1
|
Liu P, Wang L, Song Y, Pei H, Cao X. Virtual Screening of Inhibitors of Streptococcus mutans Biofilm from Lonicera japonica flos and Activity Validation. ACS Med Chem Lett 2024; 15:781-790. [PMID: 38894900 PMCID: PMC11181501 DOI: 10.1021/acsmedchemlett.4c00051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 05/07/2024] [Accepted: 05/08/2024] [Indexed: 06/21/2024] Open
Abstract
In this study, potential inhibitors of Streptococcus mutans biofilm were screened from Lonicera japonica flos using semiflexible molecular docking. A total of 88 metabolites from L. japonica flos and 14 biofilm-related proteins of S. mutans were analyzed, and 25 compounds were initially screened out. Subsequently, 9 compounds with higher availability were subjected to experimental validation, confirming that 6 of them effectively inhibit the S. mutans biofilm formation. Notably, chlorogenic acid was found to potentially disrupt the GbpC protein, which plays a role in the sucrose-dependent adhesion pathway. Similarly, oleanolic acid appeared to impede the adhesin P1 protein involved in the sucrose-independent adhesion mechanism, corroborating the computational predictions. The results of this study provide essential insights for leveraging L. japonica flos in the creation of dental-care-related products and food items aimed at oral health.
Collapse
Affiliation(s)
- Ping Liu
- Beijing Advanced
Innovation
Center for Food Nutrition and Human Health, Beijing Technology and Business University, Beijing 100048, China
| | - Lin Wang
- Beijing Advanced
Innovation
Center for Food Nutrition and Human Health, Beijing Technology and Business University, Beijing 100048, China
| | - Ya Song
- Beijing Advanced
Innovation
Center for Food Nutrition and Human Health, Beijing Technology and Business University, Beijing 100048, China
| | - Hairun Pei
- Beijing Advanced
Innovation
Center for Food Nutrition and Human Health, Beijing Technology and Business University, Beijing 100048, China
| | - Xueli Cao
- Beijing Advanced
Innovation
Center for Food Nutrition and Human Health, Beijing Technology and Business University, Beijing 100048, China
| |
Collapse
|
2
|
Chen Y, Cui G, Cui Y, Chen D, Lin H. Small molecule targeting amyloid fibrils inhibits Streptococcus mutans biofilm formation. AMB Express 2021; 11:171. [PMID: 34919191 PMCID: PMC8683520 DOI: 10.1186/s13568-021-01333-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Accepted: 12/11/2021] [Indexed: 11/10/2022] Open
Abstract
Amyloid fibrils are important scaffold in bacterial biofilms. Streptococcus mutans is an established cariogenic bacteria dwelling within biofilms, and C123 segment of P1 protein is known to form amyloid fibrils in S. mutans biofilms, among which C3 segment could serve as a promising anti-amyloid target due to its critical role in C123-P1 interactions. Recently, small molecules have been found to successfully inhibit biofilms by targeting amyloid fibrils. Thus, our study aimed to screen small molecules targeting C3 segment with the capacity to influence amyloid fibrils and S. mutans biofilms. In silico screening was utilized to discover promising small molecules, which were evaluated for their effects on bacterial cells and amyloid fibrils. We selected 99 small molecules and enrolled 55 small molecules named D1-D55 for crystal violet staining. Notably, D25 selectively inhibit S. mutans biofilms but had no significant influence on biofilms formed by Streptococcus gordonii and Streptococcus sanguinis, and D25 showed no bactericidal effects and low cytotoxicity. In addition, amyloid fibrils in free-floating bacteria, biofilms and purified C123 were quantified with ThT assays, and the differences were not statistically significant in the presence or absence of D25. Morphological changes of amyloid fibrils were visualized with TEM images, where amorphous aggregates were obvious coupled with long and atypical amyloid fibrils. Moreover, amyloid-related genes were upregulated in response to D25. In conclusion, D25 is a promising antimicrobial agent with the capacity to influence amyloid fibrils and inhibit S. mutans biofilms.
Collapse
Affiliation(s)
- Yuanyuan Chen
- Department of Preventive Dentistry, Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, Guangdong China
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, Guangdong China
| | - Guxin Cui
- Department of Preventive Dentistry, Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, Guangdong China
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, Guangdong China
| | - Yuqi Cui
- Department of Preventive Dentistry, Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, Guangdong China
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, Guangdong China
| | - Dongru Chen
- Department of Orthodontics, Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, Guangdong China
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, Guangdong China
| | - Huancai Lin
- Department of Preventive Dentistry, Hospital of Stomatology, Sun Yat-Sen University, Guangzhou, Guangdong China
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, Guangdong China
| |
Collapse
|
3
|
In Silico Selection and In Vitro Evaluation of New Molecules That Inhibit the Adhesion of Streptococcus mutants through Antigen I/II. Int J Mol Sci 2020; 22:ijms22010377. [PMID: 33396525 PMCID: PMC7795114 DOI: 10.3390/ijms22010377] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 11/26/2020] [Accepted: 12/03/2020] [Indexed: 11/23/2022] Open
Abstract
Streptococcus mutans is the main early colonizing cariogenic bacteria because it recognizes salivary pellicle receptors. The Antigen I/II (Ag I/II) of S. mutans is among the most important adhesins in this process, and is involved in the adhesion to the tooth surface and the bacterial co-aggregation in the early stage of biofilm formation. However, this protein has not been used as a target in a virtual strategy search for inhibitors. Based on the predicted binding affinities, drug-like properties and toxicity, molecules were selected and evaluated for their ability to reduce S. mutans adhesion. A virtual screening of 883,551 molecules was conducted; cytotoxicity analysis on fibroblast cells, S. mutans adhesion studies, scanning electron microscopy analysis for bacterial integrity and molecular dynamics simulation were also performed. We found three molecules ZINC19835187 (ZI-187), ZINC19924939 (ZI-939) and ZINC19924906 (ZI-906) without cytotoxic activity, which inhibited about 90% the adhesion of S. mutans to polystyrene microplates. Molecular dynamic simulation by 300 nanoseconds showed stability of the interaction between ZI-187 and Ag I/II (PDB: 3IPK). This work provides new molecules that targets Ag I/II and have the capacity to inhibit in vitro the S. mutans adhesion on polystyrene microplates.
Collapse
|
4
|
Multifunctional Amyloids in the Biology of Gram-Positive Bacteria. Microorganisms 2020; 8:microorganisms8122020. [PMID: 33348645 PMCID: PMC7766987 DOI: 10.3390/microorganisms8122020] [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] [Received: 11/26/2020] [Revised: 12/11/2020] [Accepted: 12/14/2020] [Indexed: 01/18/2023] Open
Abstract
Since they were discovered, amyloids have proven to be versatile proteins able to participate in a variety of cellular functions across all kingdoms of life. This multitask trait seems to reside in their ability to coexist as monomers, aggregates or fibrillar entities, with morphological and biochemical peculiarities. It is precisely this common molecular behaviour that allows amyloids to cross react with one another, triggering heterologous aggregation. In bacteria, many of these functional amyloids are devoted to the assembly of biofilms by organizing the matrix scaffold that keeps cells together. However, consistent with their notion of multifunctional proteins, functional amyloids participate in other biological roles within the same organisms, and emerging unprecedented functions are being discovered. In this review, we focus on functional amyloids reported in gram-positive bacteria, which are diverse in their assembly mechanisms and remarkably specific in their biological functions that they perform. Finally, we consider cross-seeding between functional amyloids as an emerging theme in interspecies interactions that contributes to the diversification of bacterial biology.
Collapse
|
5
|
Järvå MA, Hirt H, Dunny GM, Berntsson RPA. Polymer Adhesin Domains in Gram-Positive Cell Surface Proteins. Front Microbiol 2020; 11:599899. [PMID: 33324381 PMCID: PMC7726212 DOI: 10.3389/fmicb.2020.599899] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 10/28/2020] [Indexed: 01/12/2023] Open
Abstract
Surface proteins in Gram-positive bacteria are often involved in biofilm formation, host-cell interactions, and surface attachment. Here we review a protein module found in surface proteins that are often encoded on various mobile genetic elements like conjugative plasmids. This module binds to different types of polymers like DNA, lipoteichoic acid and glucans, and is here termed polymer adhesin domain. We analyze all proteins that contain a polymer adhesin domain and classify the proteins into distinct classes based on phylogenetic and protein domain analysis. Protein function and ligand binding show class specificity, information that will be useful in determining the function of the large number of so far uncharacterized proteins containing a polymer adhesin domain.
Collapse
Affiliation(s)
- Michael A Järvå
- Department of Medical Biochemistry and Biophysics, Umeå University, Umeå, Sweden
| | - Helmut Hirt
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, United States
| | - Gary M Dunny
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, United States
| | - Ronnie P-A Berntsson
- Department of Medical Biochemistry and Biophysics, Umeå University, Umeå, Sweden.,Wallenberg Centre for Molecular Medicine, Umeå University, Umeå, Sweden
| |
Collapse
|
6
|
Rivière G, Peng EQ, Brotgandel A, Andring JT, Lakshmanan RV, Agbandje-McKenna M, McKenna R, Brady LJ, Long JR. Characterization of an intermolecular quaternary interaction between discrete segments of the Streptococcus mutans adhesin P1 by NMR spectroscopy. FEBS J 2019; 287:2597-2611. [PMID: 31782893 DOI: 10.1111/febs.15158] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 09/13/2019] [Accepted: 11/27/2019] [Indexed: 11/30/2022]
Abstract
Cell surface-localized P1 adhesin (aka Antigen I/II or PAc) of the cariogenic bacterium Streptococcus mutans mediates sucrose-independent adhesion to tooth surfaces. Previous studies showed that P1's C-terminal segment (C123, AgII) is also liberated as a separate polypeptide, contributes to cellular adhesion, interacts specifically with intact P1 on the cell surface, and forms amyloid fibrils. Identifying how C123 specifically interacts with P1 at the atomic level is essential for understanding related virulence properties of S. mutans. However, with sizes of ~ 51 and ~ 185 kDa, respectively, C123 and full-length P1 are too large to achieve high-resolution data for full structural analysis by NMR. Here, we report on biologically relevant interactions of the individual C3 domain with A3VP1, a polypeptide that represents the apical head of P1 as it is projected on the cell surface. Also evaluated are C3's interaction with C12 and the adhesion-inhibiting monoclonal antibody (MAb) 6-8C. NMR titration experiments with 15 N-enriched C3 demonstrate its specific binding to A3VP1. Based on resolved C3 assignments, two binding sites, proximal and distal, are identified. Complementary NMR titration of A3VP1 with a C3/C12 complex suggests that binding of A3VP1 occurs on the distal C3 binding site, while the proximal site is occupied by C12. The MAb 6-8C binding interface to C3 overlaps with that of A3VP1 at the distal site. Together, these results identify a specific C3-A3VP1 interaction that serves as a foundation for understanding the interaction of C123 with P1 on the bacterial surface and the related biological processes that stem from this interaction. DATABASE: BMRB submission code: 27935.
Collapse
Affiliation(s)
- Gwladys Rivière
- Department of Biochemistry and Molecular Biology and McKnight Brain Institute, University of Florida, Gainesville, FL, USA.,National High Magnetic Field Laboratory, University of Florida, Gainesville, FL, USA
| | - Emily-Qingqing Peng
- Department of Biochemistry and Molecular Biology and McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Albert Brotgandel
- Department of Biochemistry and Molecular Biology and McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Jacob T Andring
- Department of Biochemistry and Molecular Biology and McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Renuk V Lakshmanan
- Department of Biochemistry and Molecular Biology and McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Mavis Agbandje-McKenna
- Department of Biochemistry and Molecular Biology and McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Robert McKenna
- Department of Biochemistry and Molecular Biology and McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - L Jeannine Brady
- College of Dentistry, Department of Oral Biology, University of Florida, Gainesville, FL, USA
| | - Joanna R Long
- Department of Biochemistry and Molecular Biology and McKnight Brain Institute, University of Florida, Gainesville, FL, USA.,National High Magnetic Field Laboratory, University of Florida, Gainesville, FL, USA
| |
Collapse
|
7
|
Rego S, Heal TJ, Pidwill GR, Till M, Robson A, Lamont RJ, Sessions RB, Jenkinson HF, Race PR, Nobbs AH. Structural and Functional Analysis of Cell Wall-anchored Polypeptide Adhesin BspA in Streptococcus agalactiae. J Biol Chem 2016; 291:15985-6000. [PMID: 27311712 DOI: 10.1074/jbc.m116.726562] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2016] [Indexed: 12/21/2022] Open
Abstract
Streptococcus agalactiae (group B Streptococcus, GBS) is the predominant cause of early-onset infectious disease in neonates and is responsible for life-threatening infections in elderly and immunocompromised individuals. Clinical manifestations of GBS infection include sepsis, pneumonia, and meningitis. Here, we describe BspA, a deviant antigen I/II family polypeptide that confers adhesive properties linked to pathogenesis in GBS. Heterologous expression of BspA on the surface of the non-adherent bacterium Lactococcus lactis confers adherence to scavenger receptor gp340, human vaginal epithelium, and to the fungus Candida albicans Complementary crystallographic and biophysical characterization of BspA reveal a novel β-sandwich adhesion domain and unique asparagine-dependent super-helical stalk. Collectively, these findings establish a new bacterial adhesin structure that has in effect been hijacked by a pathogenic Streptococcus species to provide competitive advantage in human mucosal infections.
Collapse
Affiliation(s)
- Sara Rego
- From the School of Oral and Dental Sciences, University of Bristol, Bristol BS1 2LY, United Kingdom, the School of Biochemistry, University of Bristol, Bristol BS8 1TD, United Kingdom
| | - Timothy J Heal
- the School of Biochemistry, University of Bristol, Bristol BS8 1TD, United Kingdom, the Bristol Centre for Functional Nanomaterials, University of Bristol, Bristol BS8 1TL, United Kingdom
| | - Grace R Pidwill
- From the School of Oral and Dental Sciences, University of Bristol, Bristol BS1 2LY, United Kingdom
| | - Marisa Till
- the School of Biochemistry, University of Bristol, Bristol BS8 1TD, United Kingdom, the BrisSynBio Synthetic Biology Research Centre, University of Bristol, Bristol BS8 1TQ, United Kingdom, and
| | - Alice Robson
- the School of Biochemistry, University of Bristol, Bristol BS8 1TD, United Kingdom
| | - Richard J Lamont
- the Department of Oral Immunology and Infectious Diseases, University of Louisville, Louisville, Kentucky 40202
| | - Richard B Sessions
- the School of Biochemistry, University of Bristol, Bristol BS8 1TD, United Kingdom, the BrisSynBio Synthetic Biology Research Centre, University of Bristol, Bristol BS8 1TQ, United Kingdom, and
| | - Howard F Jenkinson
- From the School of Oral and Dental Sciences, University of Bristol, Bristol BS1 2LY, United Kingdom
| | - Paul R Race
- the School of Biochemistry, University of Bristol, Bristol BS8 1TD, United Kingdom, the BrisSynBio Synthetic Biology Research Centre, University of Bristol, Bristol BS8 1TQ, United Kingdom, and
| | - Angela H Nobbs
- From the School of Oral and Dental Sciences, University of Bristol, Bristol BS1 2LY, United Kingdom,
| |
Collapse
|
8
|
Isopeptide bond in collagen- and fibrinogen-binding MSCRAMMs. Biophys Rev 2016; 8:75-83. [PMID: 28510145 DOI: 10.1007/s12551-015-0191-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 12/14/2015] [Indexed: 10/22/2022] Open
Abstract
The internal isopeptide bonds are amide bonds formed autocatalytically between the side chains of Lys and Asn/Asp residues and have been discovered recently. These bonds are well conserved in Gram-positive bacterial pilin proteins and are also observed over a wide range of Gram-positive bacterial surface proteins. The presence of these bonds confers the pilus subunits with remarkable properties in terms of thermal stability and resistance to proteases. Like pili, microbial surface components recognizing adhesive matrix molecules (MSCRAMMs) are also surface proteins found only in Gram-positive bacteria. They specifically interact with the extracellular matrix (ECM) molecules like collagen, fibrinogen, fibronectin, laminin, etc. Many biophysical and biochemical studies have been carried out to characterize the isopeptide bonds in pili proteins from Gram-positive bacteria, but no attempts have been made to study the isopeptide bonds in MSCRAMMs. This short review aims to study the significance of the isopeptide bonds in relation to their function, by analyzing the crystal structures of collagen- and fibrinogen-binding MSCRAMMs. In this analysis, interestingly, we observed that the putative isopeptide bonds are restricted to the collagen-binding MSCRAMMs. Based on analogy with bacterial pilus subunits, we hypothesize that the collagen-binding MSCRAMMs possessing putative isopeptide bonds exhibit similar structural properties, which could help the bacteria in colonizing the host and provide resistance against host-defense mechanisms.
Collapse
|
9
|
Abstract
Protein-protein interactions are fundamental to many biological processes. Yet, the weak and transient noncovalent bonds that characterize most protein-protein interactions found in nature impose limits on many bioengineering experiments. Here, a new class of genetically encodable peptide-protein pairs--isopeptag-N/pilin-N, isopeptag/pilin-C, and SpyTag/SpyCatcher--that interact through autocatalytic intermolecular isopeptide bond formation is described. Reactions between peptide-protein pairs are specific, robust, orthogonal, and able to proceed under most biologically relevant conditions both in vitro and in vivo. As fusion constructs, they provide a handle on molecules of interest, both organic and inorganic, that can be grasped with an iron grip. Such stable interactions provide robust post-translational control over biological processes and open new opportunities in synthetic biology for engineering programmable and self-assembling protein nanoarchitectures.
Collapse
Affiliation(s)
- Bijan Zakeri
- Department of Electrical Engineering and Computer Science, Department of Biological Engineering, Research Laboratory of Electronics, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA. .,MIT Synthetic Biology Center, 500 Technology Square, Cambridge, MA, 02139, USA.
| |
Collapse
|
10
|
Sullan RMA, Li JK, Crowley PJ, Brady LJ, Dufrêne YF. Binding forces of Streptococcus mutans P1 adhesin. ACS NANO 2015; 9:1448-60. [PMID: 25671413 PMCID: PMC4369792 DOI: 10.1021/nn5058886] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Streptococcus mutans is a Gram-positive oral bacterium that is a primary etiological agent associated with human dental caries. In the oral cavity, S. mutans adheres to immobilized salivary agglutinin (SAG) contained within the salivary pellicle on the tooth surface. Binding to SAG is mediated by cell surface P1, a multifunctional adhesin that is also capable of interacting with extracellular matrix proteins. This may be of particular importance outside of the oral cavity as S. mutans has been associated with infective endocarditis and detected in atherosclerotic plaque. Despite the biomedical importance of P1, its binding mechanisms are not completely understood. In this work, we use atomic force microscopy-based single-molecule and single-cell force spectroscopy to quantify the nanoscale forces driving P1-mediated adhesion. Single-molecule experiments show that full-length P1, as well as fragments containing only the P1 globular head or C-terminal region, binds to SAG with relatively weak forces (∼50 pN). In contrast, single-cell analyses reveal that adhesion of a single S. mutans cell to SAG is mediated by strong (∼500 pN) and long-range (up to 6000 nm) forces. This is likely due to the binding of multiple P1 adhesins to self-associated gp340 glycoproteins. Such a cooperative, long-range character of the S. mutans-SAG interaction would therefore dramatically increase the strength and duration of cell adhesion. We also demonstrate, at single-molecule and single-cell levels, the interaction of P1 with fibronectin and collagen, as well as with hydrophobic, but not hydrophilic, substrates. The binding mechanism (strong forces, cooperativity, broad specificity) of P1 provides a molecular basis for its multifunctional adhesion properties. Our methodology represents a valuable approach to probe the binding forces of bacterial adhesins and offers a tractable methodology to assess anti-adhesion therapy.
Collapse
Affiliation(s)
- Ruby May A. Sullan
- Institute of Life Sciences, Université Catholique de Louvain, Louvain-la-Neuve, Belgium B-1348
| | - James K. Li
- Institute for Optical Sciences, University of Toronto, Toronto, Ontario M5S 3H8, Canada
| | - Paula J. Crowley
- Department of Oral Biology, University of Florida, Gainesville, Florida 32603, United States
| | - L. Jeannine Brady
- Department of Oral Biology, University of Florida, Gainesville, Florida 32603, United States
| | - Yves F. Dufrêne
- Institute of Life Sciences, Université Catholique de Louvain, Louvain-la-Neuve, Belgium B-1348
| |
Collapse
|
11
|
Heim KP, Sullan RMA, Crowley PJ, El-Kirat-Chatel S, Beaussart A, Tang W, Besingi R, Dufrene YF, Brady LJ. Identification of a supramolecular functional architecture of Streptococcus mutans adhesin P1 on the bacterial cell surface. J Biol Chem 2015; 290:9002-19. [PMID: 25666624 DOI: 10.1074/jbc.m114.626663] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2014] [Indexed: 12/29/2022] Open
Abstract
P1 (antigen I/II) is a sucrose-independent adhesin of Streptococcus mutans whose functional architecture on the cell surface is not fully understood. S. mutans cells subjected to mechanical extraction were significantly diminished in adherence to immobilized salivary agglutinin but remained immunoreactive and were readily aggregated by fluid-phase salivary agglutinin. Bacterial adherence was restored by incubation of postextracted cells with P1 fragments that contain each of the two known adhesive domains. In contrast to untreated cells, glutaraldehyde-treated bacteria gained reactivity with anti-C-terminal monoclonal antibodies (mAbs), whereas epitopes recognized by mAbs against other portions of the molecule were masked. Surface plasmon resonance experiments demonstrated the ability of apical and C-terminal fragments of P1 to interact. Binding of several different anti-P1 mAbs to unfixed cells triggered release of a C-terminal fragment from the bacterial surface, suggesting a novel mechanism of action of certain adherence-inhibiting antibodies. We also used atomic force microscopy-based single molecule force spectroscopy with tips bearing various mAbs to elucidate the spatial organization and orientation of P1 on living bacteria. The similar rupture lengths detected using mAbs against the head and C-terminal regions, which are widely separated in the tertiary structure, suggest a higher order architecture in which these domains are in close proximity on the cell surface. Taken together, our results suggest a supramolecular organization in which additional P1 polypeptides, including the C-terminal segment originally identified as antigen II, associate with covalently attached P1 to form the functional adhesive layer.
Collapse
Affiliation(s)
- Kyle P Heim
- From the Department of Oral Biology, University of Florida, Gainesville, Florida 32610 and
| | - Ruby May A Sullan
- Institute of Life Sciences, Université catholique de Louvain, B-1348 Louvain-la-Neuve, Belgium
| | - Paula J Crowley
- From the Department of Oral Biology, University of Florida, Gainesville, Florida 32610 and
| | - Sofiane El-Kirat-Chatel
- Institute of Life Sciences, Université catholique de Louvain, B-1348 Louvain-la-Neuve, Belgium
| | - Audrey Beaussart
- Institute of Life Sciences, Université catholique de Louvain, B-1348 Louvain-la-Neuve, Belgium
| | - Wenxing Tang
- From the Department of Oral Biology, University of Florida, Gainesville, Florida 32610 and
| | - Richard Besingi
- From the Department of Oral Biology, University of Florida, Gainesville, Florida 32610 and
| | - Yves F Dufrene
- Institute of Life Sciences, Université catholique de Louvain, B-1348 Louvain-la-Neuve, Belgium
| | - L Jeannine Brady
- From the Department of Oral Biology, University of Florida, Gainesville, Florida 32610 and
| |
Collapse
|
12
|
An intramolecular lock facilitates folding and stabilizes the tertiary structure of Streptococcus mutans adhesin P1. Proc Natl Acad Sci U S A 2014; 111:15746-51. [PMID: 25331888 DOI: 10.1073/pnas.1413018111] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The cariogenic bacterium Streptococcus mutans uses adhesin P1 to adhere to tooth surfaces, extracellular matrix components, and other bacteria. A composite model of P1 based on partial crystal structures revealed an unusual complex architecture in which the protein forms an elongated hybrid alpha/polyproline type II helical stalk by folding back on itself to display a globular head at the apex and a globular C-terminal region at the base. The structure of P1's N terminus and the nature of its critical interaction with the C-terminal region remained unknown, however. We have cocrystallized a stable complex of recombinant N- and C-terminal fragments and here describe a previously unidentified topological fold in which these widely discontinuous domains are intimately associated. The structure reveals that the N terminus forms a stabilizing scaffold by wrapping behind the base of P1's elongated stalk and physically "locking" it into place. The structure is stabilized through a highly favorable ΔG(solvation) on complex formation, along with extensive hydrogen bonding. We confirm the functional relevance of this intramolecular interaction using differential scanning calorimetry and circular dichroism to show that disruption of the proper spacing of residues 989-1001 impedes folding and diminishes stability of the full-length molecule, including the stalk. Our findings clarify previously unexplained functional and antigenic properties of P1.
Collapse
|
13
|
Hall M, Nylander S, Jenkinson HF, Persson K. Structure of the C-terminal domain of AspA (antigen I/II-family) protein from Streptococcus pyogenes. FEBS Open Bio 2014; 4:283-9. [PMID: 24918040 PMCID: PMC4048849 DOI: 10.1016/j.fob.2014.02.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Revised: 02/27/2014] [Accepted: 02/28/2014] [Indexed: 01/01/2023] Open
Abstract
The structure of the C2–3-domain of AspA from S. pyogenes was determined. The C2 and C3 domains both adopt DEv-IgG folds. Conserved isopeptide bonds and calcium binding sites are observed. Distinct structural features are observed in the SspB Adherence Region (BAR).
The pathogenic bacteria Streptococcus pyogenes can cause an array of diseases in humans, including moderate infections such as pharyngitis (strep throat) as well as life threatening conditions such as necrotizing fasciitis and puerperal fever. The antigen I/II family proteins are cell wall anchored adhesin proteins found on the surfaces of most oral streptococci and are involved in host colonization and biofilm formation. In the present study we have determined the crystal structure of the C2–3-domain of the antigen I/II type protein AspA from S. pyogenes M type 28. The structure was solved to 1.8 Å resolution and shows that the C2–3-domain is comprised of two structurally similar DEv-IgG motifs, designated C2 and C3, both containing a stabilizing covalent isopeptide bond. Furthermore a metal binding site is identified, containing a bound calcium ion. Despite relatively low sequence identity, interestingly, the overall structure shares high similarity to the C2–3-domains of antigen I/II proteins from Streptococcus gordonii and Streptococcus mutans, although certain parts of the structure exhibit distinct features. In summary this work constitutes the first step in the full structure determination of the AspA protein from S. pyogenes.
Collapse
Affiliation(s)
- Michael Hall
- Department of Chemistry, Umeå University, SE-901 87 Umeå, Sweden
| | - Sa Nylander
- Department of Odontology, Division of Oral Microbiology, Umeå University, SE-901 87 Umeå, Sweden
| | - Howard F Jenkinson
- School of Oral and Dental Sciences, University of Bristol, Bristol BS1 2LY, UK
| | - Karina Persson
- Department of Chemistry, Umeå University, SE-901 87 Umeå, Sweden
| |
Collapse
|
14
|
Heim KP, Crowley PJ, Brady LJ. An intramolecular interaction involving the N terminus of a streptococcal adhesin affects its conformation and adhesive function. J Biol Chem 2013; 288:13762-74. [PMID: 23539625 DOI: 10.1074/jbc.m113.459974] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND P1 is an adhesin on the surface of Streptococcus mutans. RESULTS Destroying the high affinity interaction between the N and C termini of S. mutans P1 creates a non-adherent phenotype. CONCLUSION The N terminus facilitates proper folding, function, and stability within recombinant P1. SIGNIFICANCE The relationship between folding, maturation, and cell surface assembly is critical to understanding the P1 mechanism of action. The adhesin P1 is localized on the surface of the oral pathogen Streptococcus mutans and facilitates an interaction with the glycoprotein complex salivary agglutinin that is comprised primarily of the scavenger receptor gp340. Recent crystal structures of P1 display an unusual structure in which the protein folds back upon itself to form an elongated hybrid helical stalk with a globular head at the apex and a globular C-terminal region at the base. The N terminus of P1 has not yet been characterized. In this report we describe the contribution of an interaction between the N-terminal and C-terminal portions of the protein that is required for proper function of P1 on the surface of S. mutans. Utilizing recombinant N-terminal and C-terminal fragments, we employed isothermal titration calorimetry and native gel electrophoresis to demonstrate that these fragments form a high affinity and stable complex in solution. Furthermore, circular dichroism and surface plasmon resonance measurements indicated that the N-terminal fragment contributes to the folding and increases the functionality of the C-terminal fragment in trans. Finally, we utilized circular dichroism, surface plasmon resonance, and differential scanning calorimetry to show that an N-terminal 106-amino acid segment within P1 contributes to the proper folding and function of the full-length recombinant molecule and increases the stability of its elongated hybrid helical stalk.
Collapse
Affiliation(s)
- Kyle P Heim
- Department of Oral Biology, University of Florida, Gainesville, Florida 32610, USA
| | | | | |
Collapse
|
15
|
Oli MW, Otoo HN, Crowley PJ, Heim KP, Nascimento MM, Ramsook CB, Lipke PN, Brady LJ. Functional amyloid formation by Streptococcus mutans. MICROBIOLOGY-SGM 2012; 158:2903-2916. [PMID: 23082034 DOI: 10.1099/mic.0.060855-0] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Dental caries is a common infectious disease associated with acidogenic and aciduric bacteria, including Streptococcus mutans. Organisms that cause cavities form recalcitrant biofilms, generate acids from dietary sugars and tolerate acid end products. It has recently been recognized that micro-organisms can produce functional amyloids that are integral to biofilm development. We now show that the S. mutans cell-surface-localized adhesin P1 (antigen I/II, PAc) is an amyloid-forming protein. This conclusion is based on the defining properties of amyloids, including binding by the amyloidophilic dyes Congo red (CR) and Thioflavin T (ThT), visualization of amyloid fibres by transmission electron microscopy and the green birefringent properties of CR-stained protein aggregates when viewed under cross-polarized light. We provide evidence that amyloid is present in human dental plaque and is produced by both laboratory strains and clinical isolates of S. mutans. We provide further evidence that amyloid formation is not limited to P1, since bacterial colonies without this adhesin demonstrate residual green birefringence. However, S. mutans lacking sortase, the transpeptidase enzyme that mediates the covalent linkage of its substrates to the cell-wall peptidoglycan, including P1 and five other proteins, is not birefringent when stained with CR and does not form biofilms. Biofilm formation is inhibited when S. mutans is cultured in the presence of known inhibitors of amyloid fibrillization, including CR, Thioflavin S and epigallocatechin-3-gallate, which also inhibited ThT uptake by S. mutans extracellular proteins. Taken together, these results indicate that S. mutans is an amyloid-forming organism and suggest that amyloidogenesis contributes to biofilm formation by this oral microbe.
Collapse
Affiliation(s)
- M W Oli
- University of Florida, Department of Oral Biology, Gainesville, FL, USA
| | - H N Otoo
- University of Florida, Department of Oral Biology, Gainesville, FL, USA
| | - P J Crowley
- University of Florida, Department of Oral Biology, Gainesville, FL, USA
| | - K P Heim
- University of Florida, Department of Oral Biology, Gainesville, FL, USA
| | - M M Nascimento
- University of Florida, Department of Oral Biology, Gainesville, FL, USA
| | - C B Ramsook
- Department of Biology, Brooklyn College of the City University of New York, NY, USA
| | - P N Lipke
- Department of Biology, Brooklyn College of the City University of New York, NY, USA
| | - L J Brady
- University of Florida, Department of Oral Biology, Gainesville, FL, USA
| |
Collapse
|
16
|
Larson MR, Rajashankar KR, Crowley PJ, Kelly C, Mitchell TJ, Brady LJ, Deivanayagam C. Crystal structure of the C-terminal region of Streptococcus mutans antigen I/II and characterization of salivary agglutinin adherence domains. J Biol Chem 2011; 286:21657-66. [PMID: 21505225 DOI: 10.1074/jbc.m111.231100] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The Streptococcus mutans antigen I/II (AgI/II) is a cell surface-localized protein that adheres to salivary components and extracellular matrix molecules. Here we report the 2.5 Å resolution crystal structure of the complete C-terminal region of AgI/II. The C-terminal region is comprised of three major domains: C(1), C(2), and C(3). Each domain adopts a DE-variant IgG fold, with two β-sheets whose A and F strands are linked through an intramolecular isopeptide bond. The adherence of the C-terminal AgI/II fragments to the putative tooth surface receptor salivary agglutinin (SAG), as monitored by surface plasmon resonance, indicated that the minimal region of binding was contained within the first and second DE-variant-IgG domains (C(1) and C(2)) of the C terminus. The minimal C-terminal region that could inhibit S. mutans adherence to SAG was also confirmed to be within the C(1) and C(2) domains. Competition experiments demonstrated that the C- and N-terminal regions of AgI/II adhere to distinct sites on SAG. A cleft formed at the intersection between these C(1) and C(2) domains bound glucose molecules from the cryo-protectant solution, revealing a putative binding site for its highly glycosylated receptor SAG. Finally, electron microscopy images confirmed the elongated structure of AgI/II and enabled building a composite tertiary model that encompasses its two distinct binding regions.
Collapse
Affiliation(s)
- Matthew R Larson
- Department of Physiology and Biophysics, University of Alabama, Birmingham, Alabama 35294, USA
| | | | | | | | | | | | | |
Collapse
|