysteine transpeptidases that catalyzes the covalent attachment of surface protein to the cell wall peptidoglycan. Among the reported six classes of sortases, each distinct class of sortase plays a unique biological role in anchoring a variety of surface proteins to the peptidoglycan of both pathogenic and non-pathogenic Gram-positive bacteria. Sortases not only exhibit virulence and pathogenesis properties to host cells, but also possess a significant role in gut retention and immunomodulation in probiotic microbes. The two main distinct functions are to attach proteins directly to the cell wall or assemble pili on the microbial surface. This review provides a compendium of the distribution of different classes of sortases present in both pathogenic and non-pathogenic Gram-positive bacteria and also the noteworthy role played by them in bacterial cell wall assembly which enables each microbe to effectively interact with its environment.
Collapse
Affiliation(s)
- Aliyath Susmitha
- Microbial Processes and Technology Division, CSIR - National Institute for Interdisciplinary Science and Technology (NIIST), Trivandrum 695019, Kerala, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Harsha Bajaj
- Microbial Processes and Technology Division, CSIR - National Institute for Interdisciplinary Science and Technology (NIIST), Trivandrum 695019, Kerala, India
| | - Kesavan Madhavan Nampoothiri
- Microbial Processes and Technology Division, CSIR - National Institute for Interdisciplinary Science and Technology (NIIST), Trivandrum 695019, Kerala, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
13
|
Oba PM, Carroll MQ, Alexander C, Valentine H, Somrak AJ, Keating SCJ, Sage AM, Swanson KS. Microbiota populations in supragingival plaque, subgingival plaque, and saliva habitats of adult dogs. Anim Microbiome 2021; 3:38. [PMID: 34001282 PMCID: PMC8130298 DOI: 10.1186/s42523-021-00100-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 04/30/2021] [Indexed: 02/01/2023] Open
Abstract
Background Oral diseases are common in dogs, with microbiota playing a prominent role in the disease process. Oral cavity habitats harbor unique microbiota populations that have relevance to health and disease. Despite their importance, the canine oral cavity microbial habitats have been poorly studied. The objectives of this study were to (1) characterize the oral microbiota of different habitats of dogs and (2) correlate oral health scores with bacterial taxa and identify what sites may be good options for understanding the role of microbiota in oral diseases. We used next-generation sequencing to characterize the salivary (SAL), subgingival (SUB), and supragingival (SUP) microbial habitats of 26 healthy adult female Beagle dogs (4.0 ± 1.2 year old) and identify taxa associated with periodontal disease indices. Results Bacterial species richness was highest for SAL, moderate for SUB, and lowest for SUP samples (p < 0.001). Unweighted and weighted principal coordinates plots showed clustering by habitat, with SAL and SUP samples being the most different from one another. Bacteroidetes, Proteobacteria, Firmicutes, Fusobacteria, Actinobacteria, and Spirochaetes were the predominant phyla in all habitats. Paludibacter, Filifactor, Peptostreptococcus, Fusibacter, Anaerovorax, Fusobacterium, Leptotrichia, Desulfomicrobium, and TG5 were enriched in SUB samples, while Actinomyces, Corynebacterium, Leucobacter, Euzebya, Capnocytophaga, Bergeyella, Lautropia, Lampropedia, Desulfobulbus, Enhydrobacter, and Moraxella were enriched in SUP samples. Prevotella, SHD-231, Helcococcus, Treponema, and Acholeplasma were enriched in SAL samples. p-75-a5, Arcobacter, and Pasteurella were diminished in SUB samples. Porphyromonas, Peptococcus, Parvimonas, and Campylobacter were diminished in SUP samples, while Tannerella, Proteocalla, Schwartzia, and Neisseria were diminished in SAL samples. Actinomyces, Corynebacterium, Capnocytophaga, Leptotrichia, and Neisseria were associated with higher oral health scores (worsened health) in plaque samples. Conclusions Our results demonstrate the differences that exist among canine salivary, subgingival plaque and supragingival plaque habitats. Salivary samples do not require sedation and are easy to collect, but do not accurately represent the plaque populations that are most important to oral disease. Plaque Actinomyces, Corynebacterium, Capnocytophaga, Leptotrichia, and Neisseria were associated with higher (worse) oral health scores. Future studies analyzing samples from progressive disease stages are needed to validate these results and understand the role of bacteria in periodontal disease development.
Collapse
Affiliation(s)
- Patrícia M Oba
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, 1207 West Gregory Drive, 162 Animal Sciences Laboratory, Urbana, IL, 61801, USA
| | - Meredith Q Carroll
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, 1207 West Gregory Drive, 162 Animal Sciences Laboratory, Urbana, IL, 61801, USA
| | - Celeste Alexander
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Helen Valentine
- Division of Animal Resources, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Amy J Somrak
- Department of Veterinary Clinical Medicine, College of Veterinary Medicine, University of Illinois, Urbana, IL, 61801, USA
| | - Stephanie C J Keating
- Department of Veterinary Clinical Medicine, College of Veterinary Medicine, University of Illinois, Urbana, IL, 61801, USA
| | - Adrianna M Sage
- Department of Surgical Sciences, School of Veterinary Medicine, University of Wisconsin - Madison, 2015 Linden Dr, Madison, WI, 53706, USA
| | - Kelly S Swanson
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, 1207 West Gregory Drive, 162 Animal Sciences Laboratory, Urbana, IL, 61801, USA. .,Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.
| |
Collapse
|
14
|
Pei X, Liu M, Zhou H, Fan H. Screening for phagocytosis resistance-related genes via a transposon mutant library of Streptococcus suis serotype 2. Virulence 2021; 11:825-838. [PMID: 32614642 PMCID: PMC7567436 DOI: 10.1080/21505594.2020.1782088] [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: 12/22/2022] Open
Abstract
Streptococcus suis serotype 2 (SS2) is a serious zoonotic pathogen which causes symptoms of streptococcal toxic shock syndrome (STSS) and septicemia; these symptoms suggest that SS2 may have evade innate immunity. Phagocytosis is an important innate immunity process where phagocytosed pathogens are killed by lysosome enzymes, reactive oxygen, and nitrogen species, and acidic environments in macrophages following engulfment. A previously constructed mutant SS2 library was screened, revealing 13 mutant strains with decreased phagocytic resistance. Through inverse PCR, the transposon insertion sites were determined. Through bioinformatic analysis, the 13 disrupted genes were identified as Cps2F, 3 genes belonging to ABC transporters, WalR, TehB, rpiA, S-transferase encoding gene, prs, HsdM, GNAT family N-acetyltransferase encoding gene, proB, and upstream region of DnaK. Except for the capsular polysaccharide biosynthesis associated Cps2F, the other genes had not been linked to a role in anti-phagocytosis. The survival ability in macrophages and whole blood of randomly picked mutant strains were significantly impaired compared with wild-type ZY05719. The virulence of the mutant strains was also attenuated in a mouse infection model. In the WalR mutant, the transcription of HP1065 decreased significantly compared with wild-type strain, indicating WalR might regulated HP1065 expression and contribute to the anti-phagocytosis of SS2. In conclusion, we identified 13 genes that influenced the phagocytosis resistant ability of SS2, and many of these genes have not been reported to be associated with resistance to phagocytosis. Our work provides novel insight into resistance to phagocytosis, and furthers our understanding of the pathogenesis mechanism of SS2.
Collapse
Affiliation(s)
- Xiaomeng Pei
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University , Nanjing, China
| | - Mingxing Liu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University , Nanjing, China
| | - Hong Zhou
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University , Nanjing, China
| | - Hongjie Fan
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University , Nanjing, China.,Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University , Yangzhou, China
| |
Collapse
|
15
|
Yamaguchi M. [Investigation of pneumococcal virulence factors in the infection process]. Nihon Saikingaku Zasshi 2020; 75:173-183. [PMID: 33361653 DOI: 10.3412/jsb.75.173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
This review summarizes current knowledge regarding the pathological mechanism of Streptococcus pneumoniae, a major cause of pneumonia, sepsis, and meningitis, with focus on our previously presented studies.To identify pneumococcal adhesins or invasins on cell surfaces, we investigated several proteins with an LPXTG anchoring motif and identified one showing interaction with human fibronectin, which was designated PfbA. Next, the mechanism of pneumococcal evasion form host immunity system in blood was examined and pneumococcal α-Enolase was found to function as a neutrophil extracellular trap induction factor. Although S. pneumoniae organisms are partially killed by iron ion-induced free radicals, they have an ability to invade red blood cells and then evade antibiotics, neutrophil phagocytosis, and H2O2 killing. In addition, our findings have indicated that zinc metalloprotease ZmpC suppresses pneumococcal virulence by inhibiting bacterial invasion of the central nervous system. Since evolutionarily conserved virulence factors are potential candidate therapeutic targets, we performed molecular evolutionary analyses, which revealed that cbpJ had the highest rate of codons under negative selection to total number of codons among genes encoding choline-binding proteins. Our experimental analysis results indicated that CbpJ functions as a virulence factor in pneumococcal pneumonia by contributing to evasion of neutrophil killing.Use of a molecular biological approach based on bacterial genome sequences, clinical disease states, and molecular evolutionary analysis is an effective strategy for revealing virulence factors and important therapeutic targets.
Collapse
Affiliation(s)
- Masaya Yamaguchi
- Department of Oral and Molecular Microbiology, Osaka University Graduate School of Dentistry
| |
Collapse
|
16
|
Pei X, Liu J, Liu M, Zhou H, Wang X, Fan H. Quantitative proteomics revealed modulation of macrophages by MetQ gene of Streptococcus suis serotype 2. AMB Express 2020; 10:195. [PMID: 33125582 PMCID: PMC7599288 DOI: 10.1186/s13568-020-01131-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 10/18/2020] [Indexed: 11/10/2022] Open
Abstract
Streptococcus suis serotype 2 (SS2) is a serious zoonotic pathogen; it can lead to symptoms of streptococcal toxic shock syndrome (STSS) in humans and sepsis in pigs, and poses a great threat to public health. The SS2 MetQ gene deletion strain has attenuated antiphagocytosis, although the mechanism of antiphagocytosis and pathogenesis of MetQ in SS2 has remained unclear. In this study, stable isotope labeling by amino acids in cell culture (SILAC) based liquid chromatography–mass spectrometry (LC–MS) and subsequent bioinformatics analysis was used to determine differentially expressed proteins of RAW264.7 cells infected with △MetQ and ZY05719. Proteomic results were verified by quantitative real-time polymerase chain reaction (qRT-PCR) and Western blotting for selected proteins. Further research was focused mainly on immune system processes related to downregulated proteins, such as Src and Ccl9, and actin cytoskeleton and endocytosis related upregulated proteins, like Pstpip1 and Ppp1r9b. The proteomic results in this study shed light on the mechanism of antiphagocytosis and innate immunity of macrophages infected with △MetQ and ZY05719, which might provide novel targets to prevent or control the infection of SS2.
Collapse
|
17
|
Wang CH, Hsieh YH, Powers ZM, Kao CY. Defeating Antibiotic-Resistant Bacteria: Exploring Alternative Therapies for a Post-Antibiotic Era. Int J Mol Sci 2020; 21:E1061. [PMID: 32033477 PMCID: PMC7037027 DOI: 10.3390/ijms21031061] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 02/04/2020] [Accepted: 02/04/2020] [Indexed: 12/11/2022] Open
Abstract
Antibiotics are one of the greatest medical advances of the 20th century, however, they are quickly becoming useless due to antibiotic resistance that has been augmented by poor antibiotic stewardship and a void in novel antibiotic discovery. Few novel classes of antibiotics have been discovered since 1960, and the pipeline of antibiotics under development is limited. We therefore are heading for a post-antibiotic era in which common infections become untreatable and once again deadly. There is thus an emergent need for both novel classes of antibiotics and novel approaches to treatment, including the repurposing of existing drugs or preclinical compounds and expanded implementation of combination therapies. In this review, we highlight to utilize alternative drug targets/therapies such as combinational therapy, anti-regulator, anti-signal transduction, anti-virulence, anti-toxin, engineered bacteriophages, and microbiome, to defeat antibiotic-resistant bacteria.
Collapse
Affiliation(s)
- Chih-Hung Wang
- Department of Power Mechanical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan;
| | - Yi-Hsien Hsieh
- Institute of Biochemistry, Microbiology and Immunology, Chung Shan Medical University, Taichung 40201, Taiwan;
| | - Zachary M. Powers
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI 53706, USA;
| | - Cheng-Yen Kao
- Institute of Microbiology and Immunology, School of Life Science, National Yang-Ming University, Taipei 11221, Taiwan
| |
Collapse
|
18
|
Martini AM, Moricz BS, Ripperger AK, Tran PM, Sharp ME, Forsythe AN, Kulhankova K, Salgado-Pabón W, Jones BD. Association of Novel Streptococcus sanguinis Virulence Factors With Pathogenesis in a Native Valve Infective Endocarditis Model. Front Microbiol 2020; 11:10. [PMID: 32082276 PMCID: PMC7005726 DOI: 10.3389/fmicb.2020.00010] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 01/06/2020] [Indexed: 12/11/2022] Open
Abstract
Streptococcus sanguinis (S. sanguinis) is an abundant oral commensal which can cause disseminated human infection if it gains access to the bloodstream. The most important among these diseases is infective endocarditis (IE). While virulence phenotypes of S. sanguinis have been correlated to disease severity, genetic factors mediating these phenotypes, and contributing to pathogenesis are largely uncharacterized. In this report, we investigate the roles of 128 genes in virulence-related phenotypes of S. sanguinis and characterize the pathogenic potential of two selected mutants in a left-sided, native valve IE rabbit model. Assays determining the ability of our mutant strains to produce a biofilm, bind to and aggregate platelets, and adhere to or invade endothelial cells identified sixteen genes with novel association to these phenotypes. These results suggest the presence of many uncharacterized genes involved in IE pathogenesis which may be relevant for disease progression. Two mutants identified by the above screening process – SSA_1099, encoding an RTX-like protein, and mur2, encoding a peptidoglycan hydrolase – were subsequently evaluated in vivo. Wild type (WT) S. sanguinis reliably induced cardiac vegetations, while the SSA_1099 and mur2 mutants produced either no vegetation or vegetations of small size. Splenomegaly was reduced in both mutant strains compared to WT, while pathology of other distal organs was indistinguishable. Histopathology analyses suggest the cardiac lesions and vegetations in this model resemble those observed in humans. These data indicate that SSA_1099 and mur2 encode virulence factors in S. sanguinis which are integral to pathogenesis of IE.
Collapse
Affiliation(s)
- Anthony M Martini
- Department of Microbiology & Immunology, The Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, United States
| | - Bridget S Moricz
- Department of Microbiology & Immunology, The Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, United States
| | - Allison K Ripperger
- Department of Microbiology & Immunology, The Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, United States
| | - Phuong M Tran
- Department of Microbiology & Immunology, The Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, United States
| | - Molly E Sharp
- Department of Microbiology & Immunology, The Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, United States
| | - Ana N Forsythe
- Department of Microbiology & Immunology, The Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, United States
| | - Katarina Kulhankova
- Department of Microbiology & Immunology, The Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, United States
| | - Wilmara Salgado-Pabón
- Department of Microbiology & Immunology, The Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, United States
| | - Bradley D Jones
- Department of Microbiology & Immunology, The Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, IA, United States.,Graduate Program in Genetics, The University of Iowa, Iowa City, IA, United States
| |
Collapse
|
19
|
Shafiei Z, Rahim ZHA, Philip K, Thurairajah N, Yaacob H. Potential effects of Psidium sp., Mangifera sp., Mentha sp. and its mixture (PEM) in reducing bacterial populations in biofilms, adherence and acid production of S. sanguinis and S. mutans. Arch Oral Biol 2019; 109:104554. [PMID: 31563709 DOI: 10.1016/j.archoralbio.2019.104554] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 09/12/2019] [Accepted: 09/14/2019] [Indexed: 11/19/2022]
Abstract
OBJECTIVE Psidium sp., Mangifera sp. and Mentha sp. and its mixture (PEM) are known to have antimicrobial and anti-adherence effects. DESIGN Here, we have investigated these individual plant extracts and its synergistic mixture (PEM) for its anti-cariogenic effect to reduce populations of single and mixed-species of Streptococcus sanguinis and Streptococcus mutans in a planktonic or/and biofilm and their others reduced virulence. Bacterial populations in the biofilm after 24 h, hydrophobic cell surface activity to n-hexadecane and pH changes at 5 min' intervals until 90 min of incubation were recorded. Total phenolic content and bioactive compounds in the crude aqueous plant extracts were analysed. Regulatory gene expressions of S. mutans adhesins genes (gtfB, gtfC, gbpB and spaP) upon treatment with PEM were investigated in planktonic and biofilm conditions. RESULTS All plant extracts strongly reduced S. mutans in the biofilm compared to S. sanguinis in single and mixed-species. PEM reduced S. mutans by 84% with S. sanguinis 87% in the mixed population. Psidium sp. and PEM highly reduced cell-surface hydrophobicity of the two bacteria thus reducing adherence and biofilm formation. PEM and Mangifera sp. lowered initial pH change in the mixed populations of S. sanguinis and S. mutans. PEM downregulated the S. mutans gtfB gene expression in the single species planktonic and mixed-species biofilms. CONCLUSIONS The effectiveness of PEM in reducing S. mutans within the biofilm, cell-surface hydrophobicity, acid production and adhesin gene (gtfB) expression in mixed-species with S. sanguinis indicates its potential as an antibacterial agent against dental caries. This is attributed to the phenolic content in the PEM.
Collapse
Affiliation(s)
- Zaleha Shafiei
- Dean's office, Department of Oral and Craniofacial Sciences, Faculty of Dentistry, University of Malaya, 50603 Kuala Lumpur, Malaysia; Centre for Craniofacial Diagnostics and Biosciences, Faculty of Dentistry, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300 Kuala Lumpur, Malaysia.
| | - Zubaidah Haji Abdul Rahim
- Dean's office, Department of Oral and Craniofacial Sciences, Faculty of Dentistry, University of Malaya, 50603 Kuala Lumpur, Malaysia.
| | - Koshy Philip
- Institute of Ocean & Earth Sciences, University of Malaya, 50603 Kuala Lumpur, Malaysia; Lincoln University College, Wisma Lincoln, Jalan SS6/12, 47301 Petaling Jaya, Selangor, Malaysia.
| | - Nalina Thurairajah
- Centre for Pre-U studies, UCSI University, No.1, Jalan Menara Gading, UCSI Heights, Cheras, 56000, Kuala Lumpur, Malaysia.
| | - Hashim Yaacob
- Dean's office, Department of Oral and Craniofacial Sciences, Faculty of Dentistry, University of Malaya, 50603 Kuala Lumpur, Malaysia.
| |
Collapse
|
20
|
Hirose Y, Yamaguchi M, Goto K, Sumitomo T, Nakata M, Kawabata S. Competence-induced protein Ccs4 facilitates pneumococcal invasion into brain tissue and virulence in meningitis. Virulence 2019; 9:1576-1587. [PMID: 30251911 PMCID: PMC6177246 DOI: 10.1080/21505594.2018.1526530] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Streptococcus pneumoniae is a major pathogen that causes pneumonia, sepsis, and meningitis. The candidate combox site 4 (ccs4) gene has been reported to be a pneumococcal competence-induced gene. Such genes are involved in development of S. pneumoniae competence and virulence, though the functions of ccs4 remain unknown. In the present study, the role of Ccs4 in the pathogenesis of pneumococcal meningitis was examined. We initially constructed a ccs4 deletion mutant and complement strains, then examined their association with and invasion into human brain microvascular endothelial cells. Wild-type and Ccs4-complemented strains exhibited significantly higher rates of association and invasion as compared to the ccs4 mutant strain. Deletion of ccs4 did not change bacterial growth activity or expression of NanA and CbpA, known brain endothelial pneumococcal adhesins. Next, mice were infected either intravenously or intranasally with pneumococcal strains. In the intranasal infection model, survival rates were comparable between wild-type strain-infected and ccs4 mutant strain-infected mice, while the ccs4 mutant strain exhibited a lower level of virulence in the intravenous infection model. In addition, at 24 hours after intravenous infection, the bacterial burden in blood was comparable between the wild-type and ccs4 mutant strain-infected mice, whereas the wild-type strain-infected mice showed a significantly higher bacterial burden in the brain. These results suggest that Ccs4 contributes to pneumococcal invasion of host brain tissues and functions as a virulence factor.
Collapse
Affiliation(s)
- Yujiro Hirose
- a Department of Oral and Molecular Microbiology , Osaka University Graduate School of Dentistry , Suita , Osaka , Japan
| | - Masaya Yamaguchi
- a Department of Oral and Molecular Microbiology , Osaka University Graduate School of Dentistry , Suita , Osaka , Japan
| | - Kana Goto
- a Department of Oral and Molecular Microbiology , Osaka University Graduate School of Dentistry , Suita , Osaka , Japan
| | - Tomoko Sumitomo
- a Department of Oral and Molecular Microbiology , Osaka University Graduate School of Dentistry , Suita , Osaka , Japan
| | - Masanobu Nakata
- a Department of Oral and Molecular Microbiology , Osaka University Graduate School of Dentistry , Suita , Osaka , Japan
| | - Shigetada Kawabata
- a Department of Oral and Molecular Microbiology , Osaka University Graduate School of Dentistry , Suita , Osaka , Japan
| |
Collapse
|
21
|
Identification of evolutionarily conserved virulence factor by selective pressure analysis of Streptococcus pneumoniae. Commun Biol 2019; 2:96. [PMID: 30886906 PMCID: PMC6408437 DOI: 10.1038/s42003-019-0340-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 02/06/2019] [Indexed: 01/09/2023] Open
Abstract
Evolutionarily conserved virulence factors can be candidate therapeutic targets or vaccine antigens. Here, we investigated the evolutionary selective pressures on 16 pneumococcal choline-binding cell-surface proteins since Streptococcus pneumoniae is one of the pathogens posing the greatest threats to human health. Phylogenetic and molecular analyses revealed that cbpJ had the highest codon rates to total numbers of codons under considerable negative selection among those examined. Our in vitro and in vivo assays indicated that CbpJ functions as a virulence factor in pneumococcal pneumonia by contributing to evasion of neutrophil killing. Deficiency of cbpL under relaxed selective pressure also caused a similar tendency but showed no significant difference in mouse intranasal infection. Thus, molecular evolutionary analysis is a powerful tool that reveals the importance of virulence factors in real-world infection and transmission, since calculations are performed based on bacterial genome diversity following transmission of infection in an uncontrolled population.
Collapse
|
22
|
Genomic, Phenotypic, and Virulence Analysis of Streptococcus sanguinis Oral and Infective-Endocarditis Isolates. Infect Immun 2018; 87:IAI.00703-18. [PMID: 30396893 DOI: 10.1128/iai.00703-18] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 10/26/2018] [Indexed: 12/22/2022] Open
Abstract
Streptococcus sanguinis, an abundant and benign inhabitant of the oral cavity, is an important etiologic agent of infective endocarditis (IE), particularly in people with predisposing cardiac valvular damage. Although commonly isolated from patients with IE, little is known about the factors that make any particular S. sanguinis isolate more virulent than another or, indeed, whether significant differences in virulence exist among isolates. In this study, we compared the genomes of a collection of S. sanguinis strains comprised of both oral isolates and bloodstream isolates from patients diagnosed with IE. Oral and IE isolates could not be distinguished by phylogenetic analyses, and we did not succeed in identifying virulence genes unique to the IE strains. We then investigated the virulence of these strains in a rabbit model of IE using a variation of the Bar-seq (barcode sequencing) method wherein we pooled the strains and used Illumina sequencing to count unique barcodes that had been inserted into each isolate at a conserved intergenic region. After we determined that several of the genome sequences were misidentified in GenBank, our virulence results were used to inform our bioinformatic analyses, identifying genes that may explain the heterogeneity in virulence. We further characterized these strains by assaying for phenotypes potentially contributing to virulence. Neither strain competition via bacteriocin production nor biofilm formation showed any apparent relationship with virulence. Increased cell-associated manganese was, however, correlated with blood isolates. These results, combined with additional phenotypic assays, suggest that S. sanguinis virulence is highly variable and results from multiple genetic factors.
Collapse
|
23
|
Synergistic findings from microbiological and evolutional analyses of virulence factors among pathogenic streptococcal species. J Oral Biosci 2018. [DOI: 10.1016/j.job.2018.02.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
24
|
Novel Two-Component System of Streptococcus sanguinis Affecting Functions Associated with Viability in Saliva and Biofilm Formation. Infect Immun 2018; 86:IAI.00942-17. [PMID: 29339459 DOI: 10.1128/iai.00942-17] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 01/08/2018] [Indexed: 02/07/2023] Open
Abstract
Streptococcus sanguinis is a pioneer species of teeth and a common opportunistic pathogen of infective endocarditis. In this study, we identified a two-component system, S. sanguinis SptRS (SptRS Ss ), affecting S. sanguinis survival in saliva and biofilm formation. Isogenic mutants of sptRSs (SKsptR) and sptSSs (SKsptS) showed reduced cell counts in ex vivo assays of viability in saliva compared to those of parent strain SK36 and complemented mutants. Reduced counts of the mutants in saliva were associated with reduced growth rates in nutrient-poor medium (RPMI) and increased susceptibility to the deposition of C3b and the membrane attach complex (MAC) of the complement system, a defense component of saliva and serum. Conversely, sptRSs and sptSSs mutants showed increased biofilm formation associated with higher levels of production of H2O2 and extracellular DNA. Reverse transcription-quantitative PCR (RT-qPCR) comparisons of strains indicated a global role of SptRS Ss in repressing genes for H2O2 production (2.5- to 15-fold upregulation of spxB, spxR, vicR, tpk, and ackA in sptRSs and sptSSs mutants), biofilm formation, and/or evasion of host immunity (2.1- to 11.4-fold upregulation of srtA, pcsB, cwdP, iga, and nt5e). Compatible with the homology of SptR Ss with AraC-type regulators, duplicate to multiple conserved repeats were identified in 1,000-bp regulatory regions of downstream genes, suggesting that SptR Ss regulates transcription by DNA looping. Significant transcriptional changes in the regulatory genes vicR, spxR, comE, comX, and mecA in the sptRSs and sptSSs mutants further indicated that SptRS Ss is part of a regulatory network that coordinates cell wall homeostasis, H2O2 production, and competence. This study reveals that SptRS Ss is involved in the regulation of crucial functions for S. sanguinis persistence in the oral cavity.
Collapse
|
25
|
Pereira D, Seneviratne CJ, Koga-Ito CY, Samaranayake LP. Is the oral fungal pathogen Candida albicans a cariogen? Oral Dis 2017; 24:518-526. [PMID: 28513096 DOI: 10.1111/odi.12691] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 04/13/2017] [Accepted: 04/27/2017] [Indexed: 12/31/2022]
Abstract
Pathobiology of dental caries is complex. Data from recent molecular microbiologic studies have further redefined the role of the oral microbiome in the etiology of dental caries. This new information challenges the conventional view on the hegemony of classic cariogenic prokaryotes such as Streptococcus mutans in caries etiology, and raises the intriguing possibility of the participation of the eukaryotic oral fungal pathogen Candida in the caries process. The virulence attributes of Candida species such as their acidogenicity and aciduric nature, the ability to develop profuse biofilms, ferment and assimilate dietary sugars, and produce collagenolytic proteinases are all indicative of their latent cariogenic potential. Based on the above, oral candidal counts have been used by some as a caries risk indicator. On the contrary, other studies suggest that Candida is merely a passenger extant in an acidic cariogenic milieu, and not a true pathogen. In this review, we critically examine the varying roles of Candida, and traditionally accepted cariogens such as the mutans group of streptococci in the pathobiology of dental caries. The weight of available data tends to imply that Candida may play a pivotal role as a secondary agent perpetuating the carious process, especially in dentinal caries.
Collapse
Affiliation(s)
- Dfa Pereira
- São José dos Campos Institute of Science and Technology, Universidade Estadual Paulista/UNESP, São José dos Campos, Brazil
| | - C J Seneviratne
- Oral Sciences, Faculty of Dentistry, National University of Singapore, Singapore
| | - C Y Koga-Ito
- São José dos Campos Institute of Science and Technology, Universidade Estadual Paulista/UNESP, São José dos Campos, Brazil
| | - L P Samaranayake
- Department of Bioclinical Sciences, Kuwait University, Kuwait.,School of Dentistry, The University of Queensland, Australia
| |
Collapse
|
26
|
Millo G, Juntavee A, Ratanathongkam A, Nualkaew N, Peerapattana J, Chatchiwiwattana S. Antibacterial Inhibitory Effects of Punica Granatum Gel on Cariogenic Bacteria: An in vitro Study. Int J Clin Pediatr Dent 2017; 10:152-157. [PMID: 28890615 PMCID: PMC5571384 DOI: 10.5005/jp-journals-10005-1426] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 09/29/2016] [Indexed: 11/23/2022] Open
Abstract
AIM This study evaluated the in vitro antibacterial effects of the formulated Punica granatum (PG) gel against Streptococcus mutans, Streptococcus sanguinis, and Lactobacillus casei. MATERIALS AND METHODS The PG extract was dissolved in water at 500 mg/mL. High performance liquid chromatography (HPLC) was used for identification and quantification of chemical marker punicalagin. Minimum bactericidal concentration (MBC) and time-kill assay (TKA) were investigated. Antibacterial activities of the formulated PG gel, 2% chlorhexidine (CHX) gel and blank gel were tested by measuring the zones of inhibition through agar well diffusion method. RESULTS The HPLC results showed presence of punicalagin at 2023.58 ± 25.29 pg/mL in the aqueous PG extract and at 0.234% (w/w) in the formulated PG gel. The MBC for S. mutans, S. Sanguinis, and L. casei were 250, 125, and 500 mg/mL respectively. The TKA of 500 mg/mL aqueous PG extract showed total inhibition of S. mutans, S. Sanguinis, and L. casei at 6, 1, and 24 hours contact time respectively. Agar well diffusion revealed that for S. mutans, CHX gel > PG gel > blank gel; for S. sanguinis, CHX gel = PG gel > blank gel; for L. casei, CHX gel > PG gel = blank gel. Comparison of the PG gel potency showed that S. sanguinis = S. mutans > L. casei. CONCLUSION The PG gel equivalent to 0.234% punicalagin (w/w) inhibited S. mutans and S. sanguinis but not L. casei within 24 hours incubation period and has the potential to be used for caries prevention. HOW TO CITE THIS ARTICLE Millo G, Juntavee A, Ratanathongkam A, Nualkaew N, Peerapattana J, Chatchiwiwattana S. Antibacterial Inhibitory Effects of Punica Granatum Gel on Cariogenic Bacteria: An in vitro Study. Int J Clin Pediatr Dent 2017;10(2):152-157.
Collapse
Affiliation(s)
- Grazielle Millo
- Graduate Student, Department of Pediatric Dentistry, Faculty of Dentistry, Khon Kaen University, Khon Kaen, Thailand
| | - Apa Juntavee
- Associate Professor, Department of Pediatric Dentistry, Faculty of Dentistry, Khon Kaen University, Khon Kaen, Thailand
| | - Ariya Ratanathongkam
- Associate Professor, Department of Oral Biology, Faculty of Dentistry, Khon Kaen University, Khon Kaen, Thailand
| | - Natsajee Nualkaew
- Assistant Professor, Department of Pharmacognosy and Toxicity, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen Thailand
| | - Jomjai Peerapattana
- Associate Professor, Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen Thailand
| | - Supaporn Chatchiwiwattana
- Associate Professor, Department of Oral Biology, Faculty of Dentistry, Khon Kaen University, Khon Kaen, Thailand
| |
Collapse
|
27
|
Scoffield JA, Duan D, Zhu F, Wu H. A commensal streptococcus hijacks a Pseudomonas aeruginosa exopolysaccharide to promote biofilm formation. PLoS Pathog 2017; 13:e1006300. [PMID: 28448633 PMCID: PMC5407764 DOI: 10.1371/journal.ppat.1006300] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 03/16/2017] [Indexed: 02/05/2023] Open
Abstract
Pseudomonas aeruginosa causes devastating chronic pulmonary infections in cystic fibrosis (CF) patients. Although the CF airway is inhabited by diverse species of microorganisms interlaced within a biofilm, many studies focus on the sole contribution of P. aeruginosa pathogenesis in CF morbidity. More recently, oral commensal streptococci have been identified as cohabitants of the CF lung, but few studies have explored the role these bacteria play within the CF biofilm. We examined the interaction between P. aeruginosa and oral commensal streptococci within a dual species biofilm. Here we report that the CF P. aeruginosa isolate, FRD1, enhances biofilm formation and colonization of Drosophila melanogaster by the oral commensal Streptococcus parasanguinis. Moreover, production of the P. aeruginosa exopolysaccharide, alginate, is required for the promotion of S. parasanguinis biofilm formation and colonization. However, P. aeruginosa is not promoted in the dual species biofilm. Furthermore, we show that the streptococcal adhesin, BapA1, mediates alginate-dependent enhancement of the S. parasanguinis biofilm in vitro, and BapA1 along with another adhesin, Fap1, are required for the in vivo colonization of S. parasanguinis in the presence of FRD1. Taken together, our study highlights a new association between streptococcal adhesins and P. aeruginosa alginate, and reveals a mechanism by which S. parasanguinis potentially colonizes the CF lung and interferes with the pathogenesis of P. aeruginosa.
Collapse
Affiliation(s)
- Jessica A. Scoffield
- Department of Pediatric Dentistry, School of Dentistry, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Dingyu Duan
- Department of Pediatric Dentistry, School of Dentistry, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- State Key Laboratory of Oral Diseases, Department of Periodontology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Fan Zhu
- Department of Pediatric Dentistry, School of Dentistry, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Hui Wu
- Department of Pediatric Dentistry, School of Dentistry, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- * E-mail:
| |
Collapse
|
28
|
Evolutionary inactivation of a sialidase in group B Streptococcus. Sci Rep 2016; 6:28852. [PMID: 27352769 PMCID: PMC4926279 DOI: 10.1038/srep28852] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 06/10/2016] [Indexed: 11/17/2022] Open
Abstract
Group B Streptococcus (GBS) is a leading cause of bacterial sepsis and meningitis in newborns. GBS possesses a protein with homology to the pneumococcal virulence factor, NanA, which has neuraminidase (sialidase) activity and promotes blood-brain barrier penetration. However, phylogenetic sequence and enzymatic analyses indicate the GBS NanA ortholog has lost sialidase function – and for this distinction we designate the gene and encoded protein nonA/NonA. Here we analyze NonA function in GBS pathogenesis, and through heterologous expression of active pneumococcal NanA in GBS, potential costs of maintaining sialidase function. GBS wild-type and ΔnonA strains lack sialidase activity, but forced expression of pneumococcal NanA in GBS induced degradation of the terminal sialic acid on its exopolysaccharide capsule. Deletion of nonA did not change GBS-whole blood survival or brain microvascular cell invasion. However, forced expression of pneumococcal NanA in GBS removed terminal sialic acid residues from the bacterial capsule, restricting bacterial proliferation in human blood and in vivo upon mouse infection. GBS expressing pneumococcal NanA had increased invasion of human brain microvascular endothelial cells. Thus, we hypothesize that nonA lost enzyme activity allowing the preservation of an effective survival factor, the sialylated exopolysaccharide capsule.
Collapse
|
29
|
Zhuang PL, Yu LX, Tao Y, Zhou Y, Zhi QH, Lin HC. Effects of missense mutations in sortase A gene on enzyme activity in Streptococcus mutans. BMC Oral Health 2016; 16:47. [PMID: 27068451 PMCID: PMC4827206 DOI: 10.1186/s12903-016-0204-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 04/01/2016] [Indexed: 11/21/2022] Open
Abstract
Background Streptococcus mutans (S. mutans) is the major aetiological agent of dental caries, and the transpeptidase Sortase A (SrtA) plays a major role in cariogenicity. The T168G and G470A missense mutations in the srtA gene may be linked to caries susceptibility, as demonstrated in our previous studies. This study aimed to investigate the effects of these missense mutations of the srtA gene on SrtA enzyme activity in S. mutans. Methods The point mutated recombinant S.mutans T168G and G470A sortases were expressed in expression plasmid pET32a. S. mutans UA159 sortase coding gene srtA was used as the template for point mutation. Enzymatic activity was assessed by quantifying increases in the fluorescence intensity generated when a substrate Dabcyl-QALPNTGEE-Edans was cleaved by SrtA. The kinetic constants were calculated based on the curve fit for the Michaelis-Menten equation. Results SrtA△N40(UA159) and the mutant enzymes, SrtA△N40(D56E) and SrtA△N40(R157H), were expressed and purified. A kinetic analysis showed that the affinity of SrtA△N40(D56E) and SrtA△N40(R157H) remained approximately equal to the affinity of SrtA△N40(UA159), as determined by the Michaelis constant (Km). However, the catalytic rate constant (kcat) and catalytic efficiency (kcat/Km) of SrtA△N40(D56E) were reduced compared with those of SrtA△N40(R157H) and SrtA△N40(UA159), whereas the kcat and kcat/Km values of SrtA△N40(R157H) were slightly lower than those of SrtA△N40(UA159). Conclusions The findings of this study indicate that the T168G missense mutation of the srtA gene results in a significant reduction in enzymatic activity compared with S. mutans UA159, suggesting that the T168G missense mutation of the srtA gene may be related to low cariogenicity. Electronic supplementary material The online version of this article (doi:10.1186/s12903-016-0204-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- P L Zhuang
- Department of Preventive Dentistry, Guanghua School of Stomatology, Sun Yat-Sen University, 56 Ling Yuan Road West, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-Sen University, Guangzhou, China.,Department of Stomatology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 107 Yan Jiang Road West, Guangzhou, China
| | - L X Yu
- Department of Preventive Dentistry, Guanghua School of Stomatology, Sun Yat-Sen University, 56 Ling Yuan Road West, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-Sen University, Guangzhou, China
| | - Y Tao
- Department of Preventive Dentistry, Guanghua School of Stomatology, Sun Yat-Sen University, 56 Ling Yuan Road West, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-Sen University, Guangzhou, China
| | - Y Zhou
- Department of Preventive Dentistry, Guanghua School of Stomatology, Sun Yat-Sen University, 56 Ling Yuan Road West, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-Sen University, Guangzhou, China
| | - Q H Zhi
- Department of Preventive Dentistry, Guanghua School of Stomatology, Sun Yat-Sen University, 56 Ling Yuan Road West, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-Sen University, Guangzhou, China
| | - H C Lin
- Department of Preventive Dentistry, Guanghua School of Stomatology, Sun Yat-Sen University, 56 Ling Yuan Road West, Guangzhou, China. .,Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
30
|
Aguayo S, Donos N, Spratt D, Bozec L. Probing the nanoadhesion of Streptococcus sanguinis to titanium implant surfaces by atomic force microscopy. Int J Nanomedicine 2016; 11:1443-50. [PMID: 27103802 PMCID: PMC4827898 DOI: 10.2147/ijn.s100768] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
As titanium (Ti) continues to be utilized in great extent for the fabrication of artificial implants, it is important to understand the crucial bacterium–Ti interaction occurring during the initial phases of biofilm formation. By employing a single-cell force spectroscopy technique, the nanoadhesive interactions between the early-colonizing Streptococcus sanguinis and a clinically analogous smooth Ti substrate were explored. Mean adhesion forces between S. sanguinis and Ti were found to be 0.32±0.00, 1.07±0.06, and 4.85±0.56 nN for 0, 1, and 60 seconds contact times, respectively; while adhesion work values were reported at 19.28±2.38, 104.60±7.02, and 1,317.26±197.69 aJ for 0, 1, and 60 seconds, respectively. At 60 seconds surface delays, minor-rupture events were modeled with the worm-like chain model yielding an average contour length of 668±12 nm. The mean force for S. sanguinis minor-detachment events was 1.84±0.64 nN, and Poisson analysis decoupled this value into a short-range force component of −1.60±0.34 nN and a long-range force component of −0.55±0.47 nN. Furthermore, a solution of 2 mg/mL chlorhexidine was found to increase adhesion between the bacterial probe and substrate. Overall, single-cell force spectroscopy of living S. sanguinis cells proved to be a reliable way to characterize early-bacterial adhesion onto machined Ti implant surfaces at the nanoscale.
Collapse
Affiliation(s)
- Sebastian Aguayo
- Department of Biomaterials and Tissue Engineering, University College London, London, UK
| | - Nikolaos Donos
- Periodontology Unit, University College London, London, UK
| | - Dave Spratt
- Division of Microbial Diseases, UCL Eastman Dental Institute, University College London, London, UK
| | - Laurent Bozec
- Department of Biomaterials and Tissue Engineering, University College London, London, UK
| |
Collapse
|
31
|
Pian Y, Wang P, Liu P, Zheng Y, Zhu L, Wang H, Xu B, Yuan Y, Jiang Y. Proteomics identification of novel fibrinogen-binding proteins of Streptococcus suis contributing to antiphagocytosis. Front Cell Infect Microbiol 2015; 5:19. [PMID: 25789245 PMCID: PMC4349166 DOI: 10.3389/fcimb.2015.00019] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2014] [Accepted: 02/17/2015] [Indexed: 12/12/2022] Open
Abstract
Streptococcus suis serotype 2 (SS2) induced sepsis and meningitis are often accompanied by bacteremia. However, the mechanism whereby it helps S. suis to evade PMN-mediated phagocytosis remain unclear. Because of the central roles of bacteria-human fibrinogen (hFg) interaction in innate immunity, here, a proteomics based Far-western blotting (PBFWB) was developed to identify the fibrinogen-binding surface proteins of S. suis (SsFBPs) on a large-scale. And then thirteen potential SsFBPs were identified by PBFWB and we selected seven potential surface proteins to further confirm their binding ability to hFg, of which the gene mutant strains of MRP displayed significantly decrease in binding to immobilized hFg. Additionally, the polyclonal antibodies against Enolase were found to significantly inhibit the binding of SS2 to hFg. Strikingly, MRP and Enolase were found to improve the antiphagocytic ability of SS2 to PMNs by interacting with hFg and enhance the survival of SS2 in human blood. Taken together, the PBFWB method provides useful clues to the bacteria-host interactions. These studies firstly disclose MRP and Enolase were involved in immune evasion of SS2 at least in part by binding to Fg, which make them potential targets for therapies for SS2 infection.
Collapse
Affiliation(s)
- Yaya Pian
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing Institute of Microbiology and Epidemiology Beijing China
| | - Pingping Wang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing Institute of Microbiology and Epidemiology Beijing China
| | - Peng Liu
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing Institute of Microbiology and Epidemiology Beijing China
| | - Yuling Zheng
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing Institute of Microbiology and Epidemiology Beijing China
| | - Li Zhu
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing Institute of Microbiology and Epidemiology Beijing China
| | - Hengliang Wang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing Institute of Microbiology and Epidemiology Beijing China
| | - Bin Xu
- National Center of Biomedical Analysis, Academy of Military Medical Sciences Beijing, China
| | - Yuan Yuan
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing Institute of Microbiology and Epidemiology Beijing China
| | - Yongqiang Jiang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing Institute of Microbiology and Epidemiology Beijing China
| |
Collapse
|
32
|
Role for the A domain of unprocessed accumulation-associated protein (Aap) in the attachment phase of the Staphylococcus epidermidis biofilm phenotype. J Bacteriol 2014; 196:4268-75. [PMID: 25266380 DOI: 10.1128/jb.01946-14] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
The polysaccharide intercellular adhesin or the cell wall-anchored accumulation-associated protein (Aap) mediates cellular accumulation during Staphylococcus epidermidis biofilm maturation. Mutation of sortase, which anchors up to 11 proteins (including Aap) to the cell wall, blocked biofilm development by the cerebrospinal fluid isolate CSF41498. Aap was implicated in this phenotype when Western blots and two-dimensional (2D) electrophoresis revealed increased levels of the protein in culture supernatants. Unexpectedly, reduced levels of primary attachment were associated with impaired biofilm formation by CSF41498 srtA and aap mutants. In contrast to previous studies, which implicated Aap proteolytic cleavage and, specifically, the Aap B domains in biofilm accumulation, the CSF41498 Aap protein was unprocessed. Furthermore, aap appeared to play a less important role in the biofilm phenotype of S. epidermidis 1457, in which the Aap protein is processed. Anti-Aap A-domain IgG inhibited primary attachment and biofilm formation in strain CSF41498 but not in strain 1457. The nucleotide sequences of the aap gene A-domain region and cleavage site in strains CSF41498 and 1457 were identical, implicating altered protease activity in the differential Aap processing results in the two strains. These data reveal a new role for the A domain of unprocessed Aap in the attachment phase of biofilm formation and suggest that extracellular protease activity can influence whether Aap contributes to the attachment or accumulation phases of the S. epidermidis biofilm phenotype.
Collapse
|
33
|
Liu P, Pian Y, Li X, Liu R, Xie W, Zhang C, Zheng Y, Jiang Y, Yuan Y. Streptococcus suis adenosine synthase functions as an effector in evasion of PMN-mediated innate immunit. J Infect Dis 2014; 210:35-45. [PMID: 24446521 DOI: 10.1093/infdis/jiu050] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Streptococcus suis serotype 2 (S. suis 2) is a highly invasive pathogen in pigs and humans that can cause severe systemic infection. Sepsis and meningitis are the most common clinical manifestations of S. suis 2 infection. However, the mechanisms of S. suis 2 surviving in human blood remains unclear, so to identify novel virulence factors in evasion of polymorphonuclear leukocyte (PMN)-mediated innate immunity play important roles in developing therapies against S. suis 2 infection. Here, we found that S. suis 2 can escape phagocytic clearance by adenosine synthesis in blood. Through bioinformatics-based analyses we identified a cell wall-anchored protein harbors a 5′-nucleotidase signature sequence and evidence strongly indicated that it can convert adenosine monophosphate (AMP) to adenosine. It was designated as Ssads (the adenosine synthase of S. suis 2). Furthermore, we found that Ssads could impair PMN's defense against S. suis 2 with decreasing of oxidative activity and degranulation of PMNs in human blood via A₂a receptors. Additionally, this enzyme-deficient mutant was found to have diminished virulence in the piglet infection model. Taken together, these results indicate that Ssads play an important role in S. suis 2 escaping human innate immunity in the context of inhibiting PMN's activity by synthesis of adenosine.
Collapse
|
34
|
Yamaguchi M, Terao Y, Mori-Yamaguchi Y, Domon H, Sakaue Y, Yagi T, Nishino K, Yamaguchi A, Nizet V, Kawabata S. Streptococcus pneumoniae invades erythrocytes and utilizes them to evade human innate immunity. PLoS One 2013; 8:e77282. [PMID: 24194877 PMCID: PMC3806730 DOI: 10.1371/journal.pone.0077282] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Accepted: 08/31/2013] [Indexed: 11/18/2022] Open
Abstract
Streptococcus pneumoniae, a Gram-positive bacterium, is a major cause of invasive infection-related diseases such as pneumonia and sepsis. In blood, erythrocytes are considered to be an important factor for bacterial growth, as they contain abundant nutrients. However, the relationship between S. pneumoniae and erythrocytes remains unclear. We analyzed interactions between S. pneumoniae and erythrocytes, and found that iron ion present in human erythrocytes supported the growth of Staphylococcus aureus, another major Gram-positive sepsis pathogen, while it partially inhibited pneumococcal growth by generating free radicals. S. pneumoniae cells incubated with human erythrocytes or blood were subjected to scanning electron and confocal fluorescence microscopic analyses, which showed that the bacterial cells adhered to and invaded human erythrocytes. In addition, S. pneumoniae cells were found associated with human erythrocytes in cultures of blood from patients with an invasive pneumococcal infection. Erythrocyte invasion assays indicated that LPXTG motif-containing pneumococcal proteins, erythrocyte lipid rafts, and erythrocyte actin remodeling are all involved in the invasion mechanism. In a neutrophil killing assay, the viability of S. pneumoniae co-incubated with erythrocytes was higher than that without erythrocytes. Also, H2O2 killing of S. pneumoniae was nearly completely ineffective in the presence of erythrocytes. These results indicate that even when S. pneumoniae organisms are partially killed by iron ion-induced free radicals, they can still invade erythrocytes. Furthermore, in the presence of erythrocytes, S. pneumoniae can more effectively evade antibiotics, neutrophil phagocytosis, and H2O2 killing.
Collapse
Affiliation(s)
- Masaya Yamaguchi
- Department of Cell Membrane Biology, Institute of Scientific and Industrial Research, Osaka University, Ibaraki, Osaka, Japan
- Department of Oral and Molecular Microbiology, Osaka University Graduate School of Dentistry, Suita, Osaka, Japan
- Department of Pediatrics, University of California San Diego, La Jolla, California, United States of America
| | - Yutaka Terao
- Department of Oral and Molecular Microbiology, Osaka University Graduate School of Dentistry, Suita, Osaka, Japan
- Division of Microbiology and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, Chuo-ku, Niigata, Japan
- * E-mail:
| | - Yuka Mori-Yamaguchi
- Department of Pediatrics, University of California San Diego, La Jolla, California, United States of America
| | - Hisanori Domon
- Division of Microbiology and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, Chuo-ku, Niigata, Japan
| | - Yuuki Sakaue
- Division of Microbiology and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, Chuo-ku, Niigata, Japan
| | - Tetsuya Yagi
- Center of National University Hospital for Infection Control, Nagoya University Hospital, Nagoya, Aichi, Japan
| | - Kunihiko Nishino
- Department of Cell Membrane Biology, Institute of Scientific and Industrial Research, Osaka University, Ibaraki, Osaka, Japan
| | - Akihito Yamaguchi
- Department of Cell Membrane Biology, Institute of Scientific and Industrial Research, Osaka University, Ibaraki, Osaka, Japan
| | - Victor Nizet
- Department of Pediatrics, University of California San Diego, La Jolla, California, United States of America
| | - Shigetada Kawabata
- Department of Oral and Molecular Microbiology, Osaka University Graduate School of Dentistry, Suita, Osaka, Japan
| |
Collapse
|
35
|
Simón-Soro A, Belda-Ferre P, Cabrera-Rubio R, Alcaraz LD, Mira A. A tissue-dependent hypothesis of dental caries. Caries Res 2013; 47:591-600. [PMID: 24080530 DOI: 10.1159/000351663] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Accepted: 04/18/2013] [Indexed: 11/19/2022] Open
Abstract
Current understanding of dental caries considers this disease a demineralization of the tooth tissues due to the acid produced by sugar-fermenting microorganisms. Thus, caries is considered a diet- and pH-dependent process. We present here the first metagenomic analysis of the bacterial communities present at different stages of caries development, with the aim of determining whether the bacterial composition and biochemical profile are specific to the tissue affected. The data show that microbial composition at the initial, enamel-affecting stage of caries is significantly different from that found at subsequent stages, as well as from dental plaque of sound tooth surfaces. Although the relative proportion of Streptococcus mutans increased from 0.12% in dental plaque to 0.72% in enamel caries, Streptococcus mitis and Streptococcus sanguinis were the dominant streptococci in these lesions. The functional profile of caries-associated bacterial communities indicates that genes involved in acid stress tolerance and dietary sugar fermentation are overrepresented only at the initial stage (enamel caries), whereas other genes coding for osmotic stress tolerance as well as collagenases and other proteases enabling dentin degradation are significantly overrepresented in dentin cavities. The results support a scenario in which pH and diet are determinants of the disease during the degradation of enamel, but in dentin caries lesions not only acidogenic but also proteolytic bacteria are involved. We propose that caries disease is a process of varying etiology, in which acid-producing bacteria are the vehicle to penetrate enamel and allow dentin degrading microorganisms to expand the cavity.
Collapse
Affiliation(s)
- A Simón-Soro
- Department of Genomics and Health, Center for Advanced Research in Public Health, Valencia, Spain
| | | | | | | | | |
Collapse
|
36
|
Jin D, Chen C, Li L, Lu S, Li Z, Zhou Z, Jing H, Xu Y, Du P, Wang H, Xiong Y, Zheng H, Bai X, Sun H, Wang L, Ye C, Gottschalk M, Xu J. Dynamics of fecal microbial communities in children with diarrhea of unknown etiology and genomic analysis of associated Streptococcus lutetiensis. BMC Microbiol 2013; 13:141. [PMID: 23782707 PMCID: PMC3716944 DOI: 10.1186/1471-2180-13-141] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Accepted: 06/10/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The sequences of the 16S rRNA genes extracted from fecal samples provide insights into the dynamics of fecal microflora. This potentially gives valuable etiological information for patients whose conditions have been ascribed to unknown pathogens, which cannot be accomplished using routine culture methods. We studied 33 children with diarrhea who were admitted to the Children's Hospital in Shanxi Province during 2006. RESULTS Nineteen of 33 children with diarrhea could not be etiologically diagnosed by routine culture and polymerase chain reaction methods. Eleven of 19 children with diarrhea of unknown etiology had Streptococcus as the most dominant fecal bacterial genus at admission. Eight of nine children whom three consecutive fecal samples were collected had Streptococcus as the dominant fecal bacterial genus, including three in the Streptococcus bovis group and three Streptococcus sp., which was reduced during and after recovery. We isolated strains that were possibly from the S. bovis group from feces sampled at admission, which were then identified as Streptococcus lutetiensis from one child and Streptococcus gallolyticus subsp. pasteurianus from two children. We sequenced the genome of S. lutetiensis and identified five antibiotic islands, two pathogenicity islands, and five unique genomic islands. The identified virulence genes included hemolytic toxin cylZ of Streptococcus agalactiae and sortase associated with colonization of pathogenic streptococci. CONCLUSIONS We identified S. lutetiensis and S. gallolyticus subsp. pasteurianus from children with diarrhea of unknown etiology, and found pathogenic islands and virulence genes in the genome of S. lutetiensis.
Collapse
Affiliation(s)
- Dong Jin
- State Key Laboratory for Infectious Disease Prevention and Control, and National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Cole JN, Henningham A, Gillen CM, Ramachandran V, Walker MJ. Human pathogenic streptococcal proteomics and vaccine development. Proteomics Clin Appl 2012; 2:387-410. [PMID: 21136841 DOI: 10.1002/prca.200780048] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Gram-positive streptococci are non-motile, chain-forming bacteria commonly found in the normal oral and bowel flora of warm-blooded animals. Over the past decade, a proteomic approach combining 2-DE and MS has been used to systematically map the cellular, surface-associated and secreted proteins of human pathogenic streptococcal species. The public availability of complete streptococcal genomic sequences and the amalgamation of proteomic, genomic and bioinformatic technologies have recently facilitated the identification of novel streptococcal vaccine candidate antigens and therapeutic agents. The objective of this review is to examine the constituents of the streptococcal cell wall and secreted proteome, the mechanisms of transport of surface and secreted proteins, and describe the current methodologies employed for the identification of novel surface-displayed proteins and potential vaccine antigens.
Collapse
Affiliation(s)
- Jason N Cole
- School of Biological Sciences, University of Wollongong, Wollongong, New South Wales, Australia
| | | | | | | | | |
Collapse
|
38
|
Fhb, a novel factor H-binding surface protein, contributes to the antiphagocytic ability and virulence of Streptococcus suis. Infect Immun 2012; 80:2402-13. [PMID: 22526676 DOI: 10.1128/iai.06294-11] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Streptococcus suis serotype 2 is a Gram-positive bacterium that causes sepsis and meningitis in piglets and humans. The mechanisms of S. suis serotype 2 invasive disease are not well understood. The surface proteins of pathogens usually play important roles in infection and bacterium-host interactions. Here, we identified a novel surface protein that contributed significantly to the virulence of S. suis serotype 2 in a piglet infection model. This protein showed little similarity to other reported proteins and exhibited strong binding activity to human factor H (hFH). It was designated Fhb (factor H-binding protein). The fhb genes found in S. suis serotypes 1, 2, 4, 7, and 9 exhibited molecular polymorphism. Fhb possessed two proline-rich repeat sequences and XPZ domains, and one repeat sequence exhibited a high homology to Bac, an IgA-binding protein of Streptococcus agalactiae. Evidence strongly indicated that fhb-deficient mutants had diminished phagocytosis resistance in bactericidal assays. In addition, Fhb plays important roles in complement-mediated immunity by interacting with hFH. These findings indicated that Fhb is a crucial surface protein contributing to the virulence of S. suis, with important functions in evading innate immune defenses by interaction with host complement regulatory factor hFH.
Collapse
|
39
|
Chang OK, Perrin C, Galia W, Saulnier F, Miclo L, Roux E, Driou A, Humbert G, Dary A. Release of the cell-envelope protease PrtS in the growth medium of Streptococcus thermophilus 4F44. Int Dairy J 2012. [DOI: 10.1016/j.idairyj.2011.10.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
40
|
Abstract
Bacterial infection associated with the use of medical or dental devices is a serious concern. Although devices made of ethylene vinyl acetate (EVA) are often used in the oral cavity, there are no established standards for their storage. We investigated bacterial survival on EVA sheets under various storage conditions to establish a standard for hygienic storage of such dental devices. Bacterial counts were evaluated, which showed a significant decrease after washing with sterilized water, mechanical brushing and rinsing, and using Mouthguard Cleaner as compared to untreated samples. In addition, no bacteria were detected on samples stored 2 days or longer in a ventilated environment, whereas they were detected for up to 14 days on samples without any cleaning stored in a closed environment. Bacterial counts for the untreated samples gradually declined, while surviving bacteria on samples treated with sterilized water and mechanical brushing showed a rapid decrease. Additionally, bacterial identification using polymerase chain reaction (PCR) revealed that Streptococcus oralis was dominantly detected on salivary samples after 14 days of storage among both two subjects. For effective hygienic storage of dental devices made of EVA, washing with sterilized water is important to remove absorbed salivary compounds along with storage in a ventilated environment.
Collapse
|
41
|
Zhang XH, Zhou Y, Zhi QH, Tao Y, Lin HC. Genetic polymorphisms of the sortase A gene and early childhood caries in two-year-old children. Arch Oral Biol 2012; 57:948-53. [PMID: 22381535 DOI: 10.1016/j.archoralbio.2012.02.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2011] [Revised: 01/09/2012] [Accepted: 02/01/2012] [Indexed: 10/28/2022]
Abstract
OBJECTIVE To explore and compare the genetic polymorphisms of the sortase A (srtA) gene found in Streptococcus mutans (S. mutans) infecting two-year-old children suffering early childhood caries to those found in caries-free children through molecular identification methods. METHODS Clinical S. mutans strains were isolated from the dental plaques of two-year-old children. Fifteen strains of S. mutans from the caries-active group and 15 strains of S. mutans from the caries-free group were collected. Genomic DNA was extracted from the S. mutans isolates. DNA fragments, including the srtA gene, were amplified by PCR. The PCR products were purified, sequenced and analyzed. A chi-square test and BioEdit software were used to analyze the sequencing results. RESULTS All 30 clinically isolated S. mutans strains had a 741 base pair (bp) srtA gene. There were no nucleotide sequence insertions or deletions observed in the srtA genes. Twenty mutations were identified in the srtA genes that taken from the 30 clinical strains. There were 10 silent point mutations at the 78, 99, 150, 165, 186, 222, 249, 261, 312, and 636bp positions. The other 10 mutations were point mutations resulting in a missense mutation at the 23, 34, 36, 47, 112, 114, 168, 176, 470, and 671bp positions. None of the positions were enzyme-activity sites of srt A. The missense mutation rates of the two groups did not exhibit statistically significant differences. CONCLUSION There were no genetic polymorphisms of the sortase A gene associated with early childhood caries in two-year-old children.
Collapse
Affiliation(s)
- X H Zhang
- Department of Preventive Dentistry, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | | | | | | | | |
Collapse
|
42
|
Zheng L, Chen Z, Itzek A, Herzberg MC, Kreth J. CcpA regulates biofilm formation and competence in Streptococcus gordonii. Mol Oral Microbiol 2011; 27:83-94. [PMID: 22394467 DOI: 10.1111/j.2041-1014.2011.00633.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Streptococcus gordonii is an important member of the oral biofilm community. As an oral commensal streptococcus, S. gordonii is considered beneficial in promoting biofilm homeostasis. CcpA is known as the central regulator of carbon catabolite repression in Gram-positive bacteria and is also involved in the control of virulence gene expression. To further establish the role of CcpA as central regulator in S. gordonii, the effect of CcpA on biofilm formation and natural competence of S. gordonii was investigated. These phenotypic traits have been suggested to be important to oral streptococci in coping with environmental stress. Here we demonstrate that a CcpA mutant was severely impaired in its biofilm-forming ability, showed a defect in extracellular polysaccharide production and reduced competence. The data suggest that CcpA is involved in the regulation of biofilm formation and competence development in S. gordonii.
Collapse
Affiliation(s)
- L Zheng
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | | | | | | | | |
Collapse
|
43
|
Goh YJ, Goin C, O'Flaherty S, Altermann E, Hutkins R. Specialized adaptation of a lactic acid bacterium to the milk environment: the comparative genomics of Streptococcus thermophilus LMD-9. Microb Cell Fact 2011; 10 Suppl 1:S22. [PMID: 21995282 PMCID: PMC3231929 DOI: 10.1186/1475-2859-10-s1-s22] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Background Streptococcus thermophilus represents the only species among the streptococci that has “Generally Regarded As Safe” status and that plays an economically important role in the fermentation of yogurt and cheeses. We conducted comparative genome analysis of S. thermophilus LMD-9 to identify unique gene features as well as features that contribute to its adaptation to the dairy environment. In addition, we investigated the transcriptome response of LMD-9 during growth in milk in the presence of Lactobacillus delbrueckii ssp. bulgaricus, a companion culture in yogurt fermentation, and during lytic bacteriophage infection. Results The S. thermophilus LMD-9 genome is comprised of a 1.8 Mbp circular chromosome (39.1% GC; 1,834 predicted open reading frames) and two small cryptic plasmids. Genome comparison with the previously sequenced LMG 18311 and CNRZ1066 strains revealed 114 kb of LMD-9 specific chromosomal region, including genes that encode for histidine biosynthetic pathway, a cell surface proteinase, various host defense mechanisms and a phage remnant. Interestingly, also unique to LMD-9 are genes encoding for a putative mucus-binding protein, a peptide transporter, and exopolysaccharide biosynthetic proteins that have close orthologs in human intestinal microorganisms. LMD-9 harbors a large number of pseudogenes (13% of ORFeome), indicating that like LMG 18311 and CNRZ1066, LMD-9 has also undergone major reductive evolution, with the loss of carbohydrate metabolic genes and virulence genes found in their streptococcal counterparts. Functional genome distribution analysis of ORFeomes among streptococci showed that all three S. thermophilus strains formed a distinct functional cluster, further establishing their specialized adaptation to the nutrient-rich milk niche. An upregulation of CRISPR1 expression in LMD-9 during lytic bacteriophage DT1 infection suggests its protective role against phage invasion. When co-cultured with L. bulgaricus, LMD-9 overexpressed genes involved in amino acid transport and metabolism as well as DNA replication. Conclusions The genome of S. thermophilus LMD-9 is shaped by its domestication in the dairy environment, with gene features that conferred rapid growth in milk, stress response mechanisms and host defense systems that are relevant to its industrial applications. The presence of a unique exopolysaccharide gene cluster and cell surface protein orthologs commonly associated with probiotic functionality revealed potential probiotic applications of LMD-9.
Collapse
Affiliation(s)
- Yong Jun Goh
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, Nebraska 68583, USA
| | | | | | | | | |
Collapse
|
44
|
Okahashi N, Nakata M, Terao Y, Isoda R, Sakurai A, Sumitomo T, Yamaguchi M, Kimura RK, Oiki E, Kawabata S, Ooshima T. Pili of oral Streptococcus sanguinis bind to salivary amylase and promote the biofilm formation. Microb Pathog 2011; 50:148-54. [DOI: 10.1016/j.micpath.2011.01.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2010] [Revised: 01/04/2011] [Accepted: 01/06/2011] [Indexed: 10/18/2022]
|
45
|
Sharma A, Nigam A. Structure modeling of novel DNA glycosylase enzyme from oral pathogen Streptococcus sanguinis. Bioinformation 2010; 5:136-40. [PMID: 21364794 PMCID: PMC3040489 DOI: 10.6026/97320630005136] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2010] [Accepted: 08/26/2010] [Indexed: 11/23/2022] Open
Abstract
The novel 3-methyladenine DNA glycosylase enzyme from oral pathogen Streptococcus sanguinisin involves in DNA repair mechanisms and participates in base excision repair. Its 3D structure is still unknown which may be a potential drug target, therefore here we proposed its putative 3D structure by homology modeling approach. EsyPred3d software produced more precise modeled structure as compare to Swiss model software. The modeled structure was further verified by PROCHECK analysis and subjected to functional site prediction servers for active site residues prediction. The functional site was further validated by molecular docking approach with ligand EDA (3- [2- Deoxyribofuranosyl] - 3H- 1, 3, 4, 5A, 8-Pentaaza- Asindacene-5- monophosphate) from 1F4R. The EDR docked at the cavity of modeled structure of 3-methyladenine DNA glycosylase enzyme with highest Patchdock score of 3966 and lowest Autodock 4 docking energy of -10.30 Kcal/mol. The YA51, LA105, RA107 residues are surrounding the EDA and matching with ligand binding residues predicted by PROFUNC server.
Collapse
Affiliation(s)
- Ashwani Sharma
- Dept. of Bioscience and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai-400076 Maharashtra, India
| | - Anshul Nigam
- Dept. of Bioscience and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai-400076 Maharashtra, India
| |
Collapse
|
46
|
Leigh JA, Egan SA, Ward PN, Field TR, Coffey TJ. Sortase anchored proteins of Streptococcus uberis play major roles in the pathogenesis of bovine mastitis in dairy cattle. Vet Res 2010; 41:63. [PMID: 20519112 PMCID: PMC2898060 DOI: 10.1051/vetres/2010036] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2010] [Accepted: 06/02/2010] [Indexed: 12/22/2022] Open
Abstract
Streptococcus uberis, strain 0140J, contains a single copy sortase A (srtA), encoding a transamidase capable of covalently anchoring specific proteins to peptidoglycan. Unlike the wild-type, an isogenic mutant carrying an inactivating ISS1 insertion within srtA was only able to infect the bovine mammary gland in a transient fashion. For the first 24 h post challenge, the srtA mutant colonised at a similar rate and number to the wild type strain, but unlike the wild type did not subsequently colonise in higher numbers. Similar levels of host cell infiltration were detected in response to infection with both strains, but only in those mammary quarters infected with the wild type strain were clinical signs of disease evident. Mutants that failed to express individual sortase substrate proteins (sub0135, sub0145, sub0207, sub0241, sub0826, sub0888, sub1095, sub1154, sub1370, and sub1730) were isolated and their virulence determined in the same challenge model. This revealed that mutants lacking sub0145, sub1095 and sub1154 were attenuated in cattle. These data demonstrate that a number of sortase anchored proteins each play a distinct, non-redundant and important role in pathogenesis of S. uberis infection within the lactating bovine mammary gland.
Collapse
Affiliation(s)
- James A Leigh
- The School of Veterinary Medicine and Science, The University of Nottingham, Sutton Bonington Campus, Sutton Bonington, Leicestershire, LE12 5RD, United Kindgom.
| | | | | | | | | |
Collapse
|
47
|
Okahashi N, Nakata M, Sakurai A, Terao Y, Hoshino T, Yamaguchi M, Isoda R, Sumitomo T, Nakano K, Kawabata S, Ooshima T. Pili of oral Streptococcus sanguinis bind to fibronectin and contribute to cell adhesion. Biochem Biophys Res Commun 2009; 391:1192-6. [PMID: 20004645 DOI: 10.1016/j.bbrc.2009.12.029] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2009] [Accepted: 12/05/2009] [Indexed: 12/26/2022]
Abstract
Streptococcus sanguinis is a predominant bacterium in the human oral cavity and occasionally causes infective endocarditis. We identified a unique cell surface polymeric structure named pili in this species and investigated its functions in regard to its potential virulence. Pili of S. sanguinis strain SK36 were shown to be composed of three distinctive pilus proteins (PilA, PilB, and PilC), and a pili-deficient mutant demonstrated reduced bacterial adherence to HeLa and human oral epithelial cells. PilC showed a binding ability to fibronectin, suggesting that pili are involved in colonization by this species. In addition, ATCC10556, a standard S. sanguinis strain, was unable to produce pili due to defective pilus genes, which indicates a diversity of pilus expression among various S. sanguinis strains.
Collapse
Affiliation(s)
- Nobuo Okahashi
- Department of Oral Frontier Biology, Graduate School of Dentistry, Osaka University, 1-8 Yamadaoka, Suita-Osaka 565-0871, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Nrc of Streptococcus pneumoniae suppresses capsule expression and enhances anti-phagocytosis. Biochem Biophys Res Commun 2009; 390:155-60. [PMID: 19799870 DOI: 10.1016/j.bbrc.2009.09.099] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2009] [Accepted: 09/24/2009] [Indexed: 11/22/2022]
Abstract
Streptococcus pneumoniae is a major pathogen of community-acquired pneumonia and one of its major virulence factors is pneumolysin, which functions as a cholesterol-dependent cytolytic pore-forming toxin. In this study, we identified the ply-like gene spd0729 in a BLAST search. Unexpectedly, hemolytic and cytotoxic assays showed no significant differences between a Deltaspd0729 mutant strain and the wild-type strain, whereas the mutant strain exhibited weaker anti-phagocytic activity in human peripheral blood. In addition, real-time RT-PCR analysis revealed that four capsular biosynthesis genes in the mutant strain had expressions 7- to 432-fold greater than those of the wild type, while an enzyme-linked immunoassay showed a mean 3-fold greater amount of total capsular polysaccharide in the mutant strain. These results suggest that Spd0729 is not a cytolysin, though it plays crucial roles in anti-phagocytosis and regulation of capsule expression. Thus, we named Spd0729 as a negative regulator of capsular polysaccharide synthesis (Nrc).
Collapse
|
49
|
Comprehensive evaluation of Streptococcus sanguinis cell wall-anchored proteins in early infective endocarditis. Infect Immun 2009; 77:4966-75. [PMID: 19703977 DOI: 10.1128/iai.00760-09] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Streptococcus sanguinis is a member of the viridans group of streptococci and a leading cause of the life-threatening endovascular disease infective endocarditis. Initial contact with the cardiac infection site is likely mediated by S. sanguinis surface proteins. In an attempt to identify the proteins required for this crucial step in pathogenesis, we searched for surface-exposed, cell wall-anchored proteins encoded by S. sanguinis and then used a targeted signature-tagged mutagenesis (STM) approach to evaluate their contributions to virulence. Thirty-three predicted cell wall-anchored proteins were identified-a number much larger than those found in related species. The requirement of each cell wall-anchored protein for infective endocarditis was assessed in the rabbit model. It was found that no single cell wall-anchored protein was essential for the development of early infective endocarditis. STM screening was also employed for the evaluation of three predicted sortase transpeptidase enzymes, which mediate the cell surface presentation of cell wall-anchored proteins. The sortase A mutant exhibited a modest (approximately 2-fold) reduction in competitiveness, while the other two sortase mutants were indistinguishable from the parental strain. The combined results suggest that while cell wall-anchored proteins may play a role in S. sanguinis infective endocarditis, strategies designed to interfere with individual cell wall-anchored proteins or sortases would not be effective for disease prevention.
Collapse
|
50
|
Contribution of autolysin and Sortase a during Enterococcus faecalis DNA-dependent biofilm development. Infect Immun 2009; 77:3626-38. [PMID: 19528211 DOI: 10.1128/iai.00219-09] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Biofilm production is a major attribute of Enterococcus faecalis clinical isolates. Although some factors, such as sortases, autolysin, and extracellular DNA (eDNA), have been associated with E. faecalis biofilm production, the mechanisms underlying the contributions of these factors to this process have not been completely elucidated yet. In this study we define important roles for the major E. faecalis autolysin (Atn), eDNA, and sortase A (SrtA) during the developmental stages of biofilm formation under static and hydrodynamic conditions. Deletion of srtA affects the attachment stage and results in a deficiency in biofilm production. Atn-deficient mutants are delayed in biofilm development due to defects in primary adherence and DNA release, which we show to be particularly important during the accumulative phase for maturation and architectural stability of biofilms. Confocal laser scanning and freeze-dry electron microscopy of biofilms grown under hydrodynamic conditions revealed that E. faecalis produces a DNase I-sensitive fibrous network, which is important for biofilm stability and is absent in atn-deficient mutant biofilms. This study establishes the stage-specific requirements for SrtA and Atn and demonstrates a role for Atn in the pathway leading to DNA release during biofilm development in E. faecalis.
Collapse
|