1
|
Thayumanavan T, Harish BS, Subashkumar R, Shanmugapriya K, Karthik V. Streptococcus mutans biofilms in the establishment of dental caries: a review. 3 Biotech 2025; 15:62. [PMID: 39959706 PMCID: PMC11828782 DOI: 10.1007/s13205-025-04227-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 01/18/2025] [Indexed: 02/18/2025] Open
Abstract
Dental caries is considered as the most common and multifactorial disease worldwide, caused by a variety of oral microorganisms like Streptococcus spp., Veillonella spp., Actinomyces spp., Bifidobacterium spp., and Lactobacillus fermentum, which colonize food debris in oral cavities. Of them, Streptococcus mutans is the predominant bacterium and can induce progressive tooth destruction, especially during dentition. The superior characteristics of S. mutans, such as the presence of the cell surface protein P1 and exopolysaccharide-synthesizing enzymes, acid tolerance, biofilm-forming ability mediated by brpA gene, and multidrug resistance, render it a highly virulent pathogen in the etiology of dental caries. Given its significant role in dental caries, extensive research has been conducted over the past few decades, focusing on the development of specific antimicrobial treatments, and other innovative therapeutic approaches. To gain deeper insights into the genetic diversity and epidemiological patterns of S. mutans, various genotypic methods have been developed and successfully employed. By combining the insights gained from genetic studies of S. mutans with the suitable control measures against the biofilm, we can develop innovative and effective strategies for preventing and treating dental caries.
Collapse
Affiliation(s)
- Thangavelu Thayumanavan
- Department of Biotechnology, KIT-Kalaignarkarunanidhi Institute of Technology (Autonomous), Coimbatore, 641 402 India
| | - B. S. Harish
- Department of Biotechnology, KIT-Kalaignarkarunanidhi Institute of Technology (Autonomous), Coimbatore, 641 402 India
| | - Rathinasamy Subashkumar
- Department of Biotechnology, Sri Ramakrishna College of Arts and Science (Autonomous), Coimbatore, 641 006 India
| | - Karuppusamy Shanmugapriya
- UCD School of Biosystems and Food Engineering, University College Dublin, Dublin 4, Belfield, Ireland
| | - Velusamy Karthik
- Department of Industrial Biotechnology, Government College of Technology, Coimbatore, 641 013 India
| |
Collapse
|
2
|
Ellepola K, Shields RC, Kajfasz JK, Zhang H, Lemos JA, Wu H, Wen ZT. MecA in Streptococcus mutans is a multi-functional protein. mSphere 2024; 9:e0030824. [PMID: 39530674 PMCID: PMC11656736 DOI: 10.1128/msphere.00308-24] [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: 04/12/2024] [Accepted: 09/09/2024] [Indexed: 11/16/2024] Open
Abstract
Our recent studies have shown that deficiency of MecA in Streptococcus mutans significantly affects cell division, growth, and biofilm formation. In this study, an in vitro mixed-species model, proteomics, and affinity pull-down assays were used to further characterize the MecA-mediated regulation in S. mutans. The results showed that compared with the wild type, UA159, the mecA mutant significantly reduced its production of glucans and weakened its ability to facilitate mixed-species biofilm formation. Relative to the wild type, the mecA mutant also displayed unique characteristics, including colony morphology, growth rate, and biofilm formation that did not fully resemble any of the clpP, clpX, clpE, clpCE, and clpC individual or combinational mutants. Deletion of mecA was shown to result in alteration of >337 proteins, including down expression of GtfBC&D and adhesin P1. More than 277 proteins were differentially expressed in response to clpP deletion, including increased expression of GtfB. By cross-referencing the two proteomes, a distinctive set of proteins was found to be altered in the mecA mutant, indicating a ClpP-independent role of MecA in the regulation of S. mutans. When analyzed using affinity pull-down, ClpC, ClpX, ClpE, and CcpA were among the members identified in the MecA-associated complex. Further analysis using a bacterial two-hybrid system confirmed CcpA, ClpX, and ClpE as members of the MecA interactome. These results further suggest that MecA in S. mutans is more than an adapter of the Clp-proteolytic machinery, although the mechanism that underlies the Clp-independent regulation and its impact on S. mutans pathophysiology await further investigation. IMPORTANCE MecA is known as an adaptor protein that works in concerto with ATPase ClpC and protease ClpP in the regulated proteolysis machinery. The results presented here provide further evidence that MecA in S. mutans, a keystone cariogenic bacterium, plays a significant role in its ability to facilitate mixed-species biofilm formation, a trait critical to its cariogenicity. Proteomics analysis, along with affinity pull-down and bacterial two-hybrid system, further confirm that MecA can also regulate S. mutans physiology and biofilm formation through pathways independent of the Clp proteolytic machinery, although how it functions independently of Clp awaits further investigation.
Collapse
Affiliation(s)
- Kassapa Ellepola
- Department of Oral and Craniofacial Biology, School of Dentistry, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Robert C. Shields
- Department of Biological Sciences, Arkansas State University, Jonesboro, Arkansas, USA
| | - Jessica K. Kajfasz
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, Florida, USA
| | - Hua Zhang
- Department of Restorative Dentistry, School of Dentistry, Oregon Health and Science University, Portland, Oregon, USA
| | - Jose A. Lemos
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, Florida, USA
| | - Hui Wu
- Department of Restorative Dentistry, School of Dentistry, Oregon Health and Science University, Portland, Oregon, USA
| | - Zezhang T. Wen
- Department of Oral and Craniofacial Biology, School of Dentistry, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
- Department of Microbiology, Immunology and Parasitology, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| |
Collapse
|
3
|
Ding X, Zheng Z, Zhao G, Wang L, Wang H, Wang P. Adaptive laboratory evolution for improved tolerance of vitamin K in Bacillus subtilis. Appl Microbiol Biotechnol 2024; 108:75. [PMID: 38194140 DOI: 10.1007/s00253-023-12877-7] [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: 06/19/2023] [Revised: 10/10/2023] [Accepted: 10/30/2023] [Indexed: 01/10/2024]
Abstract
Menaquinone-7 (MK-7), a subtype of vitamin K2 (VK2), assumes crucial roles in coagulation function, calcium homeostasis, and respiratory chain transmission. The production of MK-7 via microbial fermentation boasts mild technological conditions and high biocompatibility. Nevertheless, the redox activity of MK-7 imposes constraints on its excessive accumulation in microorganisms. To address this predicament, an adaptive laboratory evolution (ALE) protocol was implemented in Bacillus subtilis BS011, utilizing vitamin K3 (VK3) as a structural analog of MK-7. The resulting strain, BS012, exhibited heightened tolerance to high VK3 concentrations and demonstrated substantial enhancements in biofilm formation and total antioxidant capacity (T-AOC) when compared to BS011. Furthermore, MK-7 production in BS012 exceeded that of BS011 by 76% and 22% under static and shaking cultivation conditions, respectively. The molecular basis underlying the superior performance of BS012 was elucidated through genome and transcriptome analyses, encompassing observations of alterations in cell morphology, variations in central carbon and nitrogen metabolism, spore formation, and antioxidant systems. In summation, ALE technology can notably enhance the tolerance of B. subtilis to VK and increase MK-7 production, thus offering a theoretical framework for the microbial fermentation production of other VK2 subtypes. Additionally, the evolved strain BS012 can be developed for integration into probiotic formulations within the food industry to maintain intestinal flora homeostasis, mitigate osteoporosis risk, and reduce the incidence of cardiovascular disease. KEY POINTS: • Bacillus subtilis was evolved for improved vitamin K tolerance and menaquinone-7 (MK-7) production • Evolved strains formed wrinkled biofilms and elongated almost twofold in length • Evolved strains induced sporulation to improve tolerance when carbon was limited.
Collapse
Affiliation(s)
- Xiumin Ding
- Institute of Intelligent Machines, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, China
- Department of Health Inspection and Quarantine, Wannan Medical College, Wuhu, China
- University of Science and Technology of China, Hefei, China
| | - Zhiming Zheng
- Institute of Intelligent Machines, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, China.
| | - Genhai Zhao
- Institute of Intelligent Machines, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, China
| | - Li Wang
- Institute of Intelligent Machines, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, China
| | - Han Wang
- Institute of Intelligent Machines, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, China
| | - Peng Wang
- Institute of Intelligent Machines, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, China.
| |
Collapse
|
4
|
Ellepola K, Guillot LC, Comeaux B, Han Y, Kajfasz JK, Bitoun JP, Spatafora G, Lemos JA, Wen ZT. Multiple factors regulate the expression of sufCDSUB in Streptococcus mutans. Front Cell Infect Microbiol 2024; 14:1499476. [PMID: 39664495 PMCID: PMC11631912 DOI: 10.3389/fcimb.2024.1499476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 11/05/2024] [Indexed: 12/13/2024] Open
Abstract
Introduction The sufCDSUB gene cluster, encoding the sole iron-sulfur (Fe-S) cluster assembly system in S. mutans, was recently shown to be up-regulated in response to oxidative stressors and Fe limitation. Methods In this study, luciferase reporter fusion assays, electrophoretic gel mobility shift assays (EMSA) and in vitro transcription assays (IVT) were used to dissect the cis- and trans-acting factors that regulate the expression of sufCDSUB. Results and discussion Results showed deletion of perR, for the only Fur-family transcriptional regulator in S. mutans, resulted in >5-fold increases in luciferase activity under the control of the sufCDSUB promoter (P<0.01), as compared to the parent strain, UA159 when the reporter strains were grown in medium with no supplemental iron. Site-directed mutagenesis of a PerR-box in the promoter region led to elevation of the reporter activity by >1.6-fold (P<0.01). In an EMSA, recombinant PerR (rPerR) was shown to bind to the cognate sufCDSUB promoter leading to mobility retardation. On the other hand, the reporter activity was increased by >84-fold (P<0.001) in response to the addition of cysteine at 4 mM to the culture medium. Deletion of cysR, for a LysR-type of transcriptional regulator, led to reduction of the reporter activity by >11.6-fold (P<0.001). Addition of recombinant CysR (rCysR) to an EMSA caused mobility shift of the sufCDSUB promoter probe, indicative of rCysR-promoter interaction, and rCysR was shown to enhance sufC transcription under the direction of sufCDSUB promoter in vitro. These results suggest that multiple factors are involved in the regulation of sufCDSUB expression in response to environmental cues, including cysteine and Fe availability, consistent with the important role of sufCDSUB in S. mutans physiology.
Collapse
Affiliation(s)
- Kassapa Ellepola
- Department of Oral and Craniofacial Biology, School of Dentistry, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Lauren C. Guillot
- Department of Oral and Craniofacial Biology, School of Dentistry, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Bradley Comeaux
- Department of Oral and Craniofacial Biology, School of Dentistry, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Yiran Han
- Department of Biology, Middlebury College, Middlebury, VT, United States
| | - Jessica K. Kajfasz
- Department of Oral Biology, School of Dentistry, University of Florida, Gainesville, FL, United States
| | - Jacob P. Bitoun
- Department of Microbiology, Tulane University, New Orleans, LA, United States
| | - Grace Spatafora
- Department of Biology, Middlebury College, Middlebury, VT, United States
| | - Jose A. Lemos
- Department of Oral Biology, School of Dentistry, University of Florida, Gainesville, FL, United States
| | - Zezhang T. Wen
- Department of Oral and Craniofacial Biology, School of Dentistry, Louisiana State University Health Sciences Center, New Orleans, LA, United States
- Department of Microbiology, Immunology and Parasitology, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| |
Collapse
|
5
|
Zou Z, Singh P, Pinkner JS, Obernuefemann CLP, Xu W, Nye TM, Dodson KW, Almqvist F, Hultgren SJ, Caparon MG. Dihydrothiazolo ring-fused 2-pyridone antimicrobial compounds treat Streptococcus pyogenes skin and soft tissue infection. SCIENCE ADVANCES 2024; 10:eadn7979. [PMID: 39093975 PMCID: PMC11296344 DOI: 10.1126/sciadv.adn7979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 06/27/2024] [Indexed: 08/04/2024]
Abstract
We have developed GmPcides from a peptidomimetic dihydrothiazolo ring-fused 2-pyridone scaffold that has antimicrobial activities against a broad spectrum of Gram-positive pathogens. Here, we examine the treatment efficacy of GmPcides using skin and soft tissue infection (SSTI) and biofilm formation models by Streptococcus pyogenes. Screening our compound library for minimal inhibitory (MIC) and minimal bactericidal (MBC) concentrations identified GmPcide PS757 as highly active against S. pyogenes. Treatment of S. pyogenes biofilm with PS757 revealed robust efficacy against all phases of biofilm formation by preventing initial biofilm development, ceasing biofilm maturation and eradicating mature biofilm. In a murine model of S. pyogenes SSTI, subcutaneous delivery of PS757 resulted in reduced levels of tissue damage, decreased bacterial burdens, and accelerated rates of wound healing, which were associated with down-regulation of key virulence factors, including M protein and the SpeB cysteine protease. These data demonstrate that GmPcides show considerable promise for treating S. pyogenes infections.
Collapse
Affiliation(s)
- Zongsen Zou
- Department of Molecular Microbiology, Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Pardeep Singh
- Department of Chemistry, Umeå University, SE-90187 Umeå, Sweden
| | - Jerome S. Pinkner
- Department of Molecular Microbiology, Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Chloe L. P. Obernuefemann
- Department of Molecular Microbiology, Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Wei Xu
- Department of Molecular Microbiology, Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Taylor M. Nye
- Department of Molecular Microbiology, Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Karen W. Dodson
- Department of Molecular Microbiology, Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | - Scott J. Hultgren
- Department of Molecular Microbiology, Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Michael G. Caparon
- Department of Molecular Microbiology, Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
6
|
Si B, Yang Y, Naveed M, Wang F, Chan MWH. Characterizations of biogenic selenium nanoparticles and their anti-biofilm potential against Streptococcus mutans ATCC 25175. J Trace Elem Med Biol 2024; 84:127448. [PMID: 38626650 DOI: 10.1016/j.jtemb.2024.127448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 04/03/2024] [Accepted: 04/05/2024] [Indexed: 04/18/2024]
Abstract
INTRODUCTION S. mutans has been identified as the primary pathogenic bacterium in biofilm-mediated dental caries. The biogenic selenium nanoparticles (SeNPs) produced by L. plantarum KNF-5 were used in this study against S. mutans ATCC 25175. OBJECTIVES The aims of this study were: (1) the biosynthesis of SeNPs by L. plantarum KNF-5, (2) the characterization of SeNPs, (3) the investigation of the inhibitory effect of biogenic SeNPs against S. mutans ATCC 25175, and (4) the determination of the anti-biofilm potential of SeNPS against S. mutans ATCC 25175. METHODOLOGY 3 mL of the culture was added to 100 mL of MRS medium and incubated. After 4 h, Na2SeO3 solution (concentration 100 μg/mL) was added and incubated at 37 °C for 36 h. The color of the culture solution changed from brownish-yellow to reddish, indicating the formation of SeNPs. The characterization of SeNPs was confirmed by UV-Vis spectrophotometry, FTIR, SEM-EDS and a particle size analyzer. The antibacterial activity was determined by the disk diffusion method, the MIC by the micro-double dilution method, and the biofilm inhibitory potential by the crystal violet method and the MTT assay. The effect of SeNPs on S. mutans ATCC 25175 was determined using SEM and CLSM spectrometry techniques. The sulfate-anthrone method was used to analyze the effect of SeNPs on insoluble extracellular polysaccharides. The expression of genes in S. mutans ATCC 25175 was analyzed by real-time quantitative polymerase chain reaction (RT-qPCR). PREPARATION OF NANOPARTICLES SeNPs produced by probiotic bacteria are considered a safe method. In this study, L. plantarum KNF-5 (probiotic strain) was used for the production of SeNPs. RESULTS The biogenic SeNPs were spherical and coated with proteins and polysaccharides and had a diameter of about 270 nm. The MIC of the SeNPs against S. mutans ATCC 25175 was 3.125 mg/mL. Biofilm growth was also significantly suppressed at this concentration. The expression of genes responsible for biofilm formation (GtfB, GtfC, BrpA and GbpB,) was reduced when S. mutans ATCC 25175 was treated with SeNPs. CONCLUSION It was concluded that the biogenic SeNPs produced by L. plantarum KNF-5 was highly effective to inhibit the growth of S. mutans ATCC 25175. NOVELTY STATEMENT The application of biogenic SeNPs, a natural anti-biofilm agent against S. mutans ATCC 25175. In the future, this study will provide a new option for the prevention and treatment of dental caries.
Collapse
Affiliation(s)
- Binbin Si
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology & Business University (BTBU), Beijing 100048, China; School of Light Industry, Beijing Technology & Business University (BTBU), Beijing 100048, China
| | - Yang Yang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology & Business University (BTBU), Beijing 100048, China; School of Light Industry, Beijing Technology & Business University (BTBU), Beijing 100048, China
| | - Muhammad Naveed
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology & Business University (BTBU), Beijing 100048, China; School of Light Industry, Beijing Technology & Business University (BTBU), Beijing 100048, China
| | - Fenghuan Wang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology & Business University (BTBU), Beijing 100048, China; School of Light Industry, Beijing Technology & Business University (BTBU), Beijing 100048, China.
| | - Malik Wajid Hussain Chan
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology & Business University (BTBU), Beijing 100048, China; School of Light Industry, Beijing Technology & Business University (BTBU), Beijing 100048, China.
| |
Collapse
|
7
|
Chamlagain M, Hu J, Sionov RV, Steinberg D. Anti-bacterial and anti-biofilm activities of arachidonic acid against the cariogenic bacterium Streptococcus mutans. Front Microbiol 2024; 15:1333274. [PMID: 38596377 PMCID: PMC11002910 DOI: 10.3389/fmicb.2024.1333274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Accepted: 02/16/2024] [Indexed: 04/11/2024] Open
Abstract
Streptococcus mutans is a Gram-positive, facultative anaerobic bacterium, which causes dental caries after forming biofilms on the tooth surface while producing organic acids that demineralize enamel and dentin. We observed that the polyunsaturated arachidonic acid (AA) (ω-6; 20:4) had an anti-bacterial activity against S. mutans, which prompted us to investigate its mechanism of action. The minimum inhibitory concentration (MIC) of AA on S. mutans was 25 μg/ml in the presence of 5% CO2, while it was reduced to 6.25-12.5 μg/ml in the absence of CO2 supplementation. The anti-bacterial action was due to a combination of bactericidal and bacteriostatic effects. The minimum biofilm inhibitory concentration (MBIC) was the same as the MIC, suggesting that part of the anti-biofilm effect was due to the anti-bacterial activity. Gene expression studies showed decreased expression of biofilm-related genes, suggesting that AA also has a specific anti-biofilm effect. Flow cytometric analyses using potentiometric DiOC2(3) dye, fluorescent efflux pump substrates, and live/dead SYTO 9/propidium iodide staining showed that AA leads to immediate membrane hyperpolarization, altered membrane transport and efflux pump activities, and increased membrane permeability with subsequent membrane perforation. High-resolution scanning electron microscopy (HR-SEM) showed remnants of burst bacteria. Furthermore, flow cytometric analysis using the redox probe 2',7'-dichlorofluorescein diacetate (DCFHDA) showed that AA acts as an antioxidant in a dose-dependent manner. α-Tocopherol, an antioxidant that terminates the radical chain, counteracted the anti-bacterial activity of AA, suggesting that oxidation of AA in bacteria leads to the production of cytotoxic radicals that contribute to bacterial growth arrest and death. Importantly, AA was not toxic to normal Vero epithelial cells even at 100 μg/ml, and it did not cause hemolysis of erythrocytes. In conclusion, our study shows that AA is a potentially safe drug that can be used to reduce the bacterial burden of cariogenic S. mutans.
Collapse
Affiliation(s)
- Manoj Chamlagain
- Institute of Biomedical and Oral Research (IBOR), The Faculty of Dental Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Biology, Hong Kong Baptist University, Kowloon, Hong Kong SAR, China
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Jieni Hu
- Institute of Biomedical and Oral Research (IBOR), The Faculty of Dental Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Ronit Vogt Sionov
- Institute of Biomedical and Oral Research (IBOR), The Faculty of Dental Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Doron Steinberg
- Institute of Biomedical and Oral Research (IBOR), The Faculty of Dental Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
8
|
Lv X, Feng Z, Luo J, Liu Z, Lu J, Han S, Wang K, Zhang L. Effects of caffeic acid phenethyl ester against multi-species cariogenic biofilms. Folia Microbiol (Praha) 2023; 68:977-989. [PMID: 37289416 DOI: 10.1007/s12223-023-01064-w] [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: 03/03/2023] [Accepted: 05/16/2023] [Indexed: 06/09/2023]
Abstract
Dental caries is a biofilm-related disease, widely perceived to be caused by oral ecological imbalance when cariogenic/aciduric bacteria obtain an ecological advantage. Compared with planktonic bacteria, dental plaques are difficult to remove under extracellular polymeric substance protection. In this study, the effect of caffeic acid phenethyl ester (CAPE) on a preformed cariogenic multi-species biofilm was evaluated, which was comprised of cariogenic bacteria (Streptococcus mutans), commensal bacteria (Streptococcus gordonii), and a pioneer colonizer (Actinomyces naeslundii). Our result revealed that treatment with 0.08 mg/mL CAPE reduced live S. mutans in the preformed multi-species biofilm while not significantly changing the quantification of live S. gordonii. CAPE significantly reduced the production of lactic acid, extracellular polysaccharide, and extracellular DNA and made the biofilm looser. Moreover, CAPE could promote the H2O2 production of S. gordonii and inhibit the expression of SMU.150 encoding mutacin to modulate the interaction among species in biofilms. Overall, our results suggested that CAPE could inhibit the cariogenic properties and change the microbial composition of the multi-species biofilms, indicating its application potential in dental caries prevention and management.
Collapse
Affiliation(s)
- Xiaohui Lv
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Disease, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3 of Renmin Road South, Chengdu, 610041, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Zening Feng
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Disease, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3 of Renmin Road South, Chengdu, 610041, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Junyuan Luo
- Department of Endodontics, College of Stomatology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhenqi Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Disease, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3 of Renmin Road South, Chengdu, 610041, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Junzhuo Lu
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Disease, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3 of Renmin Road South, Chengdu, 610041, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Sili Han
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Disease, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3 of Renmin Road South, Chengdu, 610041, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Kun Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Disease, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3 of Renmin Road South, Chengdu, 610041, China.
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| | - Linglin Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Disease, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3 of Renmin Road South, Chengdu, 610041, China.
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
9
|
Chavak Z, Mahdian N, Pakzad I, Soltani MR, Badakhsh B, Ghafourian S. In vitro anti-biofilm properties of the peel of fruite wall of acorn against Streptococcus mutans. GMS HYGIENE AND INFECTION CONTROL 2023; 18:Doc23. [PMID: 37829251 PMCID: PMC10566014 DOI: 10.3205/dgkh000449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/14/2023]
Abstract
Dental caries is a multi-factorial infectious disease. The primary cause is dental plaque, a complex of biofilm. It was postulated that the ethanolic extract of fruite wall of acorn may represent a new substance to prevent caries. Hence, the study was performed to evaluate the effect of ethanolic extract of fruite wall of acorn against biofilm formation by Streptococcus mutans, which is associated with dental plaque. The cytotoxicity of the ethanolic extract was determined against Vero cells resulting in an inhibitory concentration of 50 (IC50) of 55 µg/ml. After bacterial collection, different concentrations under the IC50 from the extract were evaluated against biofilm formation of S. mutans. 3 µg/ml of the extract inhibited the biofilm formation of S. mutans, and 1 to 3 µg/ml caused a decrease in gtfB and brpA biofilm-production genes. This study showed the potency of the ethanolic extract of fruite wall of acorn against biofilm formation by S. mutans.
Collapse
Affiliation(s)
- Zahra Chavak
- Department of Microbiology, Faculty of Medicine, Ilam University of Medical Sciences, Ilam, Iran
| | - Nahid Mahdian
- Department of Microbiology, Faculty of Medicine, Ilam University of Medical Sciences, Ilam, Iran
| | - Iraj Pakzad
- Department of Microbiology, Faculty of Medicine, Ilam University of Medical Sciences, Ilam, Iran
| | - Mohammad Reza Soltani
- Department of Operative Dentistry, School of Dentistry, Ilam University of Medical of Sciences, Ilam, Iran
| | - Behzad Badakhsh
- Department of Gastroenterology, Faculty of Medicine, IlamUniversity of Medical Sciences, Ilam, Iran
| | - Sobhan Ghafourian
- Department of Microbiology, Faculty of Medicine, Ilam University of Medical Sciences, Ilam, Iran
| |
Collapse
|
10
|
Huang X, Laird CG, Riley PP, Wen ZT. Impacts of a DUF2207 Family Protein on Streptococcus mutans Stress Tolerance Responses and Biofilm Formation. Microorganisms 2023; 11:1982. [PMID: 37630542 PMCID: PMC10457818 DOI: 10.3390/microorganisms11081982] [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: 07/07/2023] [Revised: 07/30/2023] [Accepted: 07/31/2023] [Indexed: 08/27/2023] Open
Abstract
Locus SMU.243 in Streptococcus mutans was annotated as a member of the DUF2207 family proteins highly conserved in all bacteria but with unknown function. To investigate its role in S. mutans physiology, a SMU.243-deficient mutant was constructed using allelic exchange mutagenesis, and the impacts of SMU.243 deletion on bacterial growth, stress tolerance response, and biofilm formation were analyzed. Compared to the wild-type UA159, S. mutans lacking SMU.243 displayed a reduced growth rate and a reduced overnight culture density (p < 0.01) when grown at low pH and in the presence of methyl viologen. Relative to the parent strain, the deficient mutant also had a reduced survival rate following incubation in a buffer of pH 2.8 (p < 0.01) and in a buffer containing hydrogen peroxide at 58 mM after 60 min (p < 0.001) and had a reduced capacity in biofilm formation especially in the presence of sucrose (p < 0.01). To study any ensuing functional/phenotypical links between SMU.243 and uppP, which is located immediately downstream of SMU.243 and encodes an undecaprenyl pyrophosphate phosphatase involved in recycling of carrier lipid undecaprenyl phosphate, a uppP deficient mutant was generated using allelic exchange mutagenesis. Unlike the SMU.243 mutant, deletion of uppP affected cell envelope biogenesis and caused major increases in susceptibility to bacitracin. In addition, two variant morphological mutants, one forming rough colonies and the other forming mucoid, smooth colonies, also emerged following the deletion of uppP. The results suggest that the SMU.243-encoded protein of the DUF2207 family in S. mutans plays an important role in stress tolerance response and biofilm formation, but unlike the downstream uppP, does not seem to be involved in cell envelope biogenesis, although the exact roles in S. mutans' physiology awaits further investigation.
Collapse
Affiliation(s)
- Xiaochang Huang
- Department of Oral and Craniofacial Biology, School of Dentistry, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; (X.H.); (P.P.R.)
| | - Camile G. Laird
- Department of Oral and Craniofacial Biology, School of Dentistry, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; (X.H.); (P.P.R.)
| | - Paul P. Riley
- Department of Oral and Craniofacial Biology, School of Dentistry, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; (X.H.); (P.P.R.)
| | - Zezhang Tom Wen
- Department of Oral and Craniofacial Biology, School of Dentistry, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; (X.H.); (P.P.R.)
- Department of Microbiology, Immunology and Parasitology, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| |
Collapse
|
11
|
Rose M, Wilson N, Williams E, Letner H, Bettinger R, Bouchendouka A, Batagower J, Kaspar J. Growth with Commensal Streptococci Alters Streptococcus mutans Behaviors. J Dent Res 2023; 102:450-458. [PMID: 36688378 PMCID: PMC10154915 DOI: 10.1177/00220345221145906] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
As oral bacteria grow and persist within biofilms attached to the tooth's surface, they interact with other species to form synergistic or antagonistic exchanges that govern homeostasis for the overall population. One example are the interactions between the cariogenic species Streptococcus mutans and oral commensal streptococci. Previously, we showed that the cell-cell signaling pathways of S. mutans were inhibited during coculture with other oral streptococci species, leading us to posit that the S. mutans transcriptome and behaviors are broadly altered during growth with these species. To test this hypothesis, we performed whole transcriptome sequencing (RNA-seq) on cocultures of S. mutans with either Streptococcus gordonii, Streptococcus sanguinis, or Streptococcus oralis and a quadculture containing all 4 species in comparison to S. mutans grown alone. Our results reveal that in addition to species-dependent changes to the S. mutans transcriptome, a conserved response to oral streptococci in general can be observed. We monitored the behavior of S. mutans by both microscopy imaging of biofilms and in a bacteriocin overlay assay and verified that S. mutans acts similarly with each of these species but noted divergences in phenotypes when cocultured with another cariogenic Streptococcus (Streptococcus sobrinus) or with oral nonstreptococci species. RNA-seq with oral nonstreptococci showed lack of a consistent gene expression profile and overlap of differentially expressed genes found with commensal streptococci. Finally, we investigated the role of upregulated S. mutans genes within our data sets to determine if they provided a fitness benefit during interspecies interactions. Eleven total genes were studied, and we found that a majority impacted the fitness of S. mutans in various assays, highlighted by increased biomass of commensal streptococci in mixed-species biofilms. These results confirm a common, species-independent modification of S. mutans behaviors with oral commensal streptococci that emphasizes the need to further evaluate oral bacteria within multispecies settings.
Collapse
Affiliation(s)
- M. Rose
- Division of Biosciences, The Ohio State
University College of Dentistry, Columbus, OH, USA
| | - N. Wilson
- Division of Biosciences, The Ohio State
University College of Dentistry, Columbus, OH, USA
| | - E. Williams
- Division of Biosciences, The Ohio State
University College of Dentistry, Columbus, OH, USA
| | - H. Letner
- Division of Biosciences, The Ohio State
University College of Dentistry, Columbus, OH, USA
| | - R. Bettinger
- Division of Biosciences, The Ohio State
University College of Dentistry, Columbus, OH, USA
| | - A. Bouchendouka
- Division of Biosciences, The Ohio State
University College of Dentistry, Columbus, OH, USA
| | - J. Batagower
- Division of Biosciences, The Ohio State
University College of Dentistry, Columbus, OH, USA
| | - J.R. Kaspar
- Division of Biosciences, The Ohio State
University College of Dentistry, Columbus, OH, USA
| |
Collapse
|
12
|
Antibacterial activity of oregano essential oils against Streptococcus mutans in vitro and analysis of active components. BMC Complement Med Ther 2023; 23:61. [PMID: 36810055 PMCID: PMC9942419 DOI: 10.1186/s12906-023-03890-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 02/15/2023] [Indexed: 02/23/2023] Open
Abstract
BACKGROUND Streptococcus mutans (S. mutans) is considered the most relevant bacteria during the transition of the non-pathogenic commensal oral microbial community to plaque biofilms that promote the development of dental caries. Oregano (Origanum vulgare L.), is a universally natural flavoring and its essential oil has been demonstrated to have good antibacterial effects. However, the specific antibacterial mechanism of oregano essential oil (OEO) against S. mutans is still not completely understood. METHODS In this work, the composition of two different OEOs was determined by GC‒MS. Disk-diffusion method, minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) were determined to assess their antimicrobial effect on S. mutans. The inhibition of acid production, hydrophobicity, biofilm formation and real-time PCR for gtfB/C/D, spaP, gbpB, vicR, relA and brpA mRNA expression by S. mutans were assessed to preliminarily investigate the mechanisms of action. Molecular docking was performed to simulate the interactions with the virulence proteins and active constituents. MTT test using immortalized human keratinocytes cells was also performed to investigate cytotoxicity. RESULTS Compared with the positive drug Penicillin /streptomycin 100X (DIZ: 34.13 ± 0.85 mm, MIC: 0.78125 μL/mL, MBC: 6.25 μL/mL), the essential oils of Origanum vulgare L. (DIZ: 80 mm, MIC: 0.625μL/mL, MBC:2.5μL/mL) and Origanum heracleoticum L. (DIZ: 39.67 ± 0.81 mm, MIC: 0.625μL/mL, MBC: 1.25μL/mL) could also exhibit similar effects to inhibit the acid production and reduce the hydrophobicity and biofilm formation of S. mutans at 1/2-1MIC concentration. And gene expression of gtfB/C/D, spaP, gbpB, vicR and relA were found to be downregulated. Due to the composition of essential oils from different sources being highly variable, through effective network pharmacology analysis, we found that OEOs contained many effective compounds, like carvacrol and its biosynthetic precursors γ-terpinene and p-cymene, which may directly target several virulence proteins of S. mutans. Besides, no toxic effect was instigated by OEOs at 0.1 μL/mL in the immortalized human keratinocytes cells. CONCLUSION The integrated analysis in the present study suggested that OEO might be a potential antibacterial agent for the prevention of dental caries.
Collapse
|
13
|
Zhao Z, Wu J, Sun Z, Fan J, Liu F, Zhao W, Liu WH, Zhang M, Hung WL. Postbiotics Derived from L. paracasei ET-22 Inhibit the Formation of S. mutans Biofilms and Bioactive Substances: An Analysis. Molecules 2023; 28:molecules28031236. [PMID: 36770903 PMCID: PMC9919839 DOI: 10.3390/molecules28031236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 01/19/2023] [Accepted: 01/21/2023] [Indexed: 01/31/2023] Open
Abstract
Globally, dental caries is one of the most common non-communicable diseases for patients of all ages; Streptococcus mutans (S. mutans) is its principal pathogen. Lactobacillus paracasei (L. paracasei) shows excellent anti-pathogens and immune-regulation functions in the host. The aim of this study is to evaluate the effects of L. paracasei ET-22 on the formation of S. mutans biofilms. The living bacteria, heat-killed bacteria, and secretions of L. paracasei ET-22 were prepared using the same number of bacteria. In vitro, they were added into artificial-saliva medium, and used to coculture with the S. mutans. Results showed that the living bacteria and secretions of L. paracasei ET-22 inhibited biofilm-growth, the synthesis of water-soluble polysaccharide and water-insoluble polysaccharide, and virulence-gene-expression levels related to the formation of S. mutans biofilms. Surprisingly, the heat-killed L. paracasei ET-22, which is a postbiotic, also showed a similar regulation function. Non-targeted metabonomics technology was used to identify multiple potential active-substances in the postbiotics of L. paracasei ET-22 that inhibit the formation of S. mutans biofilms, including phenyllactic acid, zidovudine monophosphate, and citrulline. In conclusion, live bacteria and its postbiotics of L. paracasei ET-22 all have inhibitory effects on the formation of S. mutans biofilm. The postbiotics of L. paracasei ET-22 may be a promising biological anticariogenic-agent.
Collapse
Affiliation(s)
- Zhi Zhao
- School of Food and Health, Beijing Technology and Business University, Beijing 100024, China
- College of Food Science and Engineering, Bohai University, Jinzhou 121013, China
| | - Jianmin Wu
- China Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China
| | - Zhe Sun
- School of Food and Health, Beijing Technology and Business University, Beijing 100024, China
| | - Jinbo Fan
- College of Food Science and Engineering, Bohai University, Jinzhou 121013, China
| | - Fudong Liu
- Inner Mongolia Dairy Technology Research Institute Co., Ltd., Hohhot 010110, China
- Inner Mongolia Yili Industrial Group Co., Ltd., Hohhot 010110, China
| | - Wen Zhao
- Inner Mongolia Dairy Technology Research Institute Co., Ltd., Hohhot 010110, China
- Inner Mongolia Yili Industrial Group Co., Ltd., Hohhot 010110, China
| | - Wei-Hsien Liu
- Inner Mongolia Dairy Technology Research Institute Co., Ltd., Hohhot 010110, China
- Inner Mongolia Yili Industrial Group Co., Ltd., Hohhot 010110, China
| | - Ming Zhang
- School of Food and Health, Beijing Technology and Business University, Beijing 100024, China
- Correspondence: (M.Z.); (W.-L.H.)
| | - Wei-Lian Hung
- Inner Mongolia Dairy Technology Research Institute Co., Ltd., Hohhot 010110, China
- Inner Mongolia Yili Industrial Group Co., Ltd., Hohhot 010110, China
- Correspondence: (M.Z.); (W.-L.H.)
| |
Collapse
|
14
|
Fixed Orthodontic Treatment Increases Cariogenicity and Virulence Gene Expression in Dental Biofilm. J Clin Med 2022; 11:jcm11195860. [PMID: 36233727 PMCID: PMC9571576 DOI: 10.3390/jcm11195860] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 09/22/2022] [Accepted: 09/29/2022] [Indexed: 11/18/2022] Open
Abstract
Background: Dental caries commonly occurs during orthodontic treatment because fixed appliances can impede effective oral hygiene practices. This study investigated the effects of fixed orthodontic treatment on dental biofilm maturity and virulence gene (gtfB, ldh, brpA, spaP, luxS, and gbpB) expression. Methods: Dental biofilms and virulence gene expression were determined in 24 orthodontic patients before and after treatment of ≥6 months. A three-tone disclosing gel was used to stain dental biofilm and assess its maturity by its color change—pink (new dental biofilm), purple (mature dental biofilm), and light blue (cariogenic dental biofilm). Gene expression levels were determined using real-time PCR. Results: After fixed orthodontic appliance insertion, the percentage of new dental biofilm decreased, whereas that of cariogenic dental biofilm significantly increased (p < 0.05). There was no significant difference in the percentage of mature dental biofilm (p > 0.05). Fixed orthodontic appliances increased gtfB, ldh, brpA, and gbpB gene expression above 1.5-fold in dental biofilm. In contrast, there was no change in spaP or luxS gene expression after treatment. Conclusions: Fixed orthodontic appliance insertion induced ecological changes and cariogenic virulence gene expression in dental biofilm.
Collapse
|
15
|
Alves-Barroco C, Botelho AMN, Américo MA, Fracalanzza SEL, de Matos APA, Guimaraes MA, Ferreira-Carvalho BT, Figueiredo AMS, Fernandes AR. Assessing in vivo and in vitro biofilm development by Streptococcus dysgalactiae subsp. dysgalactiae using a murine model of catheter-associated biofilm and human keratinocyte cell. Front Cell Infect Microbiol 2022; 12:874694. [PMID: 35928206 PMCID: PMC9343579 DOI: 10.3389/fcimb.2022.874694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Accepted: 06/29/2022] [Indexed: 11/13/2022] Open
Abstract
Streptococcus dysgalactiae subsp. dysgalactiae (SDSD) is an important agent of bovine mastitis. This infection causes an inflammatory reaction in udder tissue, being the most important disease-causing significant impact on the dairy industry. Therefore, it leads to an increase in dairy farming to meet commercial demands. As a result, there is a major impact on both the dairy industry and the environment including global warming. Recurrent mastitis is often attributed to the development of bacterial biofilms, which promote survival of sessile cells in hostile environments, and resistance to the immune system defense and antimicrobial therapy. Recently, we described the in vitro biofilm development on abiotic surfaces by bovine SDSD. In that work we integrated microbiology, imaging, and computational methods to evaluate the biofilm production capability of SDSD isolates on abiotic surfaces. Additionally, we reported that bovine SDSD can adhere and internalize human cells, including human epidermal keratinocyte (HEK) cells. We showed that the adherence and internalization rates of bovine SDSD isolates in HEK cells are higher than those of a SDSD DB49998-05 isolated from humans. In vivo, bovine SDSD can cause invasive infections leading to zebrafish morbidity and mortality. In the present work, we investigated for the first time the capability of bovine SDSD to develop biofilm in vivo using a murine animal model and ex-vivo on human HEK cells. Bovine SDSD isolates were selected based on their ability to form weak, moderate, or strong biofilms on glass surfaces. Our results showed that SDSD isolates displayed an increased ability to form biofilms on the surface of catheters implanted in mice when compared to in vitro biofilm formation on abiotic surface. A greater ability to form biofilm in vitro after animal passage was observed for the VSD45 isolate, but not for the other isolates tested. Besides that, in vitro scanning electron microscopy demonstrated that SDSD biofilm development was visible after 4 hours of SDSD adhesion to HEK cells. Cell viability tests showed an important reduction in the number of HEK cells after the formation of SDSD biofilms. In this study, the expression of genes encoding BrpA-like (biofilm regulatory protein), FbpA (fibronectin-binding protein A), HtrA (serine protease), and SagA (streptolysin S precursor) was higher for biofilm grown in vivo than in vitro, suggesting a potential role for these virulence determinants in the biofilm-development, host colonization, and SDSD infections. Taken together, these results demonstrate that SDSD can develop biofilms in vivo and on the surface of HEK cells causing important cellular damages. As SDSD infections are considered zoonotic diseases, our data contribute to a better understanding of the role of biofilm accumulation during SDSD colonization and pathogenesis not only in bovine mastitis, but they also shed some lights on the mechanisms of prosthesis-associated infection and cellulitis caused by SDSD in humans, as well.
Collapse
Affiliation(s)
- Cinthia Alves-Barroco
- UCIBIO - Applied Molecular Biosciences Unit, Dept. Ciências da Vida, NOVA School of Science and Technology, Caparica, Portugal
- i4HB, Associate Laboratory - Institute for Health and Bioeconomy, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Ana Maria Nunes Botelho
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marco Antonio Américo
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - António P. Alves de Matos
- Centro de Investigação Interdisciplinar Egas Moniz (CiiEM), Egas Moniz - Cooperativa de Ensino Superior CRL, Quinta da Granja, Portugal
| | - Márcia Aparecida Guimaraes
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Agnes Marie Sá Figueiredo
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- *Correspondence: Alexandra R. Fernandes, ; Agnes Marie Sá Figueiredo,
| | - Alexandra R. Fernandes
- UCIBIO - Applied Molecular Biosciences Unit, Dept. Ciências da Vida, NOVA School of Science and Technology, Caparica, Portugal
- i4HB, Associate Laboratory - Institute for Health and Bioeconomy, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
- *Correspondence: Alexandra R. Fernandes, ; Agnes Marie Sá Figueiredo,
| |
Collapse
|
16
|
Zhang L, Shen Y, Qiu L, Yu F, Hu X, Wang M, Sun Y, Pan Y, Zhang K. The suppression effect of SCH-79797 on Streptococcus mutans biofilm formation. J Oral Microbiol 2022; 14:2061113. [PMID: 35480051 PMCID: PMC9037171 DOI: 10.1080/20002297.2022.2061113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Background Purpose Methods and Results Conclusion
Collapse
Affiliation(s)
- Lingjun Zhang
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yan Shen
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Lili Qiu
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Fangzheng Yu
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiangyu Hu
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Min Wang
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yan Sun
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yihuai Pan
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Keke Zhang
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
17
|
Bockwoldt JA, Meng C, Ludwig C, Kupetz M, Ehrmann MA. Proteomic Analysis Reveals Enzymes for β-D-Glucan Formation and Degradation in Levilactobacillus brevis TMW 1.2112. Int J Mol Sci 2022; 23:ijms23063393. [PMID: 35328813 PMCID: PMC8951740 DOI: 10.3390/ijms23063393] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/17/2022] [Accepted: 03/18/2022] [Indexed: 02/01/2023] Open
Abstract
Bacterial exopolysaccharide (EPS) formation is crucial for biofilm formation, for protection against environmental factors, or as storage compounds. EPSs produced by lactic acid bacteria (LAB) are appropriate for applications in food fermentation or the pharmaceutical industry, yet the dynamics of formation and degradation thereof are poorly described. This study focuses on carbohydrate active enzymes, including glycosyl transferases (GT) and glycoside hydrolases (GH), and their roles in the formation and potential degradation of O2-substituted (1,3)-β-D-glucan of Levilactobacillus (L.) brevis TMW 1.2112. The fermentation broth of L. brevis TMW 1.2112 was analyzed for changes in viscosity, β-glucan, and D-glucose concentrations during the exponential, stationary, and early death phases. While the viscosity reached its maximum during the stationary phase and subsequently decreased, the β-glucan concentration only increased to a plateau. Results were correlated with secretome and proteome data to identify involved enzymes and pathways. The suggested pathway for β-glucan biosynthesis involved a β-1,3 glucan synthase (GT2) and enzymes from maltose phosphorylase (MP) operons. The decreased viscosity appeared to be associated with cell lysis as the β-glucan concentration did not decrease, most likely due to missing extracellular carbohydrate active enzymes. In addition, an operon was discovered containing known moonlighting genes, all of which were detected in both proteome and secretome samples.
Collapse
Affiliation(s)
- Julia A. Bockwoldt
- Lehrstuhl für Mikrobiologie, Technische Universität München, 85354 Freising, Germany;
| | - Chen Meng
- Bayerisches Zentrum für Biomolekulare Massenspektrometrie (BayBioMS), Technische Universität München, 85354 Freising, Germany; (C.M.); (C.L.)
| | - Christina Ludwig
- Bayerisches Zentrum für Biomolekulare Massenspektrometrie (BayBioMS), Technische Universität München, 85354 Freising, Germany; (C.M.); (C.L.)
| | - Michael Kupetz
- Lehrstuhl für Brau- und Getränketechnologie, Technische Universität München, 85354 Freising, Germany;
| | - Matthias A. Ehrmann
- Lehrstuhl für Mikrobiologie, Technische Universität München, 85354 Freising, Germany;
- Correspondence:
| |
Collapse
|
18
|
Alves-Barroco C, Rivas-García L, Fernandes AR, Baptista PV. Light Triggered Enhancement of Antibiotic Efficacy in Biofilm Elimination Mediated by Gold-Silver Alloy Nanoparticles. Front Microbiol 2022; 13:841124. [PMID: 35295305 PMCID: PMC8919054 DOI: 10.3389/fmicb.2022.841124] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 01/24/2022] [Indexed: 12/21/2022] Open
Abstract
Bacterial biofilm is a tri-dimensional complex community of cells at different metabolic stages involved in a matrix of self-produced extracellular polymeric substances. Biofilm formation is part of a defense mechanism that allows the bacteria to survive in hostile environments, such as increasing resistance or tolerance to antimicrobial agents, causing persistent infections hard to treat and impair disease eradication. One such example is bovine mastitis associated with Streptococcus dysgalactiae subsp. dysgalactiae (SDSD), whose worldwide health and economic impact is on the surge. As such, non-conventional nanobased approaches have been proposed as an alternative to tackle biofilm formation and to which pathogenic bacteria fail to adapt. Among these, metallic nanoparticles have gained significant attention, particularly gold and silver nanoparticles, due to their ease of synthesis and impact against microorganism growth. This study provides a proof-of-concept investigation into the use of gold-silver alloy nanoparticles (AuAgNPs) toward eradication of bacterial biofilms. Upon visible light irradiation of AuAgNPs there was considerable disturbance of the biofilms' matrix. The hindering of structural integrity of the biofilm matrix resulted in an increased permeability for entry of antibiotics, which then cause the eradication of biofilm and inhibit subsequent biofilm formation. Additionally, our results that AuAgNPs inhibited the formation of SDSD biofilms via distinct stress pathways that lead to the downregulation of two genes critical for biofilm production, namely, brpA-like encoding biofilm regulatory protein and fbpA fibronectin-binding protein A. This study provides useful information to assist the development of nanoparticle-based strategies for the active treatment of biofilm-related infections triggered by photoirradiation in the visible.
Collapse
Affiliation(s)
- Cinthia Alves-Barroco
- Applied Molecular Biosciences Unit, Dept. Ciências da Vida, NOVA School of Science and Technology, Costa da Caparica, Portugal
- i4HB, Associate Laboratory–Institute for Health and Bioeconomy, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Lorenzo Rivas-García
- Applied Molecular Biosciences Unit, Dept. Ciências da Vida, NOVA School of Science and Technology, Costa da Caparica, Portugal
- Biomedical Research Centre, Institute of Nutrition and Food Technology, Department of Physiology, Faculty of Pharmacy, University of Granada, Granada, Spain
| | - Alexandra R. Fernandes
- Applied Molecular Biosciences Unit, Dept. Ciências da Vida, NOVA School of Science and Technology, Costa da Caparica, Portugal
- i4HB, Associate Laboratory–Institute for Health and Bioeconomy, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Pedro Viana Baptista
- Applied Molecular Biosciences Unit, Dept. Ciências da Vida, NOVA School of Science and Technology, Costa da Caparica, Portugal
- i4HB, Associate Laboratory–Institute for Health and Bioeconomy, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| |
Collapse
|
19
|
Rajaei A, Rowe HM, Neely MN. The LCP Family Protein, Psr, Is Required for Cell Wall Integrity and Virulence in Streptococcus agalactiae. Microorganisms 2022; 10:microorganisms10020217. [PMID: 35208672 PMCID: PMC8875755 DOI: 10.3390/microorganisms10020217] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/14/2022] [Accepted: 01/15/2022] [Indexed: 02/04/2023] Open
Abstract
A robust cell envelope is the first line of protection for an infecting pathogen when encountering the immune defense of its host. In Gram-positive organisms, LytR-CpsA-Psr (LCP) family proteins play a major role in the synthesis and assembly of the cell envelope. While these proteins could be considered for potential new drug targets, not enough is known about how they function to support the integrity of the cell wall. Streptococcus agalactiae (group B streptococcus or GBS) is known to encode at least three LCP family proteins, including CpsA, LytR (BrpA) and Psr. Using strains of GBS that have mutations in two of the three LCP proteins, we were able to determine a role for these proteins in GBS cell wall integrity. The results presented here demonstrate that the absence of Psr results in a decreased growth rate, decreased viability over time, inconsistent cocci morphology and diminished cell wall integrity, as well as an increased penicillin susceptibility, decreased capsule levels and attenuation in virulence in a zebrafish model of infectious disease. A strain that is missing two of the LCP family proteins, CpsA and Psr, exhibits an increase in these defective phenotypes, indicating that CpsA and Psr are partially redundant in function.
Collapse
Affiliation(s)
- Atefeh Rajaei
- Molecular and Biomedical Sciences Department, University of Maine, Orono, ME 04469, USA;
| | - Hannah M. Rowe
- Department of Microbiology, Oregon State University, Corvallis, OR 97331, USA;
| | - Melody N. Neely
- Molecular and Biomedical Sciences Department, University of Maine, Orono, ME 04469, USA;
- Correspondence:
| |
Collapse
|
20
|
Li Y, Qiao D, Zhang Y, Hao W, Xi Y, Deng X, Ge X, Xu M. MapZ deficiency leads to defects in the envelope structure and changes stress tolerance of Streptococcus mutans. Mol Oral Microbiol 2021; 36:295-307. [PMID: 34463029 DOI: 10.1111/omi.12352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/20/2021] [Accepted: 07/28/2021] [Indexed: 11/26/2022]
Abstract
Cell division is a central process in bacteria and a prerequisite for pathogenicity. Several proteins are involved in this process to ensure the accurate localization and proper function of the division machinery. In Streptococcus mutans, MapZ marks the division sites and position of the Z-ring to regulate cell division; however, whether MapZ deficiency can impair the cariogenic virulence of S. mutans remains unclear. Here, using a phenotypic assay and RNA-seq, we investigated the role of MapZ in cell envelope maintenance, biofilm formation, and stress tolerance in S. mutans. The results show that MapZ is important for normal cell shape and envelope structure, and its deletion causes abnormal septum structure and a thin cell wall. Subsequently, we found that the absence of MapZ leads to a greater level of cell death within 12 h biofilms, but it does not seem to affect biofilm architecture and accumulation. mapZ deletion also results in a decreased acid and osmotic stress tolerance. Furthermore, RNA-seq data reveal that MapZ deficiency causes changes in the expression levels of genes involved in transport systems, sugar metabolism, nature competence, and bacteriocin synthesis. Interestingly, we found that mapZ mutation renders S. mutans more sensitive to chlorhexidine. Taken together, our study suggests that MapZ plays a role in maintaining cell envelope structure and stress tolerance in S. mutans, showing a potential application as a drug target for caries prevention.
Collapse
Affiliation(s)
- Yongliang Li
- Department of Geriatric Dentistry, Peking University Hospital of Stomatology, Beijing, P. R. China.,National Engineering Laboratory for Digital and Material Technology of Stomatology, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials, Peking University Hospital of Stomatology, Beijing, P. R. China
| | - Dan Qiao
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, Shanxi, P. R. China
| | - Yifei Zhang
- National Engineering Laboratory for Digital and Material Technology of Stomatology, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials, Peking University Hospital of Stomatology, Beijing, P. R. China.,Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, P. R. China
| | - Weifeng Hao
- National Engineering Laboratory for Digital and Material Technology of Stomatology, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials, Peking University Hospital of Stomatology, Beijing, P. R. China
| | - Yue Xi
- National Engineering Laboratory for Digital and Material Technology of Stomatology, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials, Peking University Hospital of Stomatology, Beijing, P. R. China
| | - Xuliang Deng
- Department of Geriatric Dentistry, Peking University Hospital of Stomatology, Beijing, P. R. China.,National Engineering Laboratory for Digital and Material Technology of Stomatology, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials, Peking University Hospital of Stomatology, Beijing, P. R. China
| | - Xuejun Ge
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, Shanxi, P. R. China
| | - Mingming Xu
- Department of Geriatric Dentistry, Peking University Hospital of Stomatology, Beijing, P. R. China.,National Engineering Laboratory for Digital and Material Technology of Stomatology, NMPA Key Laboratory for Dental Materials, Beijing Laboratory of Biomedical Materials, Peking University Hospital of Stomatology, Beijing, P. R. China
| |
Collapse
|
21
|
Wang Y, Hoffmann JP, Baker SM, Bentrup KHZ, Wimley WC, Fuselier JA, Bitoun JP, Morici LA. Inhibition of Streptococcus mutans biofilms with bacterial-derived outer membrane vesicles. BMC Microbiol 2021; 21:234. [PMID: 34429066 PMCID: PMC8386047 DOI: 10.1186/s12866-021-02296-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 08/13/2021] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Biofilms are microbial communities surrounded by a self-produced extracellular matrix which protects them from environmental stress. Bacteria within biofilms are 10- to 1000-fold more resistant to antibiotics, making it challenging but imperative to develop new therapeutics that can disperse biofilms and eradicate infection. Gram-negative bacteria produce outer membrane vesicles (OMV) that play critical roles in communication, genetic exchange, cargo delivery, and pathogenesis. We have previously shown that OMVs derived from Burkholderia thailandensis inhibit the growth of drug-sensitive and drug-resistant bacteria and fungi. RESULTS Here, we examine the antibiofilm activity of Burkholderia thailandensis OMVs against the oral biofilm-forming pathogen Streptococcus mutans. We demonstrate that OMV treatment reduces biofilm biomass, biofilm integrity, and bacterial cell viability. Both heat-labile and heat-stable components, including 4-hydroxy-3-methyl-2-(2-non-enyl)-quinoline and long-chain rhamnolipid, contribute to the antibiofilm activity of OMVs. When OMVs are co-administered with gentamicin, the efficacy of the antibiotic against S. mutans biofilms is enhanced. CONCLUSION These studies indicate that bacterial-derived OMVs are highly effective biological nanoparticles that can inhibit and potentially eradicate biofilms.
Collapse
Affiliation(s)
- Yihui Wang
- grid.265219.b0000 0001 2217 8588Department of Microbiology and Immunology, Tulane University School of Medicine, 1430 Tulane Ave., SL-38, LA 70112-2699 New Orleans, USA
| | - Joseph P. Hoffmann
- grid.265219.b0000 0001 2217 8588Department of Microbiology and Immunology, Tulane University School of Medicine, 1430 Tulane Ave., SL-38, LA 70112-2699 New Orleans, USA
| | - Sarah M. Baker
- grid.265219.b0000 0001 2217 8588Department of Microbiology and Immunology, Tulane University School of Medicine, 1430 Tulane Ave., SL-38, LA 70112-2699 New Orleans, USA
| | - Kerstin Höner zu Bentrup
- grid.265219.b0000 0001 2217 8588Department of Microbiology and Immunology, Tulane University School of Medicine, 1430 Tulane Ave., SL-38, LA 70112-2699 New Orleans, USA
| | - William C. Wimley
- grid.265219.b0000 0001 2217 8588Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA USA
| | - Joseph A. Fuselier
- grid.265219.b0000 0001 2217 8588Department of Medicine, Tulane University School of Medicine, New Orleans, LA USA
| | - Jacob P. Bitoun
- grid.265219.b0000 0001 2217 8588Department of Microbiology and Immunology, Tulane University School of Medicine, 1430 Tulane Ave., SL-38, LA 70112-2699 New Orleans, USA
| | - Lisa A. Morici
- grid.265219.b0000 0001 2217 8588Department of Microbiology and Immunology, Tulane University School of Medicine, 1430 Tulane Ave., SL-38, LA 70112-2699 New Orleans, USA
| |
Collapse
|
22
|
Ellepola K, Huang X, Riley RP, Bitoun JP, Wen ZT. Streptococcus mutans Lacking sufCDSUB Is Viable, but Displays Major Defects in Growth, Stress Tolerance Responses and Biofilm Formation. Front Microbiol 2021; 12:671533. [PMID: 34248879 PMCID: PMC8264796 DOI: 10.3389/fmicb.2021.671533] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 05/11/2021] [Indexed: 11/13/2022] Open
Abstract
Streptococcus mutans appears to possess a sole iron-sulfur (Fe-S) cluster biosynthesis system encoded by the sufCDSUB cluster. This study was designed to examine the role of sufCDSUB in S. mutans physiology. Allelic exchange mutants deficient of the whole sufCDSUB cluster and in individual genes were constructed. Compared to the wild-type, UA159, the sufCDSUB-deficient mutant, Δsuf::kanr, had a significantly reduced growth rate, especially in medium with the absence of isoleucine, leucine or glutamate/glutamine, amino acids that require Fe-S clusters for biosynthesis and when grown with medium adjusted to pH 6.0 and under oxidative and nitrosative stress conditions. Relative to UA159, Δsuf::kanr had major defects in stress tolerance responses with reduced survival rate of > 2-logs following incubation at low pH environment or after hydrogen peroxide challenge. When compared to UA159, Δsuf::kanr tended to form aggregates in broth medium and accumulated significantly less biofilm. As shown by luciferase reporter fusion assays, the expression of sufCDSUB was elevated by > 5.4-fold when the reporter strain was transferred from iron sufficient medium to iron-limiting medium. Oxidative stress induced by methyl viologen increased sufCDSUB expression by > 2-fold, and incubation in a low pH environment led to reduction of sufCDSUB expression by > 7-fold. These results suggest that lacking of SufCDSUB in S. mutans causes major defects in various cellular processes of the deficient mutant, including growth, stress tolerance responses and biofilm formation. In addition, the viability of the deficient mutant also suggests that SUF, the sole Fe-S cluster machinery identified is non-essential in S. mutans, which is not known in any other bacterium lacking the NIF and/or ISC system. However, how the bacterium compensates the Fe-S deficiency and if any novel Fe-S assembly systems exist in this bacterium await further investigation.
Collapse
Affiliation(s)
- Kassapa Ellepola
- Department of Oral and Craniofacial Biology, School of Dentistry, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Xiaochang Huang
- Department of Oral and Craniofacial Biology, School of Dentistry, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Ryan P Riley
- Department of Oral and Craniofacial Biology, School of Dentistry, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Jacob P Bitoun
- Department of Microbiology, Tulane University, New Orleans, LA, United States
| | - Zezhang Tom Wen
- Department of Oral and Craniofacial Biology, School of Dentistry, Louisiana State University Health Sciences Center, New Orleans, LA, United States.,Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| |
Collapse
|
23
|
Cortês IT, Rosalen PL, Berto LA, Castro ML, Pedrini DL, Porto AN, Cogo-Müller K, Nobre Franco GC. Effect of adrenaline and noradrenaline on biofilm formation and virulence factors of Streptococcus mutans UA159. Arch Oral Biol 2021; 125:105091. [PMID: 33652302 DOI: 10.1016/j.archoralbio.2021.105091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 02/19/2021] [Accepted: 02/21/2021] [Indexed: 10/22/2022]
Abstract
OBJECTIVES To evaluate in vitro the effects of adrenaline and noradrenaline on the biofilm formation on orthodontic brackets, acid production and expression of virulence genes of Streptococcus mutans UA159 (S. mutans). DESIGN S. mutans UA159 biofilm was formed on orthodontic brackets under exposure to adrenaline (100 μM), noradrenaline (50 μM) or PBS solution (control group) in triptone-yeast extract with 1 % sucrose. After 24 h, biofilm formation was quantified through Colony Forming Units / mL (CFU/mL) and RNA was extracted to perform gene expression analysis through real-time reverse transcriptase-PCR (RT-qPCR). Evaluation of acid production was carried out on planktonic cultures for 6 h. One-way ANOVA followed by Tukey's test was carried to determine statistical difference. The level of significance was set at 5 %. RESULTS Catecholamines stimulated biofilm formation of S. mutans in orthodontic brackets (p < 0,05) but did not interfere with acid production (pH reduction) or the expression of the tested genes related to biofilm formation (gtfB, gtfC, gbpA, gbpB, gbpC, gbpD and brpA), aciduric (relA) and acidogenic properties (ldh). CONCLUSIONS The present study was the first to demonstrate that catecholamines can stimulate S. mutans UA159 biofilm formation. These findings can contribute to clarify the role of stress on bacterial metabolism and contribute to the understanding of a possible role on caries development, mainly in orthodontic patients.
Collapse
Affiliation(s)
- Iago Torres Cortês
- State University of Campinas, School of Dentistry of Piracicaba, Piracicaba, SP, Brazil.
| | - Pedro Luiz Rosalen
- State University of Campinas, School of Dentistry of Piracicaba, Piracicaba, SP, Brazil; Graduate Program in Biological Sciences, Federal University of Alfenas, Alfenas, MG, Brazil.
| | - Luciana Aranha Berto
- State University of Campinas, School of Dentistry of Piracicaba, Piracicaba, SP, Brazil.
| | | | | | | | - Karina Cogo-Müller
- State University of Campinas, School of Dentistry of Piracicaba, Piracicaba, SP, Brazil; State University of Campinas, Faculty of Pharmaceutical Sciences, Campinas, SP, Brazil.
| | | |
Collapse
|
24
|
Huang W, Chen Y, Li Q, Jiang H, Lv Q, Zheng Y, Han X, Kong D, Liu P, Jiang Y. LytR plays a role in normal septum formation and contributes to full virulence in Streptococcus suis. Vet Microbiol 2021; 254:109003. [PMID: 33561639 DOI: 10.1016/j.vetmic.2021.109003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 01/24/2021] [Indexed: 10/22/2022]
Abstract
Streptococcus suis (S. suis) is a major zoonotic pathogen and is also responsible for variety of diseases in swine. LytR-CpsA-Psr (LCP) family proteins affect the biofilm formation and virulence of some Gram-positive bacteria, but we know nothing about their roles in S. suis. In this study, we constructed the LytR mutant and its revertant strains by natural transformation and verified them by PCR and western blot. We explored the effects of LytR on the cell morphology of S. suis. Transmission electron microscopic analysis showed that the mutant strain displayed aberrant septum placement with no obvious differences in capsular thickness. Crystal violet staining and laser-scanning confocal microscopy both revealed that LytR contributes to the biofilm formation of S. suis. The LytR mutant strain had reduced survival in whole human blood and was more sensitive to killing by polymorphonuclear leukocytes (PMNs). Furthermore, in a mouse infection model, the LytR mutant strain also exhibited significantly attenuated virulence and was more easily cleared in the blood. These results indicate that the LytR protein is involved in septum placement, biofilm formation and required for full virulence of S. suis during infection.
Collapse
Affiliation(s)
- Wenhua Huang
- State Key Laboratory of Pathogens and Biosecurity, Institute of Microbiology and Epidemiology, Beijing, 100071, China
| | - Ying Chen
- School of Light Industry, Beijing Technology & Business University (BTBU), Beijing, 100048, China
| | - Qian Li
- State Key Laboratory of Pathogens and Biosecurity, Institute of Microbiology and Epidemiology, Beijing, 100071, China
| | - Hua Jiang
- State Key Laboratory of Pathogens and Biosecurity, Institute of Microbiology and Epidemiology, Beijing, 100071, China
| | - Qingyu Lv
- State Key Laboratory of Pathogens and Biosecurity, Institute of Microbiology and Epidemiology, Beijing, 100071, China
| | - Yuling Zheng
- State Key Laboratory of Pathogens and Biosecurity, Institute of Microbiology and Epidemiology, Beijing, 100071, China
| | - Xuelian Han
- State Key Laboratory of Pathogens and Biosecurity, Institute of Microbiology and Epidemiology, Beijing, 100071, China
| | - Decong Kong
- State Key Laboratory of Pathogens and Biosecurity, Institute of Microbiology and Epidemiology, Beijing, 100071, China.
| | - Peng Liu
- State Key Laboratory of Pathogens and Biosecurity, Institute of Microbiology and Epidemiology, Beijing, 100071, China.
| | - Yongqiang Jiang
- State Key Laboratory of Pathogens and Biosecurity, Institute of Microbiology and Epidemiology, Beijing, 100071, China.
| |
Collapse
|
25
|
LytR-CpsA-Psr Glycopolymer Transferases: Essential Bricks in Gram-Positive Bacterial Cell Wall Assembly. Int J Mol Sci 2021; 22:ijms22020908. [PMID: 33477538 PMCID: PMC7831098 DOI: 10.3390/ijms22020908] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/14/2021] [Accepted: 01/14/2021] [Indexed: 12/28/2022] Open
Abstract
The cell walls of Gram-positive bacteria contain a variety of glycopolymers (CWGPs), a significant proportion of which are covalently linked to the peptidoglycan (PGN) scaffolding structure. Prominent CWGPs include wall teichoic acids of Staphylococcus aureus, streptococcal capsules, mycobacterial arabinogalactan, and rhamnose-containing polysaccharides of lactic acid bacteria. CWGPs serve important roles in bacterial cellular functions, morphology, and virulence. Despite evident differences in composition, structure and underlaying biosynthesis pathways, the final ligation step of CWGPs to the PGN backbone involves a conserved class of enzymes-the LytR-CpsA-Psr (LCP) transferases. Typically, the enzymes are present in multiple copies displaying partly functional redundancy and/or preference for a distinct CWGP type. LCP enzymes require a lipid-phosphate-linked glycan precursor substrate and catalyse, with a certain degree of promiscuity, CWGP transfer to PGN of different maturation stages, according to in vitro evidence. The prototype attachment mode is that to the C6-OH of N-acetylmuramic acid residues via installation of a phosphodiester bond. In some cases, attachment proceeds to N-acetylglucosamine residues of PGN-in the case of the Streptococcus agalactiae capsule, even without involvement of a phosphate bond. A novel aspect of LCP enzymes concerns a predicted role in protein glycosylation in Actinomyces oris. Available crystal structures provide further insight into the catalytic mechanism of this biologically important class of enzymes, which are gaining attention as new targets for antibacterial drug discovery to counteract the emergence of multidrug resistant bacteria.
Collapse
|
26
|
Alves-Barroco C, Paquete-Ferreira J, Santos-Silva T, Fernandes AR. Singularities of Pyogenic Streptococcal Biofilms - From Formation to Health Implication. Front Microbiol 2021; 11:584947. [PMID: 33424785 PMCID: PMC7785724 DOI: 10.3389/fmicb.2020.584947] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Accepted: 11/20/2020] [Indexed: 01/09/2023] Open
Abstract
Biofilms are generally defined as communities of cells involved in a self-produced extracellular matrix adhered to a surface. In biofilms, the bacteria are less sensitive to host defense mechanisms and antimicrobial agents, due to multiple strategies, that involve modulation of gene expression, controlled metabolic rate, intercellular communication, composition, and 3D architecture of the extracellular matrix. These factors play a key role in streptococci pathogenesis, contributing to therapy failure and promoting persistent infections. The species of the pyogenic group together with Streptococcus pneumoniae are the major pathogens belonging the genus Streptococcus, and its biofilm growth has been investigated, but insights in the genetic origin of biofilm formation are limited. This review summarizes pyogenic streptococci biofilms with details on constitution, formation, and virulence factors associated with formation.
Collapse
Affiliation(s)
- Cinthia Alves-Barroco
- UCIBIO, Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Campus de Caparica, Caparica, Portugal
| | - João Paquete-Ferreira
- UCIBIO, Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Campus de Caparica, Caparica, Portugal
| | - Teresa Santos-Silva
- UCIBIO, Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Campus de Caparica, Caparica, Portugal
| | - Alexandra R Fernandes
- UCIBIO, Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Campus de Caparica, Caparica, Portugal
| |
Collapse
|
27
|
Deciphering Streptococcal Biofilms. Microorganisms 2020; 8:microorganisms8111835. [PMID: 33233415 PMCID: PMC7700319 DOI: 10.3390/microorganisms8111835] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 11/10/2020] [Accepted: 11/17/2020] [Indexed: 12/19/2022] Open
Abstract
Streptococci are a diverse group of bacteria, which are mostly commensals but also cause a considerable proportion of life-threatening infections. They colonize many different host niches such as the oral cavity, the respiratory, gastrointestinal, and urogenital tract. While these host compartments impose different environmental conditions, many streptococci form biofilms on mucosal membranes facilitating their prolonged survival. In response to environmental conditions or stimuli, bacteria experience profound physiologic and metabolic changes during biofilm formation. While investigating bacterial cells under planktonic and biofilm conditions, various genes have been identified that are important for the initial step of biofilm formation. Expression patterns of these genes during the transition from planktonic to biofilm growth suggest a highly regulated and complex process. Biofilms as a bacterial survival strategy allow evasion of host immunity and protection against antibiotic therapy. However, the exact mechanisms by which biofilm-associated bacteria cause disease are poorly understood. Therefore, advanced molecular techniques are employed to identify gene(s) or protein(s) as targets for the development of antibiofilm therapeutic approaches. We review our current understanding of biofilm formation in different streptococci and how biofilm production may alter virulence-associated characteristics of these species. In addition, we have summarized the role of surface proteins especially pili proteins in biofilm formation. This review will provide an overview of strategies which may be exploited for developing novel approaches against biofilm-related streptococcal infections.
Collapse
|
28
|
Competence-Stimulating-Peptide-Dependent Localized Cell Death and Extracellular DNA Production in Streptococcus mutans Biofilms. Appl Environ Microbiol 2020; 86:AEM.02080-20. [PMID: 32948520 DOI: 10.1128/aem.02080-20] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 09/14/2020] [Indexed: 02/06/2023] Open
Abstract
Extracellular DNA (eDNA) is a biofilm component that contributes to the formation and structural stability of biofilms. Streptococcus mutans, a major cariogenic bacterium, induces eDNA-dependent biofilm formation under specific conditions. Since cell death can result in the release and accumulation of DNA, the dead cells in biofilms are a source of eDNA. However, it remains unknown how eDNA is released from dead cells and is localized within S. mutans biofilms. We focused on cell death induced by the extracellular signaling peptide called competence-stimulating peptide (CSP). We demonstrate that nucleic acid release into the extracellular environment occurs in a subpopulation of dead cells. eDNA production induced by CSP was highly dependent on the lytF gene, which encodes an autolysin. Although lytF expression was induced bimodally by CSP, lytF-expressing cells further divided into surviving cells and eDNA-producing dead cells. Moreover, we found that lytF-expressing cells were abundant near the bottom of the biofilm, even when all cells in the biofilm received the CSP signal. Dead cells and eDNA were also abundantly present near the bottom of the biofilm. The number of lytF-expressing cells in biofilms was significantly higher than that in planktonic cultures, which suggests that adhesion to the substratum surface is important for the induction of lytF expression. The deletion of lytF resulted in reduced adherence to a polystyrene surface. These results suggest that lytF expression and eDNA production induced near the bottom of the biofilm contribute to a firmly attached and structurally stable biofilm.IMPORTANCE Bacterial communities encased by self-produced extracellular polymeric substances (EPSs), known as biofilms, have a wide influence on human health and environmental problems. The importance of biofilm research has increased, as biofilms are the preferred bacterial lifestyle in nature. Furthermore, in recent years it has been noted that the contribution of phenotypic heterogeneity within biofilms requires analysis at the single-cell or subpopulation level to understand bacterial life strategies. In Streptococcus mutans, a cariogenic bacterium, extracellular DNA (eDNA) contributes to biofilm formation. However, it remains unclear how and where the cells produce eDNA within the biofilm. We focused on LytF, an autolysin that is induced by extracellular peptide signals. We used single-cell level imaging techniques to analyze lytF expression in the biofilm population. Here, we show that S. mutans generates eDNA by inducing lytF expression near the bottom of the biofilm, thereby enhancing biofilm adhesion and structural stability.
Collapse
|
29
|
Zhang Y, Zhu Y, Zuo Y, Tang C, Zhou F, Cui X, Wang L. Effects of Rhein-8-O-β-D-glucopyranoside on the Biofilm Formation of Streptococcus mutans. Curr Microbiol 2020; 78:323-328. [PMID: 33128581 DOI: 10.1007/s00284-020-02248-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 10/09/2020] [Indexed: 11/26/2022]
Abstract
Dental caries is the most frequent biofilm-related human infectious disease in the oral cavity. Streptococcus mutans is one of the primary etiological agents of dental caries. The aim of our study was to investigate the effects of rhein-8-O-β-D-glucopyranoside (Rg) on the development of S. mutans biofilms. Growth curves were generated, and biofilm oxygen sensitivity was detected after Rg treatment. The expression levels of luxS, brpA, ffh, recA, nth, and smx were analyzed by real-time PCR. The trypan blue exclusion assay was used to measure the effect of Rg on monocyte viability. The results showed that Rg could significantly inhibit the growth of S. mutans and suppress the biofilm formation of S. mutans in a concentration-dependent manner. In Rg-treated biofilms, the expression levels of luxS, brpA, ffh, recA, nth, and smx were all decreased. Our results further showed that Rg was nontoxic, as Rg did not affect monocyte viability or lactate dehydrogenase activity in the exposed cells. These results suggested that Rg inhibited the biofilm formation of S. mutans, and the decrease in luxS, brpA, ffh, recA, nth, and smx expression might contribute to the antibacterial effects of Rg.
Collapse
Affiliation(s)
- Yaochao Zhang
- College of Stomatology, Xi'an Medical University, South 2nd Ring Road NO.168, Yan ta District, Xi'an, Shaanxi Province, China.
| | - Yong Zhu
- College of Stomatology, Xi'an Medical University, South 2nd Ring Road NO.168, Yan ta District, Xi'an, Shaanxi Province, China
| | - Yanping Zuo
- College of Stomatology, Xi'an Medical University, South 2nd Ring Road NO.168, Yan ta District, Xi'an, Shaanxi Province, China
| | - Chengfang Tang
- College of Stomatology, Xi'an Medical University, South 2nd Ring Road NO.168, Yan ta District, Xi'an, Shaanxi Province, China
| | - Fang Zhou
- College of Stomatology, Xi'an Medical University, South 2nd Ring Road NO.168, Yan ta District, Xi'an, Shaanxi Province, China
| | - Xiaoming Cui
- College of Stomatology, Xi'an Medical University, South 2nd Ring Road NO.168, Yan ta District, Xi'an, Shaanxi Province, China
| | - Lin Wang
- College of Stomatology, Xi'an Medical University, South 2nd Ring Road NO.168, Yan ta District, Xi'an, Shaanxi Province, China
| |
Collapse
|
30
|
de Alvarenga JA, de Barros PP, de Camargo Ribeiro F, Rossoni RD, Garcia MT, Dos Santos Velloso M, Shukla S, Fuchs BB, Shukla A, Mylonakis E, Junqueira JC. Probiotic Effects of Lactobacillus paracasei 28.4 to Inhibit Streptococcus mutans in a Gellan-Based Formulation. Probiotics Antimicrob Proteins 2020; 13:506-517. [PMID: 32980974 DOI: 10.1007/s12602-020-09712-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/20/2020] [Indexed: 12/21/2022]
Abstract
Streptococcus mutans is considered to be a major bacterium involved in dental caries, and the control of virulence mechanisms is fundamental to prevent disease. Probiotics present a promising preventive method; however, the use of probiotics requires its incorporation into delivery materials to facilitate oral colonization. Thus, we performed a comprehensive study examining preventive effects of Lactobacillus paracasei 28.4-enriched gellan hydrogel materials to inhibit S. mutans in planktonic and biofilm states, addressing its influence in the production of extracellular polysaccharides (EPS) and altered gene expression of several cariogenic virulence factors. L. paracasei 28.4, a strain isolated from the oral cavity of a caries-free individual, was incorporated in three gellan hydrogels (0.5%, 0.75%, and 1% w/v). The pretreatment with probiotic-gellan formulations provided a release of L. paracasei cells over 24 h that was sufficient to inhibit the planktonic growth of S. mutans, independent of the gellan concentrations and pH variations. This pretreatment also had inhibitory activity against S. mutans biofilms, exhibiting a reduction of 0.57 to 1.54 log10 in CFU/mL (p < 0.0001) and a decrease of 68.8 to 71.3% in total biomass (p < 0.0001) compared with the control group. These inhibitory effects were associated with the decreased production of EPS by 80% (p < 0.0001) and the downregulation of luxS, brpA, gbpB, and gtfB genes. The gellan formulation containing L. paracasei 28.4 exhibited probiotic effects for preventing S. mutans growth, biofilm formation, and production of cariogenic factors to suggest possible use in tooth decay prevention.
Collapse
Affiliation(s)
- Janaína Araújo de Alvarenga
- Department of Biosciences and Oral Diagnosis, Institute of Science and Technology, São Paulo State University/UNESP, Av. Francisco José Longo 777, São Dimas, São José dos Campos, SP, 12245-000, Brazil
| | - Patrícia Pimentel de Barros
- Department of Biosciences and Oral Diagnosis, Institute of Science and Technology, São Paulo State University/UNESP, Av. Francisco José Longo 777, São Dimas, São José dos Campos, SP, 12245-000, Brazil.
| | - Felipe de Camargo Ribeiro
- Department of Biosciences and Oral Diagnosis, Institute of Science and Technology, São Paulo State University/UNESP, Av. Francisco José Longo 777, São Dimas, São José dos Campos, SP, 12245-000, Brazil
| | - Rodnei Dennis Rossoni
- Department of Biosciences and Oral Diagnosis, Institute of Science and Technology, São Paulo State University/UNESP, Av. Francisco José Longo 777, São Dimas, São José dos Campos, SP, 12245-000, Brazil
| | - Maíra Terra Garcia
- Department of Biosciences and Oral Diagnosis, Institute of Science and Technology, São Paulo State University/UNESP, Av. Francisco José Longo 777, São Dimas, São José dos Campos, SP, 12245-000, Brazil
| | - Marisol Dos Santos Velloso
- Department of Biosciences and Oral Diagnosis, Institute of Science and Technology, São Paulo State University/UNESP, Av. Francisco José Longo 777, São Dimas, São José dos Campos, SP, 12245-000, Brazil
| | - Shashank Shukla
- School of Engineering, Brown University, Providence, RI, USA
| | - Beth Burgwyn Fuchs
- Rhode Island Hospital, Alpert Medical School & Brown University, Providence, RI, USA
| | - Anita Shukla
- School of Engineering, Brown University, Providence, RI, USA
| | - Eleftherios Mylonakis
- Rhode Island Hospital, Alpert Medical School & Brown University, Providence, RI, USA
| | - Juliana Campos Junqueira
- Department of Biosciences and Oral Diagnosis, Institute of Science and Technology, São Paulo State University/UNESP, Av. Francisco José Longo 777, São Dimas, São José dos Campos, SP, 12245-000, Brazil
| |
Collapse
|
31
|
Balasubramanian AR, Vasudevan S, Shanmugam K, Lévesque CM, Solomon AP, Neelakantan P. Combinatorial effects of trans-cinnamaldehyde with fluoride and chlorhexidine on Streptococcus mutans. J Appl Microbiol 2020; 130:382-393. [PMID: 32707601 DOI: 10.1111/jam.14794] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/06/2020] [Accepted: 07/18/2020] [Indexed: 01/03/2023]
Abstract
AIMS The aim of this study was to investigate the effects of trans-cinnamaldehyde (TC) and its synergistic activity with chlorhexidine (CHX) and fluoride against Streptococcus mutans. METHODS AND RESULTS Streptococcus mutans UA159 was treated with TC alone and in combination with CHX or sodium fluoride. The synergy profile was analysed using the Zero Interaction Potency model. TC showed strong synergism (synergy score of 21·697) with CHX, but additive effect (synergy score of 5·298) with fluoride. TC and the combinations were tested for acid production (glycolytic pH drop) and biofilm formation by S. mutans, and nitric oxide production in macrophages. TC significantly inhibited sucrose-dependent biofilm formation and acid production by S. mutans. Mechanistic studies were carried out by qRT-PCR-based transcriptomic studies which showed that TC acts by impairing genes related to metabolism, quorum sensing, bacteriocin expression, stress tolerance and biofilm formation. CONCLUSIONS trans-Cinnamaldehyde potentiates CHX and sodium fluoride in inhibiting S. mutans biofilms and virulence through multiple mechanisms. This study sheds significant new light on the potential to develop TC as an anti-caries treatment. SIGNIFICANCE AND IMPACT OF THE STUDY Oral diseases were classified as a 'silent epidemic' in the US Surgeon General's Report on Oral Health. Two decades later, >4 billion people are still affected worldwide by caries, having significant effects on the quality of life. There is an urgent need to develop novel compounds and strategies to combat dental caries. Here, we prove that TC downregulates multiple pathways and potentiates the CHX and fluoride to prevent S. mutans biofilms and virulence. This study sheds significant new light on the potential to develop TC in combination with CHX or fluoride as novel treatments to arrest dental caries.
Collapse
Affiliation(s)
- A R Balasubramanian
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - S Vasudevan
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - K Shanmugam
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - C M Lévesque
- Faculty of Dentistry, University of Toronto, Toronto, ON, Canada
| | - A P Solomon
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - P Neelakantan
- Faculty of Dentistry, The University of Hong Kong, Hong Kong, Hong Kong SAR
| |
Collapse
|
32
|
Potential Risk of Spreading Resistance Genes within Extracellular-DNA-Dependent Biofilms of Streptococcus mutans in Response to Cell Envelope Stress Induced by Sub-MICs of Bacitracin. Appl Environ Microbiol 2020; 86:AEM.00770-20. [PMID: 32532873 DOI: 10.1128/aem.00770-20] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 06/08/2020] [Indexed: 02/07/2023] Open
Abstract
Antibiotics are used to treat or prevent some types of bacterial infection. The inappropriate use of antibiotics unnecessarily promotes antibiotic resistance and increases resistant bacteria, and controlling these bacteria is difficult. While the emergence of drug-resistant bacteria is a serious problem, the behavior of drug-resistant bacteria is not fully understood. In this study, we investigated the behavior of Streptococcus mutans, a major etiological agent of dental caries that is resistant to bacitracin, which is a cell wall-targeting antibiotic, and focused on biofilm formation in the presence of bacitracin. S. mutans UA159 most strongly induced extracellular DNA (eDNA)-dependent biofilm formation in the presence of bacitracin at 1/8× MIC. The ΔmbrC and ΔmbrD mutant strains, which lack bacitracin resistance, also formed biofilms in the presence of bacitracin at 1/2× MIC. This difference between the wild type and the mutants was caused by the induction of atlA expression in the mid-log phase. We also revealed that certain rgp genes involved in the synthesis of rhamnose-glucose polysaccharide related to cell wall synthesis were downregulated by bacitracin. In addition, glucosyltransferase-I was also involved in eDNA-dependent biofilm formation. The biofilm led to increased transformation efficiencies and promoted horizontal gene transfer. Biofilms were also induced by ampicillin and vancomycin, antibiotics targeting cell wall synthesis, suggesting that cell envelope stress triggers biofilm formation. Therefore, the expression of the atlA and rgp genes is regulated by S. mutans, which forms eDNA-dependent biofilms, promoting horizontal gene transfer in response to cell envelope stress induced by sub-MICs of antibiotics.IMPORTANCE Antibiotics have been reported to induce biofilm formation in many bacteria at subinhibitory concentrations. Accordingly, it is conceivable that the MIC against drug-sensitive bacteria may promote biofilm formation of resistant bacteria. Since drug-resistant bacteria have spread, it is important to understand the behavior of resistant bacteria. Streptococcus mutans is bacitracin resistant, and the 1/8× MIC of bacitracin, which is a cell wall-targeted antibiotic, induced eDNA-dependent biofilm formation. The ΔmbrC and ΔmbrD strains, which are not resistant to bacitracin, also formed biofilms in the presence of bacitracin at 1/2× MIC, and biofilms of both the wild type and mutants promoted horizontal gene transfer. Another cell wall-targeted antibiotic, vancomycin, showed effects on biofilms and gene transfer similar to those of bacitracin. Thus, treatment with cell wall-targeted antibiotics may promote the spread of drug-resistant genes in biofilms. Therefore, the behavior of resistant bacteria in the presence of antibiotics at sub-MICs should be investigated when using antibiotics.
Collapse
|
33
|
Widhianingsih D, Koontongkaew S. Enhancement of cariogenic virulence properties of dental plaque in asthmatics. J Asthma 2020; 58:1051-1057. [PMID: 32249711 DOI: 10.1080/02770903.2020.1753211] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
OBJECTIVE The aim of this study was to investigate the caries risk of asthmatics in relation to acidogenicity and the expression of caries-related genes in dental plaque. METHODS A case-control study composed of 38 asthmatics (cases) and 22 controls with an age range from 6 to 60 years. Characteristics of asthma, use of medications, oral hygiene practices and dietary habits assessed by questionnaires and interviews. The dental plaque maturity evaluated using GC Tri Plaque ID Gel TM. The expression of brpA, gtfB, gbpB, ldh, luxS and spaP genes analyzed using real-time PCR. RESULTS Asthmatics had a higher percentage of mature and acidogenic plaque than immature plaque. In contrast, immature plaque was more evident in controls. Acidogenic plaque commonly occurred in patients using 1 or a combination of two medications. High frequency in meals and sweets were found in asthmatics. Real-time PCR revealed that the expression of spaP, gtfB, gbpB, ldh, brpA and luxS were enhanced in asthmatics compared with the control group. CONCLUSION An increase in acidogenic and mature plaque is found in asthmatics. The expression of spaP, gtfB, gbpB, ldh, brpA and luxS in dental plaque are upregulated in asthmatics.
Collapse
Affiliation(s)
- Dhyani Widhianingsih
- Department of Oral Biology, Faculty of Dentistry, Thammasat University (Rangsit Campus), Pathum Thani, Thailand.,Department of Pediatric Dentistry, Faculty of Dentistry, Trisakti University, Jakarta, Indonesia
| | - Sittichai Koontongkaew
- Department of Oral Biology, Faculty of Dentistry, Thammasat University (Rangsit Campus), Pathum Thani, Thailand.,International College of Dentistry, Walailak University, Bangkok, Thailand
| |
Collapse
|
34
|
Senpuku H, Tuna EB, Nagasawa R, Nakao R, Ohnishi M. The inhibitory effects of polypyrrole on the biofilm formation of Streptococcus mutans. PLoS One 2019; 14:e0225584. [PMID: 31774855 PMCID: PMC6881011 DOI: 10.1371/journal.pone.0225584] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 11/07/2019] [Indexed: 01/18/2023] Open
Abstract
Streptococcus mutans primary thrives on the biofilm formation on the tooth surface in sticky biofilms and under certain conditions can lead to carious lesions on the tooth surface. To search for a new preventive material for oral biofilm-associated diseases, including dental caries, we investigated the effects of polypyrrole, which contains an electrochemical polymer and causes protonation and incorporation of anion under low pH condition, on the biofilm formation of S. mutans and other streptococci. In this study, polypyrrole was applied in biofilm formation assays with the S. mutans strains UA159 and its gtfB and gtfC double mutant (gtfBC mutant), S. sanguinis, S. mitis and S. gordonii on human saliva and bovine serum albumin-coated 96-well microtiter plates in tryptic soy broth supplemented with 0.25% sucrose. The effects of polypyrrole on biofilm formation were quantitatively and qualitatively observed. High concentrations of polypyrrole significantly inhibited the biofilm formation of S. mutans UA159 and S. sanguinis. As an inhibition mechanism, polypyrrole attached to the surface of bacterial cells, increased chains and aggregates, and incorporated proteins involving GTF-I and GTF-SI produced by S. mutans. In contrast, the biofilm formation of gtfBC mutant, S. sanguinis, S. mitis and S. gordonii was temporarily induced by the addition of low polypyrrole concentrations on human saliva-coated plate but not on the uncoated and bovine serum albumin-coated plates. Moreover, biofilm formation depended on live cells and, likewise, specific interaction between cells and binding components in saliva. However, these biofilms were easily removed by increased frequency of water washing. In this regard, the physical and electrochemical properties in polypyrrole worked effectively in the removal of streptococci biofilms. Polypyrrole may have the potential to alter the development of biofilms associated with dental diseases.
Collapse
Affiliation(s)
- Hidenobu Senpuku
- Department of Bacteriology I, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo, Japan
- * E-mail:
| | - Elif Bahar Tuna
- Department of Pediatric Dentistry, Faculty of Dentistry, Istanbul University, Istanbul, Turky
| | - Ryo Nagasawa
- Department of Bacteriology I, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo, Japan
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Ryoma Nakao
- Department of Bacteriology I, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo, Japan
| | - Makoto Ohnishi
- Department of Bacteriology I, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo, Japan
| |
Collapse
|
35
|
Sujitha S, Vishnu US, Karthikeyan R, Sankarasubramanian J, Gunasekaran P, Rajendhran J. Genome Investigation of a Cariogenic Pathogen with Implications in Cardiovascular Diseases. Indian J Microbiol 2019; 59:451-459. [PMID: 31762508 DOI: 10.1007/s12088-019-00823-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 08/30/2019] [Indexed: 11/24/2022] Open
Abstract
The proportion of people suffering from cardiovascular diseases has risen by 34% in the last 15 years in India. Cardiomyopathy is among the many forms of CVD s present. Infection of heart muscles is the suspected etiological agent for the same. Oral pathogens gaining entry into the bloodstream are responsible for such infections. Streptococcus mutans is an oral pathogen with implications in cardiovascular diseases. Previous studies have shown certain strains of S. mutans are found predominantly within atherosclerotic plaques and extirpated valves. To decipher the genetic differences responsible for endothelial cell invasion, we have sequenced the genome of Streptococcus mutans B14. Pan-genome analysis, search for adhesion proteins through a special algorithm, and protein-protein interactions search through HPIDB have been done. Pan-genome analysis of 187 whole genomes, assemblies revealed 6965 genes in total and 918 genes forming the core gene cluster. Adhesion to the endothelial cell is a critical virulence factor distinguishing virulent and non-virulent strains. Overall, 4% of the total proteins in S. mutans B14 were categorized as adhesion proteins. Protein-protein interaction between putative adhesion proteins and Human extracellular matrix components was predicted, revealing novel interactions. A conserved gene catalyzing the synthesis of branched-chain amino acids in S. mutans B14 shows possible interaction with isoforms of cathepsin protein of the ECM. This genome sequence analysis indicates towards other proteins in the S. mutans genome, which might have a specific role to play in host cell interaction.
Collapse
Affiliation(s)
- Srinivasan Sujitha
- 1Department of Genetics, School of Biological Sciences, Madurai Kamaraj University, Madurai, Tamil Nadu 625021 India
| | - Udayakumar S Vishnu
- 1Department of Genetics, School of Biological Sciences, Madurai Kamaraj University, Madurai, Tamil Nadu 625021 India
| | - Raman Karthikeyan
- 1Department of Genetics, School of Biological Sciences, Madurai Kamaraj University, Madurai, Tamil Nadu 625021 India
| | - Jagadesan Sankarasubramanian
- 1Department of Genetics, School of Biological Sciences, Madurai Kamaraj University, Madurai, Tamil Nadu 625021 India
| | | | - Jeyaprakash Rajendhran
- 1Department of Genetics, School of Biological Sciences, Madurai Kamaraj University, Madurai, Tamil Nadu 625021 India
| |
Collapse
|
36
|
Patras KA, Derieux J, Al-Bassam MM, Adiletta N, Vrbanac A, Lapek JD, Zengler K, Gonzalez DJ, Nizet V. Group B Streptococcus Biofilm Regulatory Protein A Contributes to Bacterial Physiology and Innate Immune Resistance. J Infect Dis 2019; 218:1641-1652. [PMID: 29868829 DOI: 10.1093/infdis/jiy341] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 06/02/2018] [Indexed: 12/18/2022] Open
Abstract
Background Streptococcus agalactiae (group B Streptococcus [GBS]) asymptomatically colonizes approximately 20% of adults; however, GBS causes severe disease in susceptible populations, including newborns, pregnant women, and elderly individuals. In shifting between commensal and pathogenic states, GBS reveals multiple mechanisms of virulence factor control. Here we describe a GBS protein that we named "biofilm regulatory protein A" (BrpA) on the basis of its homology with BrpA from Streptococcus mutans. Methods We coupled phenotypic assays, RNA sequencing, human neutrophil and whole-blood killing assays, and murine infection models to investigate the contribution of BrpA to GBS physiology and virulence. Results Sequence analysis identified BrpA as a LytR-CpsA-Psr enzyme. Targeted mutagenesis yielded a GBS mutant (ΔbrpA) with normal ultrastructural morphology but a 6-fold increase in chain length, a biofilm defect, and decreased acid tolerance. GBS ΔbrpA stimulated increased neutrophil reactive oxygen species and proved more susceptible to human and murine blood and neutrophil killing. Notably, the wild-type parent outcompeted ΔbrpA GBS in murine sepsis and vaginal colonization models. RNA sequencing of ΔbrpA uncovered multiple differences from the wild-type parent, including pathways of cell wall synthesis and cellular metabolism. Conclusions We propose that BrpA is an important virulence regulator and potential target for design of novel antibacterial therapeutics against GBS.
Collapse
Affiliation(s)
- Kathryn A Patras
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California-San Diego, La Jolla
| | - Jaclyn Derieux
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California-San Diego, La Jolla
| | - Mahmoud M Al-Bassam
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California-San Diego, La Jolla
| | - Nichole Adiletta
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California-San Diego, La Jolla
| | - Alison Vrbanac
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California-San Diego, La Jolla
| | - John D Lapek
- Department of Pharmacology, School of Medicine, University of California-San Diego, La Jolla.,Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California-San Diego, La Jolla
| | - Karsten Zengler
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California-San Diego, La Jolla
| | - David J Gonzalez
- Department of Pharmacology, School of Medicine, University of California-San Diego, La Jolla.,Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California-San Diego, La Jolla
| | - Victor Nizet
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California-San Diego, La Jolla.,Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California-San Diego, La Jolla
| |
Collapse
|
37
|
Emergent Properties in Streptococcus mutans Biofilms Are Controlled through Adhesion Force Sensing by Initial Colonizers. mBio 2019; 10:mBio.01908-19. [PMID: 31506311 PMCID: PMC6737243 DOI: 10.1128/mbio.01908-19] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Bacterial adhesion is accompanied by altered gene expression, leading to "emergent" properties of biofilm bacteria that are alien to planktonic ones. With the aim of revealing the role of environmental adhesion forces in emergent biofilm properties, genes in Streptococcus mutans UA159 and a quorum-sensing-deficient mutant were identified that become expressed after adhesion to substratum surfaces. Using atomic force microscopy, adhesion forces of initial S. mutans colonizers on four different substrata were determined and related to gene expression. Adhesion forces upon initial contact were similarly low across different substrata, ranging between 0.2 and 1.2 nN regardless of the strain considered. Bond maturation required up to 21 s, depending on the strain and substratum surface involved, but stationary adhesion forces also were similar in the parent and in the mutant strain. However, stationary adhesion forces were largest on hydrophobic silicone rubber (19 to 20 nN), while being smallest on hydrophilic glass (3 to 4 nN). brpA gene expression in thin (34 to 48 μm) 5-h S. mutans UA159 biofilms was most sensitive to adhesion forces, while expression of gbpB and comDE expressions was weakly sensitive. ftf, gtfB, vicR, and relA expression was insensitive to adhesion forces. In thicker (98 to 151 μm) 24-h biofilms, adhesion-force-induced gene expression and emergent extracellular polymeric substance (EPS) production were limited to the first 20 to 30 μm above a substratum surface. In the quorum-sensing-deficient S. mutans, adhesion-force-controlled gene expression was absent in both 5- and 24-h biofilms. Thus, initial colonizers of substratum surfaces sense adhesion forces that externally trigger emergent biofilm properties over a limited distance above a substratum surface through quorum sensing.IMPORTANCE A new concept in biofilm science is introduced: "adhesion force sensitivity of genes," defining the degree up to which expression of different genes in adhering bacteria is controlled by the environmental adhesion forces they experience. Analysis of gene expression as a function of height in a biofilm showed that the information about the substratum surface to which initially adhering bacteria adhere is passed up to a biofilm height of 20 to 30 μm above a substratum surface, highlighting the importance and limitations of cell-to-cell communication in a biofilm. Bacteria in a biofilm mode of growth, as opposed to planktonic growth, are responsible for the great majority of human infections, predicted to become the number one cause of death in 2050. The concept of adhesion force sensitivity of genes provides better understanding of bacterial adaptation in biofilms, direly needed for the design of improved therapeutic measures that evade the recalcitrance of biofilm bacteria to antimicrobials.
Collapse
|
38
|
Alves-Barroco C, Roma-Rodrigues C, Balasubramanian N, Guimarães MA, Ferreira-Carvalho BT, Muthukumaran J, Nunes D, Fortunato E, Martins R, Santos-Silva T, Figueiredo AMS, Fernandes AR, Santos-Sanches I. Biofilm development and computational screening for new putative inhibitors of a homolog of the regulatory protein BrpA in Streptococcus dysgalactiae subsp. dysgalactiae. Int J Med Microbiol 2019; 309:169-181. [PMID: 30799091 DOI: 10.1016/j.ijmm.2019.02.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Revised: 02/14/2019] [Accepted: 02/17/2019] [Indexed: 01/15/2023] Open
Abstract
Streptococcus dysgalactiae subsp. dysgalactiae (SDSD), a Lancefield group C streptococci (GCS), is a frequent cause of bovine mastitis. This highly prevalent disease is the costliest in dairy industry. Adherence and biofilm production are important factors in streptoccocal pathogenesis. We have previously described the adhesion and internalization of SDSD isolates in human cells and now we describe the biofilm production capability of this bacterium. In this work we integrated microbiology, imaging and computational methods to evaluate the biofilm production capability of SDSD isolates; to assess the presence of biofilm regulatory protein BrpA homolog in the biofilm producers; and to predict a structural model of BrpA-like protein and its binding to putative inhibitors. Our results show that SDSD isolates form biofilms on abiotic surface such as glass (hydrophilic) and polystyrene (hydrophobic), with the strongest biofilm formation observed in glass. This ability was mainly associated with a proteinaceous extracellular matrix, confirmed by the dispersion of the biofilms after proteinase K and trypsin treatment. The biofilm formation in SDSD isolates was also confirmed by confocal laser scanning microscopy (CLSM) and scanning electron microscopy (SEM). Under SEM observation, VSD16 isolate formed cell aggregates during biofilm growth while VSD9 and VSD10 formed smooth and filmy layers. We show that brpA-like gene is present and expressed in SDSD biofilm-producing isolates and its expression levels correlated with the biofilm production capability, being more expressed in the late exponential phase of planktonic growth compared to biofilm growth. Fisetin, a known biofilm inhibitor and a putative BrpA binding molecule, dramatically inhibited biofilm formation by the SDSD isolates but did not affect planktonic growth, at the tested concentrations. Homology modeling was used to predict the 3D structure of BrpA-like protein. Using high throughput virtual screening and molecular docking, we selected five ligand molecules with strong binding affinity to the hydrophobic cleft of the protein, making them potential inhibitor candidates of the SDSD BrpA-like protein. These results warrant further investigations for developing novel strategies for SDSD anti-biofilm therapy.
Collapse
Affiliation(s)
- Cinthia Alves-Barroco
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Caparica, Portugal
| | - Catarina Roma-Rodrigues
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Caparica, Portugal
| | - Natesan Balasubramanian
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Caparica, Portugal; Department of Immunology, School of Biological Sciences, Madurai Kamaraj University, Madurai, 625021, India
| | | | | | - Jayaraman Muthukumaran
- UCIBIO, Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Caparica, Portugal
| | - Daniela Nunes
- i3N/CENIMAT, Departamento de Ciência dos Materiais, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Campus de Caparica, Caparica, Portugal
| | - Elvira Fortunato
- i3N/CENIMAT, Departamento de Ciência dos Materiais, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Campus de Caparica, Caparica, Portugal
| | - Rodrigo Martins
- i3N/CENIMAT, Departamento de Ciência dos Materiais, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Campus de Caparica, Caparica, Portugal
| | - Teresa Santos-Silva
- UCIBIO, Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Caparica, Portugal.
| | - Agnes M S Figueiredo
- Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, RJ, Brazil.
| | - Alexandra R Fernandes
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Caparica, Portugal.
| | - Ilda Santos-Sanches
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Caparica, Portugal
| |
Collapse
|
39
|
Lemos JA, Palmer SR, Zeng L, Wen ZT, Kajfasz JK, Freires IA, Abranches J, Brady LJ. The Biology of Streptococcus mutans. Microbiol Spectr 2019; 7:10.1128/microbiolspec.gpp3-0051-2018. [PMID: 30657107 PMCID: PMC6615571 DOI: 10.1128/microbiolspec.gpp3-0051-2018] [Citation(s) in RCA: 399] [Impact Index Per Article: 66.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Indexed: 12/30/2022] Open
Abstract
As a major etiological agent of human dental caries, Streptococcus mutans resides primarily in biofilms that form on the tooth surfaces, also known as dental plaque. In addition to caries, S. mutans is responsible for cases of infective endocarditis with a subset of strains being indirectly implicated with the onset of additional extraoral pathologies. During the past 4 decades, functional studies of S. mutans have focused on understanding the molecular mechanisms the organism employs to form robust biofilms on tooth surfaces, to rapidly metabolize a wide variety of carbohydrates obtained from the host diet, and to survive numerous (and frequent) environmental challenges encountered in oral biofilms. In these areas of research, S. mutans has served as a model organism for ground-breaking new discoveries that have, at times, challenged long-standing dogmas based on bacterial paradigms such as Escherichia coli and Bacillus subtilis. In addition to sections dedicated to carbohydrate metabolism, biofilm formation, and stress responses, this article discusses newer developments in S. mutans biology research, namely, how S. mutans interspecies and cross-kingdom interactions dictate the development and pathogenic potential of oral biofilms and how next-generation sequencing technologies have led to a much better understanding of the physiology and diversity of S. mutans as a species.
Collapse
Affiliation(s)
- J A Lemos
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, FL 32610
| | - S R Palmer
- Division of Biosciences, College of Dentistry, Ohio State University, Columbus, OH 43210
| | - L Zeng
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, FL 32610
| | - Z T Wen
- Department of Comprehensive Dentistry and Biomaterials and Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA 70112
| | - J K Kajfasz
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, FL 32610
| | - I A Freires
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, FL 32610
| | - J Abranches
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, FL 32610
| | - L J Brady
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, FL 32610
| |
Collapse
|
40
|
Wen ZT, Scott-Anne K, Liao S, De A, Luo M, Kovacs C, Narvaez BS, Faustoferri R, Yu Q, Taylor CM, Quivey RG. Deficiency of BrpA in Streptococcus mutans reduces virulence in rat caries model. Mol Oral Microbiol 2018; 33:353-363. [PMID: 29888871 PMCID: PMC6158100 DOI: 10.1111/omi.12230] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/07/2018] [Indexed: 01/09/2023]
Abstract
Our recent studies have shown that BrpA in Streptococcus mutans plays a critical role in cell envelope biogenesis, stress responses, and biofilm formation. In this study, a 10-species consortium was used to assess how BrpA deficiency influences the establishment, persistence, and competitiveness of S. mutans during growth in a community under conditions typical of the oral cavity. Results showed that, like the wild-type, the brpA mutant was able to colonize and establish on the surfaces tested. Relative to the wild-type, however, the brpA mutant had a reduced ability to persist and grow in the 10-species consortium (P < .001). A rat caries model was also used to examine the effect of BrpA, as well as Psr, a BrpA paralog, on S. mutans cariogenicity. The results showed no major differences in infectivity between the wild-type and the brpA and psr mutants. Unlike the wild-type, however, infection with the brpA mutant, but not the psr mutant, showed no significant differences in both total numbers of carious lesions and caries severity, compared with the control group that received bacterial growth medium (P > .05). Metagenomic and quantitative polymerase chain reaction analysis showed that S. mutans infection caused major alterations in the composition of the rats' plaque microbiota and that significantly less S. mutans was identified in the rats infected with the brpA mutant compared with those infected with the wild-type and the psr mutant. These results further suggest that BrpA plays a critical role in S. mutans pathophysiology and that BrpA has potential as a therapeutic target in the modulation of S. mutans virulence.
Collapse
Affiliation(s)
- Zezhang T. Wen
- Department of Comprehensive Dentistry and Biomaterials, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
- Center of Oral and Craniofacial Biology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Kathy Scott-Anne
- Center of Oral Biology, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Sumei Liao
- Center of Oral and Craniofacial Biology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Arpan De
- Department of Comprehensive Dentistry and Biomaterials, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Meng Luo
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Christopher Kovacs
- Center of Oral Biology, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | | | - Roberta Faustoferri
- Center of Oral Biology, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Qingzhao Yu
- Department of Biostatistics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Christopher M. Taylor
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Robert G. Quivey
- Center of Oral Biology, University of Rochester School of Medicine and Dentistry, Rochester, NY
| |
Collapse
|
41
|
De A, Jorgensen AN, Beatty WL, Lemos J, Wen ZT. Deficiency of MecA in Streptococcus mutans Causes Major Defects in Cell Envelope Biogenesis, Cell Division, and Biofilm Formation. Front Microbiol 2018; 9:2130. [PMID: 30254619 PMCID: PMC6141683 DOI: 10.3389/fmicb.2018.02130] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 08/20/2018] [Indexed: 12/28/2022] Open
Abstract
MecA is an adaptor protein that guides the ClpC/P-mediated proteolysis. A S. mutans MecA-deficient mutant was constructed by double-crossover allelic exchange and analyzed for the effects of such a deficiency on cell biology and biofilm formation. Unlike the wild-type, UA159, the mecA mutant, TW416, formed mucoid and smooth colonies, severely clumped in broth and had a reduced growth rate. Transmission electron microscopy analysis revealed that TW416 grows primarily in chains of giant “swollen” cells with multiple asymmetric septa, unlike the coccoid form of UA159. As compared to UA159, biofilm formation by TW416 was significantly reduced regardless of the carbohydrate sources used for growth (P < 0.001). Western blot analysis of TW416 whole cell lysates showed a reduced expression of the glucosyltransferase GtfC and GtfB, as well as the P1 and WapA adhesins providing an explanation for the defective biofilm formation of TW416. When analyzed by a colorimetric assay, the cell wall phosphate of the mutant murein sacculi was almost 20-fold lower than the parent strain (P < 0.001). Interestingly, however, when analyzed using immunoblotting of the murein sacculi preps with UA159 whole cell antiserum as a probe, TW416 was shown to possess significantly higher signal intensity as compared to the wild-type. There is also evidence that MecA in S. mutans is more than an adaptor protein, although how it modulates the bacterial pathophysiology, including cell envelope biogenesis, cell division, and biofilm formation awaits further investigation.
Collapse
Affiliation(s)
- Arpan De
- Department of Comprehensive Dentistry and Biomaterials, University of Florida, Gainesville, FL, United States
| | - Ashton N Jorgensen
- Department of Comprehensive Dentistry and Biomaterials, University of Florida, Gainesville, FL, United States
| | - Wandy L Beatty
- Center of Oral and Craniofacial Biology, University of Florida, Gainesville, FL, United States
| | - Jose Lemos
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, United States
| | - Zezhang T Wen
- Department of Comprehensive Dentistry and Biomaterials, University of Florida, Gainesville, FL, United States.,Department of Molecular Microbiology, Washington University in St. Louis, St. Louis, MO, United States.,Department of Oral Biology, University of Florida, Gainesville, FL, United States
| |
Collapse
|
42
|
Structural Characterization of the Lactobacillus Plantarum FlmC Protein Involved in Biofilm Formation. Molecules 2018; 23:molecules23092252. [PMID: 30181476 PMCID: PMC6225345 DOI: 10.3390/molecules23092252] [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: 08/03/2018] [Revised: 08/27/2018] [Accepted: 08/31/2018] [Indexed: 12/25/2022] Open
Abstract
Lactobacillus plantarum is one of the most predominant species in the human gut microbiota of healthy individuals. We have previously characterized some probiotic features of L. plantarum LM3, as the high resistance to different stress, the binding ability toward some extracellular matrix proteins and plasminogen and the immunomodulatory role of the surface expressed adhesin EnoA1. We have also identified the flmA, flmB and flmC genes, coding for putative proteins named FlmA, FlmB and FlmC, whose null mutations partially impaired biofilm development; the L. plantarum LM3–6 strain, carrying a deletion in flmC, showed a high rate of autolysis, supporting the hypothesis that FlmC might be involved in cell wall integrity. Here, we report the in-silico characterization of ΔTM-FlmC, a portion of the FlmC protein. The protein has been also expressed, purified and characterized by means of CD spectroscopy, ICP-mass and UHPLC-HRMS. The obtained experimental data validated the predicted model unveiling also the presence of a bound lipid molecule and of a Mg(II) ion. Overall, we provide strong evidences that ΔTM-FlmC belongs to the LytR-CpsA-Psr (LCP) family of domains and is involved in cell envelope biogenesis.
Collapse
|
43
|
Wang Y, Wang X, Jiang W, Wang K, Luo J, Li W, Zhou X, Zhang L. Antimicrobial peptide GH12 suppresses cariogenic virulence factors of Streptococcus mutans. J Oral Microbiol 2018; 10:1442089. [PMID: 29503706 PMCID: PMC5827641 DOI: 10.1080/20002297.2018.1442089] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 02/14/2018] [Indexed: 02/05/2023] Open
Abstract
Cariogenic virulence factors of Streptococcus mutans include acidogenicity, aciduricity, and extracellular polysaccharides (EPS) synthesis. The de novo designed antimicrobial peptide GH12 has shown bactericidal effects on S. mutans, but its interaction with virulence and regulatory systems of S. mutans remains to be elucidated. The objectives were to investigate the effects of GH12 on virulence factors of S. mutans, and further explore the function mechanisms at enzymatic and transcriptional levels. To avoid decrease in bacterial viability, we limited GH12 to subinhibitory levels. We evaluated effects of GH12 on acidogenicity of S. mutans by pH drop, lactic acid measurement and lactate dehydrogenase (LDH) assay, on aciduricity through survival rate at pH 5.0 and F1F0-ATPase assay, and on EPS synthesis using quantitative measurement, morphology observation, vertical distribution analyses and biomass calculation. Afterwards, we conducted quantitative real-time PCR to acquire the expression profile of related genes. GH12 at 1/2 MIC (4 mg/L) inhibited acid production, survival rate, EPS synthesis, and biofilm formation. The enzymatic activity of LDH and F1F0-ATPase was inhibited, and ldh, gtfBCD, vicR, liaR, and comDE genes were significantly downregulated. In conclusion, GH12 inhibited virulence factors of S. mutans, through reducing the activity of related enzymes, downregulating virulence genes, and inactivating specific regulatory systems.
Collapse
Affiliation(s)
- Yufei Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Centre for Oral Disease, Department of Cariology and Endodontics West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xiuqing Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Centre for Oral Disease, Department of Cariology and Endodontics West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Wentao Jiang
- State Key Laboratory of Oral Diseases & National Clinical Research Centre for Oral Disease, Department of Cariology and Endodontics West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Kun Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Centre for Oral Disease, Department of Cariology and Endodontics West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Junyuan Luo
- State Key Laboratory of Oral Diseases & National Clinical Research Centre for Oral Disease, Department of Cariology and Endodontics West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Wei Li
- State Key Laboratory of Oral Diseases & National Clinical Research Centre for Oral Disease, Department of Cariology and Endodontics West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases & National Clinical Research Centre for Oral Disease, Department of Cariology and Endodontics West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Linglin Zhang
- State Key Laboratory of Oral Diseases & National Clinical Research Centre for Oral Disease, Department of Cariology and Endodontics West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
44
|
Cao L, Zhang ZZ, Xu SB, Ma M, Wei X. Farnesol inhibits development of caries by augmenting oxygen sensitivity and suppressing virulence-associated gene expression inStreptococcus mutans. J Biomed Res 2017; 31:333-343. [PMID: 28808205 PMCID: PMC5548994 DOI: 10.7555/jbr.31.20150151] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Streptococcus mutans is a primary etiological agent of dental caries. Farnesol, as a potential antimicrobial agent, inhibits the development ofS. mutans biofilm. In this study, we hypothesized that farnesol inhibits caries development in vitro and interferes with biofilm formation by regulating virulence-associated gene expression. The inhibitory effects of farnesol to S. mutans biofilms on enamel surfaces were investigated by determining micro-hardness and calcium measurements. Additionally, the morphological changes ofS. mutans biofilms were compared using field emission scanning electron microscopy and confocal laser scanning microscopy, and the vitality and oxygen sensitivity ofS. mutans biofilms were compared using MTT assays. To investigate the molecular mechanisms of farnesol's effects, expressions of possible target genesluxS, brpA, ffh, recA, nth, and smx were analyzed using reverse-transcription polymerase chain reaction (PCR) and quantitative PCR. Farnesol-treated groups exhibited significantly higher micro-hardness on the enamel surface and lower calcium concentration of the supernatants as compared to the-untreated control. Microscopy revealed that a thinner film with less extracellular matrix formed in the farnesol-treated groups. As compared to the-untreated control, farnesol inhibited biofilm formation by 26.4% with 500 µmol/L and by 37.1% with 1,000 µmol/L (P<0.05). Last, decreased transcription levels of luxS, brpA, ffh, recA, nth, and smx genes were expressed in farnesol-treated biofilms. In vitrofarnesol inhibits caries development and S. mutans biofilm formation. The regulation of luxS, brpA, ffh, recA, nth, and smx genes may contribute to the inhibitory effects of farnesol.
Collapse
Affiliation(s)
- Li Cao
- Jiangsu Key Laboratory of Oral Diseases, Department of Operative Dentistry and Endodontics, School of Stomatology, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Zhen-Zhen Zhang
- Jiangsu Key Laboratory of Oral Diseases, Department of Operative Dentistry and Endodontics, School of Stomatology, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Shuang-Bo Xu
- Jiangsu Key Laboratory of Oral Diseases, Department of Operative Dentistry and Endodontics, School of Stomatology, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Ming Ma
- Jiangsu Key Laboratory of Oral Diseases, Department of Operative Dentistry and Endodontics, School of Stomatology, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Xin Wei
- Jiangsu Key Laboratory of Oral Diseases, Department of Operative Dentistry and Endodontics, School of Stomatology, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| |
Collapse
|
45
|
Bacusmo JM, Orsini SS, Hu J, DeMott M, Thiaville PC, Elfarash A, Paulines MJ, Rojas-Benítez D, Meineke B, Deutsch C, Iwata-Reuyl D, Limbach PA, Dedon PC, Rice KC, Shuman S, Crécy-Lagard VD. The t 6A modification acts as a positive determinant for the anticodon nuclease PrrC, and is distinctively nonessential in Streptococcus mutans. RNA Biol 2017; 15:508-517. [PMID: 28726545 DOI: 10.1080/15476286.2017.1353861] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Endoribonuclease toxins (ribotoxins) are produced by bacteria and fungi to respond to stress, eliminate non-self competitor species, or interdict virus infection. PrrC is a bacterial ribotoxin that targets and cleaves tRNALysUUU in the anticodon loop. In vitro studies suggested that the post-transcriptional modification threonylcarbamoyl adenosine (t6A) is required for PrrC activity but this prediction had never been validated in vivo. Here, by using t6A-deficient yeast derivatives, it is shown that t6A is a positive determinant for PrrC proteins from various bacterial species. Streptococcus mutans is one of the few bacteria where the t6A synthesis gene tsaE (brpB) is dispensable and its genome encodes a PrrC toxin. We had previously shown using an HPLC-based assay that the S. mutans tsaE mutant was devoid of t6A. However, we describe here a novel and a more sensitive hybridization-based t6A detection method (compared to HPLC) that showed t6A was still present in the S. mutans ΔtsaE, albeit at greatly reduced levels (93% reduced compared with WT). Moreover, mutants in 2 other S. mutans t6A synthesis genes (tsaB and tsaC) were shown to be totally devoid of the modification thus confirming its dispensability in this organism. Furthermore, analysis of t6A modification ratios and of t6A synthesis genes mRNA levels in S. mutans suggest they may be regulated by growth phase.
Collapse
Affiliation(s)
- Jo Marie Bacusmo
- a Department of Microbiology and Cell Science, IFAS , University of Florida , Gainesville , FL , USA
| | - Silvia S Orsini
- a Department of Microbiology and Cell Science, IFAS , University of Florida , Gainesville , FL , USA
| | - Jennifer Hu
- b Center for Environmental Health Sciences, Department of Biological Engineering , Massachusetts Institute of Technology , Cambridge , MA , USA
| | - Michael DeMott
- b Center for Environmental Health Sciences, Department of Biological Engineering , Massachusetts Institute of Technology , Cambridge , MA , USA
| | - Patrick C Thiaville
- a Department of Microbiology and Cell Science, IFAS , University of Florida , Gainesville , FL , USA.,c Genetics and Genomics Graduate Program , University of Florida , Gainesville , USA.,d University of Florida Genetics Institute, University of Florida , Gainesville , FL , USA
| | - Ameer Elfarash
- a Department of Microbiology and Cell Science, IFAS , University of Florida , Gainesville , FL , USA.,e Genetic Department, Faculty of Agriculture , Assiut University , Assuit , Egypt
| | - Mellie June Paulines
- f Rieveschl Laboratories for Mass Spectrometry, Department of Chemistry , University of Cincinnati , Cincinnati OH , USA
| | - Diego Rojas-Benítez
- g Centro de Regulación del Genoma. Facultad de Ciencias - Universidad de Chile , Santiago , Chile
| | - Birthe Meineke
- h Molecular Biology Program , Sloan-Kettering Institute , New York , NY , USA
| | - Chris Deutsch
- i Department of Chemistry , Portland State University , Portland , OR , USA
| | - Dirk Iwata-Reuyl
- i Department of Chemistry , Portland State University , Portland , OR , USA
| | - Patrick A Limbach
- f Rieveschl Laboratories for Mass Spectrometry, Department of Chemistry , University of Cincinnati , Cincinnati OH , USA
| | - Peter C Dedon
- b Center for Environmental Health Sciences, Department of Biological Engineering , Massachusetts Institute of Technology , Cambridge , MA , USA
| | - Kelly C Rice
- a Department of Microbiology and Cell Science, IFAS , University of Florida , Gainesville , FL , USA
| | - Stewart Shuman
- h Molecular Biology Program , Sloan-Kettering Institute , New York , NY , USA
| | - Valérie de Crécy-Lagard
- a Department of Microbiology and Cell Science, IFAS , University of Florida , Gainesville , FL , USA.,d University of Florida Genetics Institute, University of Florida , Gainesville , FL , USA
| |
Collapse
|
46
|
Liao S, De A, Thompson T, Chapman L, Bitoun JP, Yao X, Yu Q, Ma F, Wen ZT. Expression of BrpA in Streptococcus mutans is regulated by FNR-box mediated repression. Mol Oral Microbiol 2017; 32:517-525. [PMID: 28744965 DOI: 10.1111/omi.12193] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/21/2017] [Indexed: 11/30/2022]
Abstract
Our previous studies showed that brpA in Streptococcus mutans, which encodes a member of the LytR-CpsA-Psr family of proteins, can be co-transcribed with brpB upstream as a bicistronic operon, and the intergenic region also has strong promoter activity. To elucidate how brpA expression is regulated, the promoter regions were analyzed using polymerase chain reaction-based deletions and site-directed mutagenesis and a promoterless luciferase gene as a reporter. Allelic exchange mutagenesis was also used to examine genes encoding putative trans-acting factors, and the impact of such mutations on brpA expression was analyzed by reporter assays. Multiple elements in the short brpA promoter (nucleotide -1 to -344 relative to start cordon ATG) were shown to have a major impact on brpA expression, including an FNR-box, for a putative binding site of an FNR-type of transcriptional regulator. When compared with the intact brpA promoter, mutations of the highly conserved nucleotides in FNR-box from TTGATgtttAcCtt to TTACAgaaaGtTac resulted in 1362-fold increases of luciferase activity (P < .001), indicative of the FNR-box-mediated repression as a major mechanism in regulation of brpA expression. When luciferase reporter was fused to the upstream brpBA promoter (nucleotides -784 to -1144), luciferase activity was decreased by 4.5-fold (P < .001) in the brpA mutant, TW14D, and by 67.7-fold (P < .001) in the brpB mutant, JB409, compared with the wild-type, UA159. However, no such effects were observed when the reporter gene was fused to the short brpA promoter and its derivatives. These results also suggest that brpA expression in S. mutans is auto-regulated through the upstream brpBA promoter.
Collapse
Affiliation(s)
- S Liao
- Center of Oral and Craniofacial Biology, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - A De
- Department of Comprehensive Dentistry and Biomaterials, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - T Thompson
- Department of Comprehensive Dentistry and Biomaterials, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - L Chapman
- Center of Oral and Craniofacial Biology, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - J P Bitoun
- Center of Oral and Craniofacial Biology, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - X Yao
- Center of Oral and Craniofacial Biology, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Q Yu
- Department of Biostatistics, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - F Ma
- Center for Virology, University of Nebraska, Lincoln, NE, USA
| | - Z T Wen
- Center of Oral and Craniofacial Biology, Louisiana State University Health Sciences Center, New Orleans, LA, USA.,Department of Comprehensive Dentistry and Biomaterials, Louisiana State University Health Sciences Center, New Orleans, LA, USA.,Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| |
Collapse
|
47
|
Deficiency of RgpG Causes Major Defects in Cell Division and Biofilm Formation, and Deficiency of LytR-CpsA-Psr Family Proteins Leads to Accumulation of Cell Wall Antigens in Culture Medium by Streptococcus mutans. Appl Environ Microbiol 2017; 83:AEM.00928-17. [PMID: 28687645 DOI: 10.1128/aem.00928-17] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 06/29/2017] [Indexed: 12/20/2022] Open
Abstract
Streptococcus mutans is known to possess rhamnose-glucose polysaccharide (RGP), a major cell wall antigen. S. mutans strains deficient in rgpG, encoding the first enzyme of the RGP biosynthesis pathway, were constructed by allelic exchange. The rgpG deficiency had no effect on growth rate but caused major defects in cell division and altered cell morphology. Unlike the coccoid wild type, the rgpG mutant existed primarily in chains of swollen, "squarish" dividing cells. Deficiency of rgpG also causes significant reduction in biofilm formation (P < 0.01). Double and triple mutants with deficiency in brpA and/or psr, genes coding for the LytR-CpsA-Psr family proteins BrpA and Psr, which were previously shown to play important roles in cell envelope biogenesis, were constructed using the rgpG mutant. There were no major differences in growth rates between the wild-type strain and the rgpG brpA and rgpG psr double mutants, but the growth rate of the rgpG brpA psr triple mutant was reduced drastically (P < 0.001). Under transmission electron microscopy, both double mutants resembled the rgpG mutant, while the triple mutant existed as giant cells with multiple asymmetric septa. When analyzed by immunoblotting, the rgpG mutant displayed major reductions in cell wall antigens compared to the wild type, while little or no signal was detected with the double and triple mutants and the brpA and psr single mutants. These results suggest that RgpG in S. mutans plays a critical role in cell division and biofilm formation and that BrpA and Psr may be responsible for attachment of cell wall antigens to the cell envelope.IMPORTANCEStreptococcus mutans, a major etiological agent of human dental caries, produces rhamnose-glucose polysaccharide (RGP) as the major cell wall antigen. This study provides direct evidence that deficiency of RgpG, the first enzyme of the RGP biosynthesis pathway, caused major defects in cell division and morphology and reduced biofilm formation by S. mutans, indicative of a significant role of RGP in cell division and biofilm formation in S. mutans These results are novel not only in S. mutans, but also other streptococci that produce RGP. This study also shows that the LytR-CpsA-Psr family proteins BrpA and Psr in S. mutans are involved in attachment of RGP and probably other cell wall glycopolymers to the peptidoglycan. In addition, the results also suggest that BrpA and Psr may play a direct role in cell division and biofilm formation in S. mutans This study reveals new potential targets to develop anticaries therapeutics.
Collapse
|
48
|
Lan L, Bowen Z, Jiyao L. [Development of transcriptional regulators of Streptococcus mutans in cariogenic virulence]. HUA XI KOU QIANG YI XUE ZA ZHI = HUAXI KOUQIANG YIXUE ZAZHI = WEST CHINA JOURNAL OF STOMATOLOGY 2016; 34:643-646. [PMID: 28318169 DOI: 10.7518/hxkq.2016.06.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Some transcriptional regulators contribute to the expression of Streptococcus mutans (S. mutans) cariogenic virulence factors. Although the target sequence transcriptional regulators anchored on the cell wall and the molecular mechanism of the regulation of S. mutans are yet to be clarified, certain global regulators potentially associated with the cariogenicity of S. mutans have been identified. This review is about these related transcriptional regulators, their function, and possible mechanisms.
Collapse
Affiliation(s)
- Li Lan
- State Key Laboratory of Oral Diseases, Dept. of Conservative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Zhang Bowen
- State Key Laboratory of Oral Diseases, Dept. of Head and Nech Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Li Jiyao
- State Key Laboratory of Oral Diseases, Dept. of Conservative Dentistry and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| |
Collapse
|
49
|
Kaufman G, Skrtic D. Structural and recovery mechanisms of 3D dental pulp cell microtissues challenged with Streptococcusmutans in extracellular matrix environment. J Med Microbiol 2016; 65:1332-1340. [PMID: 27638752 DOI: 10.1099/jmm.0.000353] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Cariopathogen Streptococcus mutans exists in infected dental pulp of deciduous teeth and is frequently linked with heart diseases. Organotypic (3D) dental pulp stem cell (DPSC) cultures/microtissues, developed to mimic the physiological conditions in vivo, were utilized to assess the bacterial impact on their (i) 3D structural configuration and (ii) recovery mechanisms. The cultures, developed in extracellular matrix (ECM) bio-scaffold (Matrigel™), interacted with WT and GFP-tagged bacterial biofilms by permitting their infiltration through the ECM. Challenged cell constructs were visualized by F-actin/nuclei staining. Their pluripotency (Sox2) and differentiation (osteocalcin) markers were assessed by immunocytochemistry. Secreted mineral was detected by alizarin red, and 3D structural arrangements were analysed by epi-fluorescence and confocal scanning microscopy. Bacterial biofilm/ECM-embedded DPSC interactions appeared in distinct areas of the microtissues. Bacterial attachment to the cell surface occurred without evidence of invasion. Surface architecture of the challenged versus unchallenged microtissues was apparently unaltered. However, significant increases in thickness (138.42 vs 106.51 µm) and bacterial penetration were detected in challenged structures causing canal-like microstructures with various diameters (12.94 -42.88 µm) and average diameter of 20.66 to 33.42 µm per microtissue. Challenged constructs expressed pluripotency and differentiation markers and secreted the mineral. Presented model shows strong potential for assessing pulp-pathogen interactions in vivo. S. mutans infiltrated and penetrated the microtissues but did not invade the cells or compromise major cell repair mechanisms. These findings would suggest reexamining the role of S. mutans as an endodontic pathogen and investigating DPSC resistance to its pathogenicity.
Collapse
Affiliation(s)
- Gili Kaufman
- Volpe Research Center, American Dental Association Foundation, Gaithersburg, MD 20899, USA
| | - Drago Skrtic
- Volpe Research Center, American Dental Association Foundation, Gaithersburg, MD 20899, USA
| |
Collapse
|
50
|
Tsai CJY, Loh JMS, Proft T. Galleria mellonella infection models for the study of bacterial diseases and for antimicrobial drug testing. Virulence 2016; 7:214-29. [PMID: 26730990 PMCID: PMC4871635 DOI: 10.1080/21505594.2015.1135289] [Citation(s) in RCA: 506] [Impact Index Per Article: 56.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Galleria mellonella (greater wax moth or honeycomb moth) has been introduced as an alternative model to study microbial infections. G. mellonella larvae can be easily and inexpensively obtained in large numbers and are simple to use as they don't require special lab equipment. There are no ethical constraints and their short life cycle makes them ideal for large-scale studies. Although insects lack an adaptive immune response, their innate immune response shows remarkable similarities with the immune response in vertebrates. This review gives a current update of what is known about the immune system of G. mellonella and provides an extensive overview of how G. mellonella is used to study the virulence of Gram-positive and Gram-negative bacteria. In addition, the use of G. mellonella to evaluate the efficacy of antimicrobial agents and experimental phage therapy are also discussed. The review concludes with a critical assessment of the current limitatons of G. mellonella infection models.
Collapse
Affiliation(s)
- Catherine Jia-Yun Tsai
- a Department of Molecular Medicine & Pathology , School of Medical Sciences, University of Auckland , Auckland , New Zealand.,b Maurice Wilkins Center, University of Auckland , Auckland , New Zealand
| | - Jacelyn Mei San Loh
- a Department of Molecular Medicine & Pathology , School of Medical Sciences, University of Auckland , Auckland , New Zealand.,b Maurice Wilkins Center, University of Auckland , Auckland , New Zealand
| | - Thomas Proft
- a Department of Molecular Medicine & Pathology , School of Medical Sciences, University of Auckland , Auckland , New Zealand.,b Maurice Wilkins Center, University of Auckland , Auckland , New Zealand
| |
Collapse
|