1
|
Conquista CM, Braga AS, Francese MM, Ferrari CR, Silva AL, Pollo LHD, Santos PSDS, Magalhães AC. Effect of 70 Gy tumor therapeutic radiation applied intermittently or directly on microcosm biofilm composition and dental hard tissues and its potential to cause dental caries. J Dent 2025; 156:105678. [PMID: 40064461 DOI: 10.1016/j.jdent.2025.105678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 03/05/2025] [Accepted: 03/07/2025] [Indexed: 03/15/2025] Open
Abstract
OBJECTIVES This study compared total (70 Gy, one session) and intermittent (35 sessions of 2 Gy) tumor radiation protocols on tooth morphology (n=5) using Scanning Electron Microscopy-Energy Dispersive X-ray (SEM-EDX) and on microcosm biofilm microbiota (n=12) through colony-forming unit (CFU) counts for Candida spp., total microorganisms, Streptococcus mutans, and total lactobacillus. It also assessed "radiation caries" development via Transverse Microradiography (TMR, n=12). METHODS Bovine enamel and root dentin were divided into three groups (n=17): total radiation (1); intermittent radiation (2); and no radiation-control (3). Biofilm was produced using saliva from irradiated (for 1 and 2) or non-irradiated patients (for 3) (n=3 donors) combined with McBain saliva with 0.2 % sucrose for 5 days. Data were analyzed using ANOVA/Tukey, t-test, and Kruskal-Wallis/Dunn's tests (p<0.05). RESULTS S. mutans and Candida spp. were observed on irradiated dentin compared to control, but these microorganisms were absent in enamel biofilm (S. mutans only in 50 % and 12.5 % of intermittent and total irradiated enamel). Total microorganisms and lactobacillus numbers were similar between groups, except for total microorganisms in irradiated enamel vs. control (p<0.037). No significant differences in mineral loss or lesion depth were detected between protocols or tissues (p>0.05). SEM-EDX revealed slight differences in magnesium (p=0.0439) and calcium (p=0.0216) content in intermittently irradiated dentin. CONCLUSIONS Despite increased cariogenic microorganisms in irradiated biofilm, no greater susceptibility to "radiation caries" was observed under this model. CLINICAL SIGNIFICANCE Although the radiotherapy alters oral microbiota and dental tissue morphology, these changes alone do not increase radiation-induced caries risk. Other factors, as salivary changes and diet, need to be better studied.
Collapse
Affiliation(s)
- Camila Médola Conquista
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Al. Octávio Pinheiro Brisolla, 9-75, Bauru, SP, 17012-901, Brazil
| | - Aline Silva Braga
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Al. Octávio Pinheiro Brisolla, 9-75, Bauru, SP, 17012-901, Brazil; Department of Conservative Dentistry, Periodontology and Endodontology, Faculty of Medicine of the University Tübingen, University Hospital Tübingen, Germany
| | - Monique Malta Francese
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Al. Octávio Pinheiro Brisolla, 9-75, Bauru, SP, 17012-901, Brazil
| | - Carolina Ruis Ferrari
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Al. Octávio Pinheiro Brisolla, 9-75, Bauru, SP, 17012-901, Brazil
| | - André Luis Silva
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Al. Octávio Pinheiro Brisolla, 9-75, Bauru, SP, 17012-901, Brazil
| | - Luiz Henrique Dias Pollo
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Al. Octávio Pinheiro Brisolla, 9-75, Bauru, SP, 17012-901, Brazil
| | - Paulo Sérgio da Silva Santos
- Department of Surgery, Stomatology, Pathology and Radiology, Bauru School of Dentistry, University of São Paulo, Bauru, SP, Brazil
| | - Ana Carolina Magalhães
- Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo, Al. Octávio Pinheiro Brisolla, 9-75, Bauru, SP, 17012-901, Brazil.
| |
Collapse
|
2
|
Moon J, Seo K, Kwon JS. Novel two-stage expansion of Streptococcus mutans biofilm supports EPS-targeted prevention strategies for early childhood caries. NPJ Biofilms Microbiomes 2025; 11:65. [PMID: 40274812 PMCID: PMC12022157 DOI: 10.1038/s41522-025-00699-6] [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: 12/24/2024] [Accepted: 04/08/2025] [Indexed: 04/26/2025] Open
Abstract
Early childhood caries (ECC) affects nearly half of preschool children worldwide and characterized by rapid progression across multiple teeth. While Streptococcus mutans (S. mutans) is a keystone species in dental caries, its process for rapid biofilm expansion remains unclear. Using an air-solid interface model simulating the oral environment, we uncovered a novel expansion for S. mutans biofilms. Our findings reveal that S. mutans employs a distinct two-step expansion strategy. Through osmotic pressure, extracellular polymeric substances (EPS) spread and transport bacterial clusters to new sites. Subsequently, the hydroxyapatite surface enables new colony formation. Hydroxyapatite's acid-neutralization properties appear critical for bacterial growth and colonization. Despite successful EPS spreading, environments without hydroxyapatite failed to support new colony formation. These results reveal the unique pattern of rapid ECC progression in sugar-rich environments and establish EPS as a promising therapeutic target, advancing understanding of cariogenic biofilm behavior and preventative strategies for ECC prevention.
Collapse
Affiliation(s)
- Jeongmi Moon
- Department and Research Institute of Dental Biomaterials and Bioengineering, Yonsei University College of Dentistry, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
- BK21 FOUR Project, Yonsei University College of Dentistry, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Kyoungjin Seo
- Department and Research Institute of Dental Biomaterials and Bioengineering, Yonsei University College of Dentistry, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
- BK21 FOUR Project, Yonsei University College of Dentistry, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Jae-Sung Kwon
- Department and Research Institute of Dental Biomaterials and Bioengineering, Yonsei University College of Dentistry, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.
- BK21 FOUR Project, Yonsei University College of Dentistry, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.
| |
Collapse
|
3
|
Alexa VT, Fratila AD, Oancea R, Galuscan A, Balean O, Bolchis V, Buzatu BLR, Obistioiu D, Suleiman MA, Jumanca D. Molecular Docking and Experimental Analysis of Essential Oil-Based Preparations on Biofilm Formation on Orthodontic Archwires. Int J Mol Sci 2024; 25:13378. [PMID: 39769141 PMCID: PMC11678866 DOI: 10.3390/ijms252413378] [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: 10/15/2024] [Revised: 11/28/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Good oral hygiene is crucial during treatment with fixed appliances, emphasising the need for additional or alternative oral health methods during orthodontic treatment. This study investigates the effect of essential oil (EO)-based preparations on biofilm adhesion to orthodontic archwires. Five identical-sized orthodontic archwires of different materials were tested using therapeutic and preventive applications of essential oils. This study also used molecular docking to explore how essential oil compounds interact with key proteins of common oral pathogens like Staphylococcus aureus and Streptococcus mutans. We found that the constituent materials heavily influence the antimicrobial effects of essential oils on different orthodontic archwires. Stainless steel-based orthodontic archwires demonstrated the highest efficacy in antimicrobial protection against S. mutans strains (maximum BIP = 28.82% on the epoxy-coated SS). Conversely, inhibition effects in preventive applications against S. aureus were observed exclusively with titanium-molybdenum alloy orthodontic archwires across all tested emulsions (maximum BIP = 29.44%). CuNiTi alloys showed ineffectiveness in preventive treatments, as none of the EO mixtures inhibited biofilm development on this material. After biofilm contamination with S. mutans and S. aureuss strains, the ternary emulsion was most effective for four out of five orthodontic archwires. Computational analysis revealed strong binding interactions between essential oil compounds and key proteins of S. aureus and S. mutans, highlighting specific amino acid residues that are critical for these interactions. Based on the results, stainless steel with epoxy coating or TMA archwires, combined with BEO/CEO/OEO ternary mixture, are recommended for optimal antibacterial protection against biofilm formation on orthodontic archwires.
Collapse
Affiliation(s)
- Vlad Tiberiu Alexa
- Clinic of Preventive, Community Dentistry and Oral Health, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Sq. no 2, 300041 Timisoara, Romania; (V.T.A.); (A.G.); (O.B.); (V.B.); (B.L.R.B.); (D.J.)
- Translational and Experimental Clinical Research Center in Oral Health (TEXC-OH), Department of Preventive, Community Dentistry and Oral Health, Victor Babes University of Medicine and Pharmacy 14A Tu-dorVladimirescu Ave., 300173 Timisoara, Romania
| | - Aurora Doris Fratila
- Faculty of Dental Medicine, Ludwig-Maximilian-University Munich, Goethestraße 70, 80336 München, Germany;
| | - Roxana Oancea
- Clinic of Preventive, Community Dentistry and Oral Health, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Sq. no 2, 300041 Timisoara, Romania; (V.T.A.); (A.G.); (O.B.); (V.B.); (B.L.R.B.); (D.J.)
- Translational and Experimental Clinical Research Center in Oral Health (TEXC-OH), Department of Preventive, Community Dentistry and Oral Health, Victor Babes University of Medicine and Pharmacy 14A Tu-dorVladimirescu Ave., 300173 Timisoara, Romania
| | - Atena Galuscan
- Clinic of Preventive, Community Dentistry and Oral Health, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Sq. no 2, 300041 Timisoara, Romania; (V.T.A.); (A.G.); (O.B.); (V.B.); (B.L.R.B.); (D.J.)
- Translational and Experimental Clinical Research Center in Oral Health (TEXC-OH), Department of Preventive, Community Dentistry and Oral Health, Victor Babes University of Medicine and Pharmacy 14A Tu-dorVladimirescu Ave., 300173 Timisoara, Romania
| | - Octavia Balean
- Clinic of Preventive, Community Dentistry and Oral Health, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Sq. no 2, 300041 Timisoara, Romania; (V.T.A.); (A.G.); (O.B.); (V.B.); (B.L.R.B.); (D.J.)
- Translational and Experimental Clinical Research Center in Oral Health (TEXC-OH), Department of Preventive, Community Dentistry and Oral Health, Victor Babes University of Medicine and Pharmacy 14A Tu-dorVladimirescu Ave., 300173 Timisoara, Romania
| | - Vanessa Bolchis
- Clinic of Preventive, Community Dentistry and Oral Health, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Sq. no 2, 300041 Timisoara, Romania; (V.T.A.); (A.G.); (O.B.); (V.B.); (B.L.R.B.); (D.J.)
- Translational and Experimental Clinical Research Center in Oral Health (TEXC-OH), Department of Preventive, Community Dentistry and Oral Health, Victor Babes University of Medicine and Pharmacy 14A Tu-dorVladimirescu Ave., 300173 Timisoara, Romania
| | - Berivan Laura Rebeca Buzatu
- Clinic of Preventive, Community Dentistry and Oral Health, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Sq. no 2, 300041 Timisoara, Romania; (V.T.A.); (A.G.); (O.B.); (V.B.); (B.L.R.B.); (D.J.)
- Translational and Experimental Clinical Research Center in Oral Health (TEXC-OH), Department of Preventive, Community Dentistry and Oral Health, Victor Babes University of Medicine and Pharmacy 14A Tu-dorVladimirescu Ave., 300173 Timisoara, Romania
| | - Diana Obistioiu
- Faculty of Agriculture, University of Life Sciences “King Michael I” from Timisoara, Calea Aradului 119, 300645 Timisoara, Romania;
| | - Mukhtar Adeiza Suleiman
- Faculty of Life Science, Department of Biochemistry, Ahmadu Bello University, Zaria 810107, Kaduna State, Nigeria;
| | - Daniela Jumanca
- Clinic of Preventive, Community Dentistry and Oral Health, Victor Babes University of Medicine and Pharmacy, Eftimie Murgu Sq. no 2, 300041 Timisoara, Romania; (V.T.A.); (A.G.); (O.B.); (V.B.); (B.L.R.B.); (D.J.)
- Translational and Experimental Clinical Research Center in Oral Health (TEXC-OH), Department of Preventive, Community Dentistry and Oral Health, Victor Babes University of Medicine and Pharmacy 14A Tu-dorVladimirescu Ave., 300173 Timisoara, Romania
| |
Collapse
|
4
|
Meng X, Huang Y, Zhou J, Yin X, Han Q, Huo L, Lei Y. The Effect of Bovine Trypsin on the Adhesion and pH of Dental Plaque Biofilms: An In Vitro Study. Int Dent J 2024; 74:1266-1276. [PMID: 38760192 PMCID: PMC11551584 DOI: 10.1016/j.identj.2024.04.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 04/11/2024] [Accepted: 04/25/2024] [Indexed: 05/19/2024] Open
Abstract
OBJECTIVE The aim of this study was to investigate the effect of bovine trypsin on the adhesion and pH of dental plaque biofilms. METHODS A multispecies dental plaque biofilm model and a single-species dental plaque biofilm model were established in vitro. Three groups were tested: (1) blank control group (aseptic ultrapure water); (2) negative control group (1M Tris-HCl buffer, pH = 7.4); and (3) experimental group (bovine trypsin). Adhesion ability was measured using an automatic microplate reader and visualised by confocal laser scanning microscopy (CLSM). The pH was measured using a pH meter. The expression of gtfB, gtfC, and gtfD was analysed using quantitative real-time polymerase chain reaction. RESULTS Adhesion ability in the experimental group was significantly lower than that in the blank group and the negative control group (P < .05); readhesion ability in the experimental group was inhibited for a certain period of time (24-hour multispecies biofilms were inhibited from 4 to 8 hours, and the 48- and 72-hour multispecies biofilms were inhibited from 2 to 6 hours; P < .05). The decrease in pH was inhibited for a certain period of time (24-hour multispecies biofilms were inhibited from 2 to 8 hours, and the 48- and 72-hour multispecies biofilms were inhibited from 1 to 8 hours; P < .05). Expression levels of gtfB, gtfC, gtfD, and ldh in the experimental group were significantly lower than those in the blank group (P < .05). CONCLUSIONS Bacterial adhesion, and readhesion, decreasd pH, and expression of adhesion- and acid-related genes by Streptococcus mutans in biofilms could be reduced by bovine trypsin for a certain period of time.
Collapse
Affiliation(s)
- Xinhui Meng
- Department of Operative Dentistry, Preventive Dentistry and Endodontics, Kunming Medical University School and Hospital of Stomatology, Kunming 650106, China; Yunnan Key Laboratory of Stomatology, Kunming 650106, China
| | - Yinxue Huang
- Department of Operative Dentistry, Preventive Dentistry and Endodontics, Kunming Medical University School and Hospital of Stomatology, Kunming 650106, China; Yunnan Key Laboratory of Stomatology, Kunming 650106, China
| | - Jing Zhou
- Department of Operative Dentistry, Preventive Dentistry and Endodontics, Kunming Medical University School and Hospital of Stomatology, Kunming 650106, China; Yunnan Key Laboratory of Stomatology, Kunming 650106, China
| | - Xintong Yin
- Department of Operative Dentistry, Preventive Dentistry and Endodontics, Kunming Medical University School and Hospital of Stomatology, Kunming 650106, China; Yunnan Key Laboratory of Stomatology, Kunming 650106, China
| | - Qunchao Han
- Department of Operative Dentistry, Preventive Dentistry and Endodontics, Kunming Medical University School and Hospital of Stomatology, Kunming 650106, China; Yunnan Key Laboratory of Stomatology, Kunming 650106, China
| | - Lijun Huo
- Department of Operative Dentistry, Preventive Dentistry and Endodontics, Kunming Medical University School and Hospital of Stomatology, Kunming 650106, China; Yunnan Key Laboratory of Stomatology, Kunming 650106, China.
| | - Yayan Lei
- Department of Operative Dentistry, Preventive Dentistry and Endodontics, Kunming Medical University School and Hospital of Stomatology, Kunming 650106, China; Yunnan Key Laboratory of Stomatology, Kunming 650106, China
| |
Collapse
|
5
|
Jeyasri R, Muthuramalingam P, Priya A, Alexpandi R, Shanmugam NRS, Nivetha S, Shin H, Pandian SK, Ravi AV, Ramesh M. Comprehensive in vitro and in vivo evaluation of therapeutic potential of Bacopa-derived asiatic acid against a human oral pathogen Streptococcus mutans. Front Microbiol 2024; 15:1404012. [PMID: 38983632 PMCID: PMC11231090 DOI: 10.3389/fmicb.2024.1404012] [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: 03/20/2024] [Accepted: 05/20/2024] [Indexed: 07/11/2024] Open
Abstract
Dental caries is a common human oral disease worldwide, caused by an acid-producing bacteria Streptococcus mutans. The use of synthetic drugs and antibiotics to prevent dental caries has been increasing, but this can lead to severe side effects. To solve this issue, developing and developed countries have resorted to herbal medicines as an alternative to synthetic drugs for the treatment and prevention of dental caries. Therefore, there is an urgent need for plant-derived products to treat such diseases. Bacopa monnieri, a well-documented medicinal plant, contains 52 phytocompounds, including the pentacyclic triterpenoid metabolite known as asiatic acid (ASTA). Hence, this study aimed to demonstrate, for the first time, the antibacterial activity of phytocompound ASTA against S. mutans. The findings revealed that ASTA significantly inhibited the growth of S. mutans and the production of virulence factors such as acidurity, acidogenicity, and eDNA synthesis. Molecular docking analysis evaluated the potential activity of ASTA against S. mutans virulence genes, including VicR and GtfC. Furthermore, toxicity assessment of ASTA in human buccal epithelial cells was performed, and no morphological changes were observed. An in vivo analysis using Danio rerio (zebrafish) confirmed that the ASTA treatment significantly increased the survival rates of infected fish by hindering the intestinal colonization of S. mutans. Furthermore, the disease protection potential of ASTA against the pathognomonic symptom of S. mutans infection was proven by the histopathological examination of the gills, gut, and kidney. Overall, these findings suggest that ASTA may be a promising therapeutic and alternative drug for the treatment and prevention of oral infection imposed by S. mutans.
Collapse
Affiliation(s)
- Rajendran Jeyasri
- Department of Biotechnology, Science Campus, Alagappa University, Karaikudi, Tamil Nadu, India
| | - Pandiyan Muthuramalingam
- Department of Biotechnology, Science Campus, Alagappa University, Karaikudi, Tamil Nadu, India
- Division of Horticultural Science, College of Agriculture and Life Sciences, Gyeongsang National University, Jinju, South Korea
| | - Arumugam Priya
- Department of Biotechnology, Science Campus, Alagappa University, Karaikudi, Tamil Nadu, India
- Department of Medicine, Division of Gastroenterology and Hepatology, Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Rajaiah Alexpandi
- Department of Biotechnology, Science Campus, Alagappa University, Karaikudi, Tamil Nadu, India
- School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, China
| | - N. R. Siva Shanmugam
- Department of Food Science and Technology, Nebraska Food for Health Center, University of Nebraska - Lincoln, Lincoln, NE, United States
| | - Saminathan Nivetha
- Department of Biotechnology, Science Campus, Alagappa University, Karaikudi, Tamil Nadu, India
| | - Hyunsuk Shin
- Division of Horticultural Science, College of Agriculture and Life Sciences, Gyeongsang National University, Jinju, South Korea
| | | | - Arumugam Veera Ravi
- Department of Biotechnology, Science Campus, Alagappa University, Karaikudi, Tamil Nadu, India
| | - Manikandan Ramesh
- Department of Biotechnology, Science Campus, Alagappa University, Karaikudi, Tamil Nadu, India
| |
Collapse
|
6
|
Zakis DR, Brandt BW, van der Waal SV, Keijser BJF, Crielaard W, van der Plas DW, Volgenant CM, Zaura E. The effect of different sweeteners on the oral microbiome: a randomized clinical exploratory pilot study. J Oral Microbiol 2024; 16:2369350. [PMID: 38919384 PMCID: PMC11198155 DOI: 10.1080/20002297.2024.2369350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 06/07/2024] [Indexed: 06/27/2024] Open
Abstract
Introduction The aim of the study was to evaluate the modulating effects of five commonly used sweetener (glucose, inulin, isomaltulose, tagatose, trehalose) containing mouth rinses on the oral microbiome. Methods A single-centre, double-blind, parallel randomized clinical trial was performed with healthy, 18-55-year-old volunteers (N = 65), who rinsed thrice-daily for two weeks with a 10% solution of one of the allocated sweeteners. Microbiota composition of supragingival dental plaque and the tongue dorsum coating was analysed by 16S RNA gene amplicon sequencing of the V4 hypervariable region (Illumina MiSeq). As secondary outcomes, dental plaque red fluorescence and salivary pH were measured. Results Dental plaque microbiota changed significantly for two groups: inulin (F = 2.0239, p = 0.0006 PERMANOVA, Aitchison distance) and isomaltulose (F = 0.67, p = 0.0305). For the tongue microbiota, significant changes were observed for isomaltulose (F = 0.8382, p = 0.0452) and trehalose (F = 1.0119, p = 0.0098). In plaque, 13 species changed significantly for the inulin group, while for tongue coating, three species changed for the trehalose group (ALDEx2, p < 0.1). No significant changes were observed for the secondary outcomes. Conclusion The effects on the oral microbiota were sweetener dependant with the most pronounced effect on plaque microbiota. Inulin exhibited the strongest microbial modulating potential of the sweeteners tested. Further full-scale clinical studies are required.
Collapse
Affiliation(s)
- Davis R. Zakis
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam, Vrije Universiteit Amsterdam and University of Amsterdam, Amsterdam, The Netherlands
- Department of Cariology, Academic Centre for Dentistry Amsterdam, Vrije Universiteit Amsterdam and University of Amsterdam, Amsterdam, The Netherlands
| | - Bernd W. Brandt
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam, Vrije Universiteit Amsterdam and University of Amsterdam, Amsterdam, The Netherlands
| | - Suzette V. van der Waal
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam, Vrije Universiteit Amsterdam and University of Amsterdam, Amsterdam, The Netherlands
| | - Bart J. F. Keijser
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam, Vrije Universiteit Amsterdam and University of Amsterdam, Amsterdam, The Netherlands
- Research Group Microbiology and Systems Biology, TNO, Leiden, The Netherlands
| | - Wim Crielaard
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam, Vrije Universiteit Amsterdam and University of Amsterdam, Amsterdam, The Netherlands
| | - Derek W.K. van der Plas
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam, Vrije Universiteit Amsterdam and University of Amsterdam, Amsterdam, The Netherlands
| | - Catherine M.C. Volgenant
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam, Vrije Universiteit Amsterdam and University of Amsterdam, Amsterdam, The Netherlands
- Department of Cariology, Academic Centre for Dentistry Amsterdam, Vrije Universiteit Amsterdam and University of Amsterdam, Amsterdam, The Netherlands
| | - Egija Zaura
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam, Vrije Universiteit Amsterdam and University of Amsterdam, Amsterdam, The Netherlands
- Department of Cariology, Academic Centre for Dentistry Amsterdam, Vrije Universiteit Amsterdam and University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
7
|
Lu L, Zhao Y, Li M, Wang X, Zhu J, Liao L, Wang J. Contemporary strategies and approaches for characterizing composition and enhancing biofilm penetration targeting bacterial extracellular polymeric substances. J Pharm Anal 2024; 14:100906. [PMID: 38634060 PMCID: PMC11022105 DOI: 10.1016/j.jpha.2023.11.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 11/08/2023] [Accepted: 11/26/2023] [Indexed: 04/19/2024] Open
Abstract
Extracellular polymeric substances (EPS) constitutes crucial elements within bacterial biofilms, facilitating accelerated antimicrobial resistance and conferring defense against the host's immune cells. Developing precise and effective antibiofilm approaches and strategies, tailored to the specific characteristics of EPS composition, can offer valuable insights for the creation of novel antimicrobial drugs. This, in turn, holds the potential to mitigate the alarming issue of bacterial drug resistance. Current analysis of EPS compositions relies heavily on colorimetric approaches with a significant bias, which is likely due to the selection of a standard compound and the cross-interference of various EPS compounds. Considering the pivotal role of EPS in biofilm functionality, it is imperative for EPS research to delve deeper into the analysis of intricate compositions, moving beyond the current focus on polymeric materials. This necessitates a shift from heavy reliance on colorimetric analytic methods to more comprehensive and nuanced analytical approaches. In this study, we have provided a comprehensive summary of existing analytical methods utilized in the characterization of EPS compositions. Additionally, novel strategies aimed at targeting EPS to enhance biofilm penetration were explored, with a specific focus on highlighting the limitations associated with colorimetric methods. Furthermore, we have outlined the challenges faced in identifying additional components of EPS and propose a prospective research plan to address these challenges. This review has the potential to guide future researchers in the search for novel compounds capable of suppressing EPS, thereby inhibiting biofilm formation. This insight opens up a new avenue for exploration within this research domain.
Collapse
Affiliation(s)
- Lan Lu
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education Department, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, 610000, China
| | - Yuting Zhao
- Meishan Pharmaceutical Vocational College, School of Pharmacy, Meishan, Sichuan, 620200, China
| | - Mingxing Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Xiaobo Wang
- Hepatobiliary Surgery, Langzhong People's Hospital, Langzhong, Sichuan, 646000, China
| | - Jie Zhu
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education Department, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, 610000, China
| | - Li Liao
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education Department, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, 610000, China
| | - Jingya Wang
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education Department, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, 610000, China
| |
Collapse
|
8
|
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
|
9
|
Luo SC, Wei SM, Luo XT, Yang QQ, Wong KH, Cheung PCK, Zhang BB. How probiotics, prebiotics, synbiotics, and postbiotics prevent dental caries: an oral microbiota perspective. NPJ Biofilms Microbiomes 2024; 10:14. [PMID: 38402294 PMCID: PMC10894247 DOI: 10.1038/s41522-024-00488-7] [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: 12/07/2023] [Accepted: 02/12/2024] [Indexed: 02/26/2024] Open
Abstract
Dental caries, a highly prevalent oral disease, impacts a significant portion of the global population. Conventional approaches that indiscriminately eradicate microbes disrupt the natural equilibrium of the oral microbiota. In contrast, biointervention strategies aim to restore this balance by introducing beneficial microorganisms or inhibiting cariogenic ones. Over the past three decades, microbial preparations have garnered considerable attention in dental research for the prevention and treatment of dental caries. However, unlike related pathologies in the gastrointestinal, vaginal, and respiratory tracts, dental caries occurs on hard tissues such as tooth enamel and is closely associated with localized acid overproduction facilitated by cariogenic biofilms. Therefore, it is insufficient to rely solely on previous mechanisms to delineate the role of microbial preparations in the oral cavity. A more comprehensive perspective should involve considering the concepts of cariogenic biofilms. This review elucidates the latest research progress, mechanisms of action, challenges, and future research directions regarding probiotics, prebiotics, synbiotics, and postbiotics for the prevention and treatment of dental caries, taking into account the unique pathogenic mechanisms of dental caries. With an enhanced understanding of oral microbiota, personalized microbial therapy will emerge as a critical future research trend.
Collapse
Affiliation(s)
- Si-Chen Luo
- Guangdong Provincial Key Laboratory of Marine Biology, Department of Biology, College of Science, Shantou University, Shantou, 515063, Guangdong, PR China
| | - Si-Min Wei
- Guangdong Provincial Key Laboratory of Marine Biology, Department of Biology, College of Science, Shantou University, Shantou, 515063, Guangdong, PR China
| | - Xin-Tao Luo
- Guangdong Provincial Key Laboratory of Marine Biology, Department of Biology, College of Science, Shantou University, Shantou, 515063, Guangdong, PR China
| | - Qiong-Qiong Yang
- Guangdong Provincial Key Laboratory of Marine Biology, Department of Biology, College of Science, Shantou University, Shantou, 515063, Guangdong, PR China
| | - Ka-Hing Wong
- Research Institute for Future Food, Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hong Kong, PR China
| | - Peter C K Cheung
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, PR China
| | - Bo-Bo Zhang
- Guangdong Provincial Key Laboratory of Marine Biology, Department of Biology, College of Science, Shantou University, Shantou, 515063, Guangdong, PR China.
| |
Collapse
|
10
|
Kaur P, Vyas M, Sharma S. Dental Caries: Unveiling the State-of-the-art Insights and Crafting Hypotheses for Oral Health. Curr Pharm Des 2024; 30:2667-2670. [PMID: 38994613 DOI: 10.2174/0113816128318101240708095951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 06/16/2024] [Indexed: 07/13/2024]
Abstract
The pathophysiological understanding of dental caries explains that the primary factor responsible is linked to an imbalance in microbial composition within the oral cavity, stemming from both artificial and natural sources. Streptococcus mutans (S. mutans) is the most accountable and prevalent pathogen for caries development among the diverse pool. S. mutans, an acidogenic bacterium, lowers oral pH through the metabolic conversion of dietary sugar into organic acids, leading to enamel demineralization and dental caries. Numerous antibacterial interventions have been employed in the past to address this issue. However, adopting such an approach poses the risk of exacerbating concerns related to Antimicrobial Resistance (AMR) and long-term oral cytotoxicity. In response to this, a sustainable strategy is suggested, involving the utilization of L-Arginine (L-Arg) as a probiotic nutrient supplement for non-pathogenic microbes. It will help in creating a natural competitive environment against the pathogenic microbes responsible for initiating dental caries. The hypothesis involves utilizing a combination of a nutrient supplement and the repurposed drug Piceatannol, specifically for its anti-biofilm properties. This combination synergistically improves the effectiveness of the therapy by converting the complex microbial biofilm into a planktonic state.
Collapse
Affiliation(s)
- Palwinder Kaur
- Department of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Manish Vyas
- Department of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Sandeep Sharma
- Department of Medical Laboratory Sciences, Lovely Professional University, Phagwara, Punjab, India
| |
Collapse
|
11
|
Wang XL, Xu HW, Liu NN. Oral Microbiota: A New Insight into Cancer Progression, Diagnosis and Treatment. PHENOMICS (CHAM, SWITZERLAND) 2023; 3:535-547. [PMID: 37881320 PMCID: PMC10593652 DOI: 10.1007/s43657-023-00124-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 08/04/2023] [Accepted: 08/07/2023] [Indexed: 10/27/2023]
Abstract
The polymorphic microbiome has been defined as one of the "Hallmarks of Cancer". Extensive studies have now uncovered the role of oral microbiota in cancer development and progression. Bacteria, fungi, archaea, and viruses in the oral cavity interact dynamically with the oral microenvironment to maintain the oral micro-ecological homeostasis. This complex interaction is influenced by many factors, such as maternal transmission, personal factors and environmental factors. Dysbiosis of oral microbiota can disturbed this host-microbiota interaction, leading to systemic diseases. Numerous studies have shown the potential associations between oral microbiota and a variety of cancers. However, the underlying mechanisms and therapeutic insights are still poorly understood. In this review, we mainly focus on the following aspects: (1) the factors affect oral microbiota composition and function; (2) the interaction between microenvironment and oral microbiota; (3) the role of multi-kingdom oral microbiota in human health; (4) the potential underlying mechanisms and therapeutic benefits of oral microbiota against cancer. Finally, we aim to describe the impact of oral microbiota on cancer progression and provide novel therapeutic insights into cancer prevention and treatment by targeting oral microbiota.
Collapse
Affiliation(s)
- Xiu-Li Wang
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025 China
| | - Hua-Wen Xu
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025 China
| | - Ning-Ning Liu
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025 China
| |
Collapse
|
12
|
Du J, Huang S, Wu M, Chen S, Zhou W, Zhan L, Huang X. Dlt operon regulates physiological function and cariogenic virulence in Streptococcus mutans. Future Microbiol 2023; 18:225-233. [PMID: 37097048 DOI: 10.2217/fmb-2022-0165] [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] [Indexed: 04/26/2023] Open
Abstract
Streptococcus mutans is one of the major cariogenic pathogens in the oral cavity. The dlt operon is responsible for the process of D-alanylation of lipoteichoic acid and is related to the virulence of S. mutans. The dlt operon contributes to the adhesion, biofilm formation, stress response, interspecies competitiveness and autolysis of S. mutans. In addition, we have summarized the possible regulatory networks of the dlt operon. This review highlights the significant role of the dlt operon in S. mutans and provides new ideas for ecological caries prevention.
Collapse
Affiliation(s)
- Jingyun Du
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key lab of Fujian College & University, School & Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Shan Huang
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key lab of Fujian College & University, School & Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Minjing Wu
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key lab of Fujian College & University, School & Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Shuai Chen
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key lab of Fujian College & University, School & Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Wen Zhou
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key lab of Fujian College & University, School & Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Ling Zhan
- Division of Pediatric Dentistry, Department of Orofacial Sciences, Department of Preventive & Restorative Dental Sciences, University of California, San Francisco, CA, USA
| | - Xiaojing Huang
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key lab of Fujian College & University, School & Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| |
Collapse
|
13
|
Pezzotti G, Ofuji S, Imamura H, Adachi T, Yamamoto T, Kanamura N, Ohgitani E, Marin E, Zhu W, Mazda O, Togo A, Kimura S, Iwata T, Shiba H, Ouhara K, Aoki T, Kawai T. In Situ Raman Analysis of Biofilm Exopolysaccharides Formed in Streptococcus mutans and Streptococcus sanguinis Commensal Cultures. Int J Mol Sci 2023; 24:ijms24076694. [PMID: 37047667 PMCID: PMC10095091 DOI: 10.3390/ijms24076694] [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: 02/17/2023] [Revised: 03/23/2023] [Accepted: 03/27/2023] [Indexed: 04/14/2023] Open
Abstract
This study probed in vitro the mechanisms of competition/coexistence between Streptococcus sanguinis (known for being correlated with health in the oral cavity) and Streptococcus mutans (responsible for aciduric oral environment and formation of caries) by means of quantitative Raman spectroscopy and imaging. In situ Raman assessments of live bacterial culture/coculture focusing on biofilm exopolysaccharides supported the hypothesis that both species engaged in antagonistic interactions. Experiments of simultaneous colonization always resulted in coexistence, but they also revealed fundamental alterations of the biofilm with respect to their water-insoluble glucan structure. Raman spectra (collected at fixed time but different bacterial ratios) showed clear changes in chemical bonds in glucans, which pointed to an action by Streptococcus sanguinis to discontinue the impermeability of the biofilm constructed by Streptococcus mutans. The concurrent effects of glycosidic bond cleavage in water-insoluble α - 1,3-glucan and oxidation at various sites in glucans' molecular chains supported the hypothesis that secretion of oxygen radicals was the main "chemical weapon" used by Streptococcus sanguinis in coculture.
Collapse
Affiliation(s)
- Giuseppe Pezzotti
- Ceramic Physics Laboratory, Kyoto Institute of Technology, Sakyo-ku, Matsugasaki, Kyoto 606-8585, Japan
- Department of Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, 465 Kajii-cho, Kyoto 602-8566, Japan
- Department of Orthopedic Surgery, Tokyo Medical University, 6-7-1 Nishi-Shinjuku, Shinjuku-ku, Tokyo 160-0023, Japan
- Department of Dental Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
- Department of Applied Science and Technology, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Torino, Italy
- Department of Molecular Science and Nanosystems, Ca' Foscari University of Venice, Via Torino 155, 30172 Venice, Italy
| | - Satomi Ofuji
- Ceramic Physics Laboratory, Kyoto Institute of Technology, Sakyo-ku, Matsugasaki, Kyoto 606-8585, Japan
| | - Hayata Imamura
- Ceramic Physics Laboratory, Kyoto Institute of Technology, Sakyo-ku, Matsugasaki, Kyoto 606-8585, Japan
- Department of Dental Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Tetsuya Adachi
- Department of Dental Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Toshiro Yamamoto
- Department of Dental Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Narisato Kanamura
- Department of Dental Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Eriko Ohgitani
- Department of Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, 465 Kajii-cho, Kyoto 602-8566, Japan
| | - Elia Marin
- Ceramic Physics Laboratory, Kyoto Institute of Technology, Sakyo-ku, Matsugasaki, Kyoto 606-8585, Japan
- Department of Dental Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Wenliang Zhu
- Ceramic Physics Laboratory, Kyoto Institute of Technology, Sakyo-ku, Matsugasaki, Kyoto 606-8585, Japan
| | - Osam Mazda
- Department of Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, 465 Kajii-cho, Kyoto 602-8566, Japan
| | - Azusa Togo
- Department of Biomaterial Sciences, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Satoshi Kimura
- Department of Biomaterial Sciences, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Tadahisa Iwata
- Department of Biomaterial Sciences, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Hideki Shiba
- Department of Biological Endodontics, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan
| | - Kazuhisa Ouhara
- Department of Periodontal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan
| | - Takashi Aoki
- Faculty of Fiber Science and Engineering, Kyoto Institute of Technology, Sakyo-ku, Matsugasaki, Kyoto 606-8585, Japan
| | - Toshihisa Kawai
- Department of Oral Science and Translational Research, College of Dental Medicine, Nova Southeastern University, 3301 College Ave, Fort Lauderdale, FL 33314, USA
| |
Collapse
|
14
|
Yan Y, Hailun H, Fenghui Y, Pingting L, Lei L, Zhili Z, Tao H. Streptococcus mutans dexA affects exopolysaccharides production and biofilm homeostasis. Mol Oral Microbiol 2023; 38:134-144. [PMID: 36270969 DOI: 10.1111/omi.12395] [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: 04/12/2022] [Revised: 09/25/2022] [Accepted: 10/12/2022] [Indexed: 11/28/2022]
Abstract
OBJECTIVES The study aimed to evaluate the role of Streptococcus mutans (S. mutans) dexA gene on biofilm structure and microecological distribution in multispecies biofilms. MATERIALS AND METHODS A multispecies biofilm model consisting of S. mutans and its dexA mutants, Streptococcus gordonii (S. gordonii) and Streptococcus sanguinis (S. sanguinis) was constructed, and bacterial growth, biofilm architecture and microbiota composition were determined to study the effect of the S. mutans dexA on multispecies biofilms. RESULTS Our results showed that either deletion or overexpression of S. mutans dexA had no effect on the planktonic growth of bacterium, while S. mutans dominated in the multispecies biofilms to form cariogenic biofilms. Furthermore, we revealed that the SmudexA+ group showed structural abnormality in the form of more fractures and blank areas. The morphology of the SmudexA group was sparser and more porous, with reduced and less agglomerated exopolysaccharides scaffold. Interestingly, the microbiota composition analysis provided new insights that the inhibition of S. gordonii and S. sanguinis was alleviated in the SmudexA group compared to the significantly suppressed condition in the other groups. CONCLUSION In conclusion, deletion of S. mutans dexA gene re-modules biofilm structure and microbiota composition, thereby leading to decreased cariogenicity. Thus, the S. mutans dexA may be an important target for regulating the cariogenicity of dental plaque biofilms, expecting to be a probiotic for caries control.
Collapse
Affiliation(s)
- Yang Yan
- Hunan Key Laboratory of Oral Health Research & Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, China
| | - He Hailun
- School of Life Sciences, Central South University, Changsha, China
| | - Yang Fenghui
- School of Life Sciences, Central South University, Changsha, China
| | - Liu Pingting
- Hunan Key Laboratory of Oral Health Research & Xiangya Stomatological Hospital & Xiangya School of Stomatology, Central South University, Changsha, China
| | - Lei Lei
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Preventive Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Zhao Zhili
- Department of Oral and Maxillofacial Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Hu Tao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Preventive Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
15
|
Zaidi S, Ali K, Chawla YM, Khan AU. mltG gene deletion mitigated virulence potential of Streptococcus mutans: An in-vitro, ex-situ and in-vivo study. AMB Express 2023; 13:19. [PMID: 36806997 PMCID: PMC9941400 DOI: 10.1186/s13568-023-01526-x] [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: 02/03/2023] [Accepted: 02/07/2023] [Indexed: 02/22/2023] Open
Abstract
Bacterial cells are surrounded by a peptidoglycan (PG) cell wall, which is essential for cell integrity and intrinsic biogenesis pathways; hence, the cell wall is a potential target for several antibiotics. Among several lytic transglycosylases (LTs), the mltG gene plays a crucial role in the synthesis of peripheral PG. It localises the re-modelled PGs for septum formation and cleavage across the bacterial cell wall during daughter cells separation. However, the role of mltG gene in bacterial virulence, particularly in Gram-positive bacteria during dentine biofilm and caries development, has remained unexplored. Hence, we exploited Gram-positive Streptococcus mutans cells for the very first time to construct a mltG knock-out bacterial strain, e.g., ΔmltG S. mutans. Systematic comparative investigations revealed that doubling time (Td), survival, enzymatic efficiencies, pH tolerance, bio-synthesise of lipid, proteins and DNA, biofilm formation and dentine lesions were significantly (p < 0.001) compromised in case of ΔmltG S. mutans than wild type strain. The qRT-PCR based gene expression profiling revealed that transcriptional expression of critically important genes involved in biofilm, metabolism, and stress response were dysregulated in the mutant. Besides, an incredible reduction in dentine caries development was found in the molar teeth of Wistar rats and also in human extracted teeth. Concisely, these trends obtained evidently advocated the fact that the deletion of mltG gene can be a potential target to impair the S. mutans virulence through severe growth retardation, thereby reducing the virulence potential of S. mutans.
Collapse
Affiliation(s)
- Sahar Zaidi
- grid.411340.30000 0004 1937 0765Medical Microbiology and Molecular Biology Laboratory, Interdisciplinary, Biotechnology Unit, Aligarh Muslim University, Aligarh, 202002 UP India
| | - Khursheed Ali
- grid.411340.30000 0004 1937 0765Medical Microbiology and Molecular Biology Laboratory, Interdisciplinary, Biotechnology Unit, Aligarh Muslim University, Aligarh, 202002 UP India
| | - Yadya M. Chawla
- grid.425195.e0000 0004 0498 7682ICGEB-Emory Vaccine Center, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Asad U. Khan
- grid.411340.30000 0004 1937 0765Medical Microbiology and Molecular Biology Laboratory, Interdisciplinary, Biotechnology Unit, Aligarh Muslim University, Aligarh, 202002 UP India
| |
Collapse
|
16
|
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
|
17
|
Zeng L, Walker AR, Burne RA, Taylor ZA. Glucose Phosphotransferase System Modulates Pyruvate Metabolism, Bacterial Fitness, and Microbial Ecology in Oral Streptococci. J Bacteriol 2023; 205:e0035222. [PMID: 36468868 PMCID: PMC9879115 DOI: 10.1128/jb.00352-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 11/08/2022] [Indexed: 12/12/2022] Open
Abstract
Spontaneous mutants with defects in the primary glucose phosphotransferase permease (manLMNO) of Streptococcus sanguinis SK36 showed enhanced fitness at low pH. Transcriptomics and metabolomics with a manL deletion mutant (SK36/manL) revealed redirection of pyruvate to production of acetate and formate, rather than lactate. These observations were consistent with measurements of decreased lactic acid accumulation and increased excretion of acetate, formate, pyruvate, and H2O2. Genes showing increased expression in SK36/manL included those encoding carbohydrate transporters, extracellular glycosidases, intracellular polysaccharide metabolism, and arginine deiminase and pathways for metabolism of acetoin, ethanolamine, ascorbate, and formate, along with genes required for membrane biosynthesis and adhesion. Streptococcus mutans UA159 persisted much better in biofilm cocultures with SK36/manL than with SK36, an effect that was further enhanced by culturing the biofilms anaerobically but dampened by adding arginine to the medium. We posited that the enhanced persistence of S. mutans with SK36/manL was in part due to excess excretion of pyruvate by the latter, as addition of pyruvate to S. mutans-S. sanguinis cocultures increased the proportions of UA159 in the biofilms. Reducing the buffer capacity or increasing the concentration of glucose benefited UA159 when cocultured with SK36, but not with SK36/manL, likely due to the altered metabolism and enhanced acid tolerance of the mutant. When manL was deleted in S. mutans or Streptococcus gordonii, the mutants presented altered fitness characteristics. Our study demonstrated that phosphotransferase system (PTS)-dependent modulation of central metabolism can profoundly affect streptococcal fitness and metabolic interactions, revealing another dimension in commensal-pathogen relationships influencing dental caries development. IMPORTANCE Dental caries is underpinned by a dysbiotic microbiome and increased acid production. As beneficial bacteria that can antagonize oral pathobionts, oral streptococci such as S. sanguinis and S. gordonii can ferment many carbohydrates, despite their relative sensitivity to low pH. We characterized the molecular basis for why mutants of glucose transporter ManLMNO of S. sanguinis showed enhanced production of hydrogen peroxide and ammonia and improved persistence under acidic conditions. A metabolic shift involving more than 300 genes required for carbohydrate transport, energy production, and envelope biogenesis was observed. Significantly, manL mutants engineered in three different oral streptococci displayed altered capacities for acid production and interspecies antagonism, highlighting the potential for targeting the glucose-PTS to modulate the pathogenicity of oral biofilms.
Collapse
Affiliation(s)
- Lin Zeng
- Department of Oral Biology, University of Florida, Gainesville, Florida, USA
| | - Alejandro R. Walker
- Department of Oral Biology, University of Florida, Gainesville, Florida, USA
| | - Robert A. Burne
- Department of Oral Biology, University of Florida, Gainesville, Florida, USA
| | - Zachary A. Taylor
- Department of Oral Biology, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
18
|
Sun Y, Chen H, Xu M, He L, Mao H, Yang S, Qiao X, Yang D. Exopolysaccharides metabolism and cariogenesis of Streptococcus mutans biofilm regulated by antisense vicK RNA. J Oral Microbiol 2023; 15:2204250. [PMID: 37138664 PMCID: PMC10150615 DOI: 10.1080/20002297.2023.2204250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2023] Open
Abstract
Background Streptococcus mutans (S. mutans) is a pivotal cariogenic pathogen contributing to its multiple virulence factors, one of which is synthesizing exopolysaccharides (EPS). VicK, a sensor histidine kinase, plays a major role in regulating genes associated with EPS synthesis and adhesion. Here we first identified an antisense vicK RNA (ASvicK) bound with vicK into double-stranded RNA (dsRNA). Objective This study aims to investigate the effect and mechanism of ASvicK in the EPS metabolism and cariogenesis of S. mutans. Methods The phenotypes of biofilm were detected by scanning electron microscopy (SEM), gas chromatography-mass spectrometery (GC-MS) , gel permeation chromatography (GPC) , transcriptome analysis and Western blot. Co-immunoprecipitation (Co-ip) assay and enzyme activity experiment were adopted to investigate the mechanism of ASvicK regulation. Caries animal models were developed to study the relationship between ASvicK and cariogenicity of S. mutans. Results Overexpression of ASvicK can inhibit the growth of biofilm, reduce the production of EPS and alter genes and protein related to EPS metabolism. ASvicK can adsorb RNase III to regulate vicK and affect the cariogenicity of S. mutans. Conclusions ASvicK regulates vicK at the transcriptional and post-transcriptional levels, effectively inhibits EPS synthesis and biofilm formation and reduces its cariogenicity in vivo.
Collapse
Affiliation(s)
- Yuting Sun
- Department of Endodontics, Stomatological Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Hong Chen
- Department of Endodontics, Stomatological Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Mengmeng Xu
- Department of Endodontics, Stomatological Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Liwen He
- Department of Endodontics, Stomatological Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Hongchen Mao
- Department of Endodontics, Stomatological Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Shiyao Yang
- Department of Endodontics, Stomatological Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Xin Qiao
- Department of Endodontics, Stomatological Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Deqin Yang
- Department of Endodontics, Stomatological Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
- CONTACT Deqin Yang Department of Endodontics, Stomatological Hospital of Chongqing Medical University, Chongqing4404100, China
| |
Collapse
|
19
|
Gu M, Cho JH, Suh JW, Cheng J. Potential oral probiotic Lactobacillus pentosus MJM60383 inhibits Streptococcus mutans biofilm formation by inhibiting sucrose decomposition. J Oral Microbiol 2022; 15:2161179. [PMID: 36605406 PMCID: PMC9809368 DOI: 10.1080/20002297.2022.2161179] [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: 06/03/2022] [Accepted: 12/16/2022] [Indexed: 12/29/2022] Open
Abstract
Streptococcus mutans is known as a contributor to dental caries. In this work, Lactobacillus pentosus MJM60383 was selected for its strong antagonistic activity against S. mutans and was characterized by good oral probiotic properties including lysozyme tolerance, adhesive ability to oral cells, good aggregation (auto-aggregation, co-aggregation) ability, hydrogen peroxide production and inhibition of biofilm formation of S. mutans. L. pentosus MJM60383 also exhibited safety as a probiotic characterized by no hemolytic activity, no D-lactate production, no biogenic amine production, and susceptibility to antibiotics. Furthermore, the biofilm formation of S. mutans was also significantly inhibited by the supernatant of L. pentosus MJM60383. An anti-biofilm mechanism study revealed that sucrose decomposition and the production of water-insoluble exopolysaccharides by S. mutans were inhibited by the treatment with L. pentosus MJM60383 supernatant. Real-time PCR analysis indicated that the supernatant of L. pentosus MJM60383 significantly inhibited the mRNA expression of S. mutans glycosyltransferases, which synthesize glucan to construct biofilm architecture and mediate bacterial adherence. Our study demonstrated L. pentosus MJM60383 as a potential oral probiotic and revealed its anti-biofilm mechanism.
Collapse
Affiliation(s)
- Mingkun Gu
- Interdisciplinary Program of Biomodulation, Myongji University, Yongin, Republic of Korea
| | - Joo-Hyung Cho
- Myongji Bioefficacy Research Center, Myongji University, Yongin, Republic of Korea
| | - Joo-Won Suh
- Myongji Bioefficacy Research Center, Myongji University, Yongin, Republic of Korea
| | - Jinhua Cheng
- Myongji Bioefficacy Research Center, Myongji University, Yongin, Republic of Korea
| |
Collapse
|
20
|
Negrini TDC, Ren Z, Miao Y, Kim D, Simon-Soro Á, Liu Y, Koo H, Arthur RA. Dietary sugars modulate bacterial-fungal interactions in saliva and inter-kingdom biofilm formation on apatitic surface. Front Cell Infect Microbiol 2022; 12:993640. [PMID: 36439211 PMCID: PMC9681999 DOI: 10.3389/fcimb.2022.993640] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 10/19/2022] [Indexed: 11/10/2022] Open
Abstract
Bacteria and fungi can interact to form inter-kingdom biofilms in the oral cavity. Streptococcus mutans and Candida albicans are frequently detected in saliva and in dental biofilms associated with early childhood caries (tooth-decay), a prevalent oral disease induced by dietary sugars. However, how different sugars influence this bacterial-fungal interaction remains unclear. Here, we investigate whether specific sugars affect the inter-kingdom interaction in saliva and subsequent biofilm formation on tooth-mimetic surfaces. The microbes were incubated in saliva containing common dietary sugars (glucose and fructose, sucrose, starch, and combinations) and analyzed via fluorescence imaging and quantitative computational analyses. The bacterial and fungal cells in saliva were then transferred to hydroxyapatite discs (tooth mimic) to allow microbial binding and biofilm development. We found diverse bacterial-fungal aggregates which varied in size, structure, and spatial organization depending on the type of sugars. Sucrose and starch+sucrose induced the formation of large mixed-species aggregates characterized by bacterial clusters co-bound with fungal cells, whereas mostly single-cells were found in the absence of sugar or in the presence of glucose and fructose. Notably, both colonization and further growth on the apatitic surface were dependent on sugar-mediated aggregation, leading to biofilms with distinctive spatial organizations and 3D architectures. Starch+sucrose and sucrose-mediated aggregates developed into large and highly acidogenic biofilms with complex network of bacterial and fungal cells (yeast and hyphae) surrounded by an intricate matrix of extracellular glucans. In contrast, biofilms originated from glucose and fructose-mediated consortia (or without sugar) were sparsely distributed on the surface without structural integration, growing predominantly as individual species with reduced acidogenicity. These findings reveal the impact of dietary sugars on inter-kingdom interactions in saliva and how they mediate biofilm formation with distinctive structural organization and varying acidogenicity implicated with human tooth-decay.
Collapse
Affiliation(s)
- Thais de Cássia Negrini
- Biofilm Research Laboratories, Center for Innovation & Precision Dentistry, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States,Department of Orthodontics and Divisions of Pediatric Dentistry & Community Oral Health, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States,Department of Clinical Analysis, School of Pharmaceutical Sciences, Sao Paulo State University, Araraquara, Brazil
| | - Zhi Ren
- Biofilm Research Laboratories, Center for Innovation & Precision Dentistry, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States,Department of Orthodontics and Divisions of Pediatric Dentistry & Community Oral Health, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Yilan Miao
- Biofilm Research Laboratories, Center for Innovation & Precision Dentistry, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States,Department of Orthodontics and Divisions of Pediatric Dentistry & Community Oral Health, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Dongyeop Kim
- Biofilm Research Laboratories, Center for Innovation & Precision Dentistry, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States,Department of Orthodontics and Divisions of Pediatric Dentistry & Community Oral Health, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States,Department of Preventive Dentistry, School of Dentistry, and Institute of Oral Bioscience, Jeonbuk National University, Jeonju, South Korea
| | - Áurea Simon-Soro
- Biofilm Research Laboratories, Center for Innovation & Precision Dentistry, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States,Department of Orthodontics and Divisions of Pediatric Dentistry & Community Oral Health, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States,Department of Stomatology, School of Dentistry, University of Seville, Seville, Spain
| | - Yuan Liu
- Biofilm Research Laboratories, Center for Innovation & Precision Dentistry, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States,Department of Orthodontics and Divisions of Pediatric Dentistry & Community Oral Health, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States,Preventive & Restorative Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Hyun Koo
- Biofilm Research Laboratories, Center for Innovation & Precision Dentistry, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States,Department of Orthodontics and Divisions of Pediatric Dentistry & Community Oral Health, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Rodrigo Alex Arthur
- Biofilm Research Laboratories, Center for Innovation & Precision Dentistry, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States,Department of Orthodontics and Divisions of Pediatric Dentistry & Community Oral Health, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States,Department of Preventive and Community Dentistry, Dental School, Federal University of Rio Grande do Sul, Porto Alegre, Brazil,*Correspondence: Rodrigo Alex Arthur,
| |
Collapse
|
21
|
Abstract
Lysine acetylation, a ubiquitous and dynamic regulatory posttranslational modification (PTM), affects hundreds of proteins across all domains of life. In bacteria, lysine acetylation can be found in many essential pathways, and it is also crucial for bacterial virulence. However, the biological significance of lysine acetylation events to bacterial virulence factors remains poorly characterized. In Streptococcus mutans, the acetylome profiles help identify several lysine acetylation sites of lactate dehydrogenase (LDH), which catalyzes the conversion of pyruvate to lactic acid, causing the deterioration of teeth. We investigated the regulatory mechanism of LDH acetylation and characterized the effect of LDH acetylation on its function. We overexpressed the 15 Gcn5 N-acetyltransferases (GNAT) family members in S. mutans and showed that the acetyltransferase ActA impaired its acidogenicity by acetylating LDH. Additionally, enzymatic acetyltransferase reactions demonstrated that purified ActA could acetylate LDH in vitro, and 10 potential lysine acetylation sites of LDH were identified by mass spectrometry, 70% of which were also detected in vivo. We further demonstrated that the lysine acetylation of LDH inhibited its enzymatic activity, and a subsequent rat caries model showed that ActA impaired the cariogenicity of S. mutans. Collectively, we demonstrated that ActA, the first identified and characterized acetyltransferase in S. mutans, acetylated the LDH enzymatically and inhibited its enzymatic activity, thereby providing a starting point for the further analysis of the biological significance of lysine acetylation in the virulence of S. mutans.
Collapse
|
22
|
Boddapati S, Gummadi SN. A comprehensive review on mutan (a mixed linkage of α-1-3 and α-1-6 glucans) from bacterial sources. Biotechnol Genet Eng Rev 2021; 37:208-237. [PMID: 34816783 DOI: 10.1080/02648725.2021.2003072] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Mutan is an extracellular sticky polymer having α-1-3 and α-1-6 glycosidic linkages with a large diversity in molecular weights and structures depending on the source. These compounds are reported to be highly thermostable and also have potential physiochemical and biological applications. The main aim of this review is to provide an overview of glucosyltransferases and their role in mutan synthesis. The production strategies and structural properties of bacterial mutans are discussed with a goal to improve production efficiency. The physicochemical features, chemical modifications, potential industrial applications and future prospects are also discussed. According to data, mutan and its derivatives will play a larger role in medicinal sectors and as thermoplastics in the near future.Abbreviations: ABTS: 2,2'-azino-bis-3-ethylbenzthiazoline-6-sulphonic acid; BHI: Brain heart infusion broth; 13C (HSQC) NMR: Heteronuclear Single Quantum Coherence NMR; CBMs: Carbohydrate binding modules; DPPH: 2,2-diphenyl-1-picrylhydrazyl; FTIR: Fourier-transform infrared spectroscopy; GC-MS: Gas chromatography-mass spectrometry; GPC: Gel permeation chromatography; Gtfs: Glucosyltransferases; 1H (DQF-COSY): Double-quantum filtered correlation spectroscopy; HPAEC-PAD: High-performance anion exchange chromatography with pulsed amperometric detection; HPLC: High performance liquid chromatography; HPSEC-RI: High-performance size exclusive chromatography coupled with refractive index; HPSEC-MALLS: High-performance size exclusive chromatography with multi-angle laser light scattering detection; MALDI-TOF: Matrix-Assisted Laser Desorption/Ionization-Time of Flight mass spectrometry; Mw: Weight-average molecular weight; MWD: Molecular weight distribution; NMR: Nuclear magnetic resonance spectroscopy; TEM: Transmission electron microscopy; THB: Todd Hewitt Broth; TTY: Tryticase tryptose yeast extract broth.
Collapse
Affiliation(s)
- Sirisha Boddapati
- Applied and Industrial Microbiology Laboratory, Department of Biotechnology, Bjm School of Biosciences, Indian Institute of Technology-Madras, Chennai, India
| | - Sathyanaryana N Gummadi
- Applied and Industrial Microbiology Laboratory, Department of Biotechnology, Bjm School of Biosciences, Indian Institute of Technology-Madras, Chennai, India
| |
Collapse
|
23
|
Zhou Y, Deng W, Mo M, Luo D, Liu H, Jiang Y, Chen W, Xu C. Stimuli-Responsive Nanoplatform-Assisted Photodynamic Therapy Against Bacterial Infections. Front Med (Lausanne) 2021; 8:729300. [PMID: 34604266 PMCID: PMC8482315 DOI: 10.3389/fmed.2021.729300] [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: 06/22/2021] [Accepted: 08/13/2021] [Indexed: 11/17/2022] Open
Abstract
Bacterial infections are common diseases causing tremendous deaths in clinical settings. It has been a big challenge to human beings because of the antibiotics abuse and the newly emerging microbes. Photodynamic therapy (PDT) is a reactive oxygen species-based therapeutic technique through light-activated photosensitizer (PS). Recent studies have highlighted the potential of PDT as an alternative method of antibacterial treatment for its broad applicability and high efficiency. However, there are some shortcomings due to the low selectivity and specificity of PS. Growing evidence has shown that drug delivery nanoplatforms have unique advantages in enhancing therapeutic efficacy of drugs. Particularly, stimuli-responsive nanoplatforms, as a promising delivery system, provide great opportunities for the effective delivery of PS. In the present mini-review, we briefly introduced the unique microenvironment in bacterial infection tissues and the application of PDT on bacterial infections. Then we review the stimuli-responsive nanoplatforms (including pH-, enzymes-, redox-, magnetic-, and electric-) used in PDT against bacterial infections. Lastly, some perspectives have also been proposed to further promote the future developments of antibacterial PDT.
Collapse
Affiliation(s)
- You Zhou
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & National Medical Products Administration Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Wenmin Deng
- Department of Clinical Pharmacy, The People's Hospital of Dianbai District, Maoming, China
| | - Mulan Mo
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & National Medical Products Administration Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Dexu Luo
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & National Medical Products Administration Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Houhe Liu
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & National Medical Products Administration Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yuan Jiang
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & National Medical Products Administration Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.,Department of Rehabilitation Medicine, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Wenjie Chen
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & National Medical Products Administration Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.,State Key Laboratory of Respiratory Disease, Guangdong-Hongkong-Macao Joint Laboratory of Respiratory Infectious Disease, Guangzhou, China.,Sydney Vital Translational Cancer Research Centre, Sydney, NSW, Australia
| | - Chuanshan Xu
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State & National Medical Products Administration Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
24
|
Deacidification of Cranberry Juice Reduces Its Antibacterial Properties against Oral Streptococci but Preserves Barrier Function and Attenuates the Inflammatory Response of Oral Epithelial Cells. Foods 2021; 10:foods10071634. [PMID: 34359504 PMCID: PMC8305880 DOI: 10.3390/foods10071634] [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: 05/18/2021] [Revised: 06/28/2021] [Accepted: 07/10/2021] [Indexed: 02/07/2023] Open
Abstract
Cranberry (Vaccinium macrocarpon) may be a potent natural adjuvant for the prevention of oral diseases due to its anti-adherence, anti-cariogenic, and anti-inflammatory properties. However, the high titrable acidity of cranberry juice (CJ) has been reported to cause gastrointestinal discomfort, leading consumers to restrict their intake of this beverage. Electrodialysis with a bipolar membrane (EDBM) can reduce the organic acid content of CJ while retaining the flavonoids associated with potential health benefits. This study aimed to assess how the deacidification of CJ by EDBM impacts the antibacterial properties of the beverage against cariogenic (Streptococcus mutans, Streptococcus sobrinus) and commensal (Streptococcus gordonii, Streptococcus oralis, Streptococcus salivarius) streptococci, and how it affects oral epithelial barrier function and inflammatory response in an in vitro model. The removal of organic acids from CJ (deacidification rate ≥42%) reduced the bactericidal activity of the beverage against planktonic S. mutans and S. gordonii after a 15-min exposure, whereas only the viability of S. gordonii was significantly impacted by CJ deacidification rate when the bacteria were embedded in a biofilm. Moreover, conditioning saliva-coated hydroxyapatite with undiluted CJ samples significantly lowered the adherence of S. mutans, S. sobrinus, and S. oralis. With respect to epithelial barrier function, exposure to CJ deacidified at a rate of ≥19% maintained the integrity of a keratinocyte monolayer over the course of 24 h compared to raw CJ, as assessed by the determination of transepithelial electrical resistance (TER) and fluorescein isothiocyanate-conjugated dextran paracellular transport. These results can be in part attributed to the inability of the deacidified CJ to disrupt two tight junction proteins, zonula occludens-1 and occludin, following exposure, unlike raw CJ. Deacidification of CJ impacted the secretion of IL-6, but not of IL-8, by oral epithelial cells. In conclusion, deacidification of CJ appears to provide benefits with respect to the maintenance of oral health.
Collapse
|
25
|
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
|
26
|
Flemming J, Meyer-Probst CT, Speer K, Kölling-Speer I, Hannig C, Hannig M. Preventive Applications of Polyphenols in Dentistry-A Review. Int J Mol Sci 2021; 22:4892. [PMID: 34063086 PMCID: PMC8124254 DOI: 10.3390/ijms22094892] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/27/2021] [Accepted: 04/27/2021] [Indexed: 01/18/2023] Open
Abstract
Polyphenols are natural substances that have been shown to provide various health benefits. Antioxidant, anti-inflammatory, and anti-carcinogenic effects have been described. At the same time, they inhibit the actions of bacteria, viruses, and fungi. Thus, studies have also examined their effects within the oral cavity. This review provides an overview on the different polyphenols, and their structure and interactions with the tooth surface and the pellicle. In particular, the effects of various tea polyphenols on bioadhesion and erosion have been reviewed. The current research confirms that polyphenols can reduce the growth of cariogenic bacteria. Furthermore, they can decrease the adherence of bacteria to the tooth surface and improve the erosion-protective properties of the acquired enamel pellicle. Tea polyphenols, especially, have the potential to contribute to an oral health-related diet. However, in vitro studies have mainly been conducted. In situ studies and clinical studies need to be extended and supplemented in order to significantly contribute to additive prevention measures in caries prophylaxis.
Collapse
Affiliation(s)
- Jasmin Flemming
- Clinic of Operative Dentistry, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Fetscherstraße 74, D-01307 Dresden, Germany; (J.F.); (C.H.)
| | - Clara Theres Meyer-Probst
- Clinic of Operative Dentistry, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Fetscherstraße 74, D-01307 Dresden, Germany; (J.F.); (C.H.)
| | - Karl Speer
- Special Food Chemistry and Food Production, TU Dresden, Bergstraße 66, D-01069 Dresden, Germany; (K.S.); (I.K.-S.)
| | - Isabelle Kölling-Speer
- Special Food Chemistry and Food Production, TU Dresden, Bergstraße 66, D-01069 Dresden, Germany; (K.S.); (I.K.-S.)
| | - Christian Hannig
- Clinic of Operative Dentistry, Medical Faculty Carl Gustav Carus, Technische Universität Dresden, Fetscherstraße 74, D-01307 Dresden, Germany; (J.F.); (C.H.)
| | - Matthias Hannig
- Clinic of Operative Dentistry, Periodontology and Preventive Dentistry, University Hospital, Saarland University, Building 73, D-66421 Homburg, Germany;
| |
Collapse
|
27
|
Yuan K, Hou L, Jin Q, Niu C, Mao M, Wang R, Huang Z. Comparative transcriptomics analysis of Streptococcus mutans with disruption of LuxS/AI-2 quorum sensing and recovery of methyl cycle. Arch Oral Biol 2021; 127:105137. [PMID: 33965851 DOI: 10.1016/j.archoralbio.2021.105137] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 04/23/2021] [Accepted: 04/24/2021] [Indexed: 11/16/2022]
Abstract
OBJECTIVE The LuxS/AI-2 quorum sensing (QS) system has critical roles in Streptococcus mutans cariogenicity. Whereas the molecular and cellular mechanisms of the LuxS/AI-2 QS system are not thoroughly understood. Given that LuxS has roles in QS and methyl cycle, its mutation can cause QS deficiency and methyl cycle disruption. The aim of this study was to investigate effects of the LuxS/AI-2 QS system on gene expression in Streptococcus mutans when methyl cycle was recovered with exogenous sahH gene. METHODS Our previous study introduced the exogenous sahH gene from Pseudomonas aeruginosa into an S. mutans luxS-null strain to restore the disrupted methyl cycle, and this produced the solely the LuxS/AI-2 QS system deficient strain. Here, we analyzed the transcriptomics of this strain to get insights into the molecular mechanisms of the LuxS/AI-2 QS system applying RNA-seq. RESULTS With recovery of methyl cycle, 84 genes didn't change in expression trends in S. mutans luxS-null strain. These genes mainly encode the ABC transporters, sugar transporter EII and enzymes of carbohydrate metabolism, and are rich in the Phosphotransferase system, Fructose and mannose metabolism, Amino sugar and nucleotide sugar metabolism, Galactose metabolism, Glycolysis/Gluconeogenesis, RNA degradation, Lysine biosynthesis, and Glycine, serine and threonine metabolism. CONCLUSIONS The LuxS/AI-2 QS system may mainly affect ABC transporters and carbohydrate transport, transformation and metabolism via EII subunits and enzymes to influence virulence-associated traits without effects of methyl cycle inStreptococcus mutans.
Collapse
Affiliation(s)
- Keyong Yuan
- Department of Endodontics, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai JiaoTong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Lili Hou
- Department of Nursing, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Qiaoqiao Jin
- Department of Endodontics, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai JiaoTong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Chenguang Niu
- Department of Endodontics, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai JiaoTong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Mengying Mao
- Department of Endodontics, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai JiaoTong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China
| | - Ruirui Wang
- Department of Stomatology, Minhang Branch, Zhongshan Hospital, Fudan University, China.
| | - Zhengwei Huang
- Department of Endodontics, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai JiaoTong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, China.
| |
Collapse
|
28
|
André CB, Rosalen PL, Giannini M, Bueno-Silva B, Pfeifer CS, Ferracane JL. Incorporation of Apigenin and tt-Farnesol into dental composites to modulate the Streptococcus mutans virulence. Dent Mater 2021; 37:e201-e212. [PMID: 33422299 PMCID: PMC7981265 DOI: 10.1016/j.dental.2020.12.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 12/05/2020] [Accepted: 12/22/2020] [Indexed: 11/30/2022]
Abstract
OBJECTIVES The aim of this in vitro study was to incorporate two anti-caries agents, Apigenin and tt-Farnesol, to resin composite and resin cement to reduce the virulence of Streptococcus mutans around dental restorations. METHODS Apigenin (Api, 5 mM) and tt-Farnesol (Far, 5 mM) were added alone, together, and combined with fluoride (F). Biofilm of S. mutans was grown on composite discs, and the dry-weight, bacterial viability, and the polysaccharides (alkali-soluble, intracellular and water-soluble) were quantified. CLSM images of the S. mutans biofilm were obtained after three years of water-storage. The effect of the additions on the physicochemical properties and the composite colorimetric parameters were also analyzed. RESULTS The additions did not affect bacterial viability. Api alone and combined with Far or combined with Far and F decreased the bacterial dry-weight, alkali-soluble and intracellular polysaccharides. After three years, the composites containing the additions presented a greater EPS matrix on the top of biofilm. Statistical difference was obtained for the degree of conversion; however, the maximum polymerization rate and curing kinetics were unaffected by the additions. No difference was observed for the water-soluble polysaccharides, flexural strength, and elastic modulus. Api increased the yellowness of the composites. SIGNIFICANCE Api, alone and combined, reduced the expression of virulence of S. mutans without jeopardizing the physicochemical properties of the composites.
Collapse
Affiliation(s)
- Carolina Bosso André
- Department of Restorative Dentistry, School of Dentistry, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil.
| | - Pedro Luiz Rosalen
- Department of Biosciences, Piracicaba Dental School, University of Campinas, Piracicaba, SP, Brazil; Biological Sciences Graduate Program, Federal University of Alfenas, Alfenas, MG, Brazil.
| | - Marcelo Giannini
- Department of Restorative Dentistry, Piracicaba Dental School, University of Campinas, Piracicaba, SP, Brazil.
| | - Bruno Bueno-Silva
- Dental Research Division, School of Dentistry, Guarulhos University, Guarulhos, SP, Brazil.
| | - Carmem Silvia Pfeifer
- Department of Biomaterials and Biomechanics, School of Dentistry, Oregon Health & Science University, Portland, OR, USA.
| | - Jack Liborio Ferracane
- Department of Biomaterials and Biomechanics, School of Dentistry, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
29
|
Miran W, Naradasu D, Okamoto A. Pathogens electrogenicity as a tool for in-situ metabolic activity monitoring and drug assessment in biofilms. iScience 2021; 24:102068. [PMID: 33554070 PMCID: PMC7859304 DOI: 10.1016/j.isci.2021.102068] [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] [Indexed: 11/26/2022] Open
Abstract
Concerns regarding increased antibiotic resistance arising from the emergent properties of biofilms have spurred interest in the discovery of novel antibiotic agents and techniques to directly estimate metabolic activity in biofilms. Although a number of methods have been developed to quantify biofilm formation, real-time quantitative assessment of metabolic activity in label-free biofilms remains a challenge. Production of electrical current via extracellular electron transport (EET) has recently been found in pathogens and appears to correlate with their metabolic activity. Accordingly, monitoring the production of electrical currents as an indicator of cellular metabolic activity in biofilms represents a new direction for research aiming to assess and screen the effects of antimicrobials on biofilm activity. In this article, we reviewed EET-capable pathogens and the methods to monitor biofilm activity to discuss advantages of using the capability of pathogens to produce electrical currents and effective combination of these methods. Moreover, we discussed EET mechanisms by pathogenic and environmental bacteria and open questions for the physiological roles of EET in pathogen's biofilm. The present limitations and possible future directions of in situ biofilm metabolic activity assessment for large-scale screening of antimicrobials are also discussed.
Collapse
Affiliation(s)
- Waheed Miran
- International Center for Materials Nanoarchitectonics, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan
| | - Divya Naradasu
- International Center for Materials Nanoarchitectonics, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan
| | - Akihiro Okamoto
- International Center for Materials Nanoarchitectonics, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan
- Graduate School of Chemical Sciences and Engineering, Hokkaido University, North 13 West 8, Kita-ku, Sapporo, Hokkaido 060-8628, Japan
| |
Collapse
|
30
|
Inhibitory Potential of Mangiferin on Glucansucrase Producing Streptococcus mutans Biofilm in Dental Plaque. APPLIED SCIENCES-BASEL 2020. [DOI: 10.3390/app10228297] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Glucansucrase secreted by Streptococcus mutans and composed of virulence genes alters oral microbiota, creating adherent environment for structural bacteria colony forming dental biofilm. The present investigation studied the inhibitory and binding potentials of mangiferin against S. mutans and its enzyme glucansucrase implicated in biofilm formation. Antibacterial activity against planktonic S. mutans was carried out. Using reverse transcription PCR, the expression of crucial virulence genes, gtfB, gtfC, gtfD, gbpB, and comDE were determined. The effect of mangiferin on teeth surfaces biofilm was ascertained by scanning electron microscopy (SEM). Docking analysis of S. mutans glucansucrase and mangiferin revealed the binding energy of −7.35 and ten hydrogen interactions. Antibacterial study revealed that mangiferin was not lethal to planktonic S. mutans, but a concentration-dependent inhibition of glucansucrase activity was observed. The inhibitory effect of water-insoluble glucan synthesis was apparently more marked relative to water-soluble glucan synthesis attenuation. Mangiferin significantly downregulated the expression of the virulence genes, indicating a mechanism involving glucanotranferases, specifically inhibiting colony formation by attenuating bacterial adherence. SEM images revealed that S. mutans biofilm density was scanty in mangiferin treated teeth compared to non-treated control teeth. Our data therefore suggest that mangiferin inhibited S. mutans biofilms formation by attenuating glucansucrase activities without affecting bacteria growth.
Collapse
|
31
|
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
|
32
|
Boddapati S, Rai R, Gummadi SN. Structural analysis and antioxidative properties of mutan (water-insoluble glucan) and carboxymethyl mutan from Streptococcus mutans. Process Biochem 2020. [DOI: 10.1016/j.procbio.2020.07.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
33
|
Sims KR, Maceren JP, Liu Y, Rocha GR, Koo H, Benoit DSW. Dual antibacterial drug-loaded nanoparticles synergistically improve treatment of Streptococcus mutans biofilms. Acta Biomater 2020; 115:418-431. [PMID: 32853808 PMCID: PMC7530141 DOI: 10.1016/j.actbio.2020.08.032] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 08/12/2020] [Accepted: 08/13/2020] [Indexed: 12/12/2022]
Abstract
Dental caries (i.e., tooth decay), which is caused by biofilm formation on tooth surfaces, is the most prevalent oral disease worldwide. Unfortunately, many anti-biofilm drugs lack efficacy within the oral cavity due to poor solubility, retention, and penetration into biofilms. While drug delivery systems (DDS) have been developed to overcome these hurdles and improve traditional antimicrobial treatments, including farnesol, efficacy is still modest due to myriad resistance mechanisms employed by biofilms, suggesting that synergistic drug treatments may be more efficacious. Streptococcus mutans (S. mutans), a cariogenic pathogen and biofilm forming model organism, has several key virulence factors including acidogenicity and exopolysaccharide (EPS) matrix synthesis. Flavonoids, such as myricetin, can reduce both biofilm acidogenicity and EPS synthesis. Therefore, a nanoparticle carrier (NPC) DDS with flexibility to co-load farnesol in the hydrophobic core and myricetin within the cationic corona, was tested in vitro using established and developing S. mutans biofilms. Co-loaded NPC treatments effectively disrupted biofilm biomass (i.e., dry weight) and reduced biofilm viability by ~3 log CFU/mL versus single drug-only controls in developing biofilms, suggesting dual-drug delivery exhibits synergistic anti-biofilm effects. Mechanistic studies revealed that co-loaded NPCs synergistically inhibited planktonic bacterial growth compared to controls and reduced S. mutans acidogenicity due to decreased atpD expression, a gene associated with acid tolerance. Moreover, the myricetin-loaded NPC corona enhanced NPC binding to tooth-mimetic surfaces, which can increase drug efficacy through improved retention at the biofilm-apatite interface. Altogether, these findings suggest promise for co-delivery of myricetin and farnesol DDS as an alternative anti-biofilm treatment to prevent dental caries.
Collapse
Affiliation(s)
- Kenneth R Sims
- University of Rochester School of Medicine and Dentistry, Translational Biomedical Science, Rochester, NY, United States; University of Rochester, Department of Biomedical Engineering, Rochester, NY, United States
| | - Julian P Maceren
- University of Rochester, Department of Chemistry, Rochester, NY, United States
| | - Yuan Liu
- University of Pennsylvania, Center for Innovation and Precision Dentistry, School of Dental Medicine, Department of Orthodontics, Philadelphia, PA, United States
| | - Guilherme R Rocha
- University of Rochester, Department of Biomedical Engineering, Rochester, NY, United States; São Paulo State University, Department of Dental Materials and Prosthodontics, Araraquara, São Paulo, Brazil
| | - Hyun Koo
- University of Pennsylvania, Center for Innovation and Precision Dentistry, School of Dental Medicine, Department of Orthodontics, Philadelphia, PA, United States
| | - Danielle S W Benoit
- University of Rochester, Department of Biomedical Engineering, Rochester, NY, United States; University of Rochester, Materials Science Program, NY, United States; University of Rochester, Department of Orthopaedics and Center for Musculoskeletal Research, NY, United States; University of Rochester, Center for Oral Biology, NY, United States; University of Rochester, Department of Chemical Engineering, NY, United States.
| |
Collapse
|
34
|
Li B, Pan T, Lin H, Zhou Y. The enhancing antibiofilm activity of curcumin on Streptococcus mutans strains from severe early childhood caries. BMC Microbiol 2020; 20:286. [PMID: 32938379 PMCID: PMC7493841 DOI: 10.1186/s12866-020-01975-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 09/10/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Streptococcus mutans (S. mutans) is one of the main cariogenic bacteria for caries. It was found that the clinical strains of S. mutans isolated from caries active population have stronger cariogenic ability than the isolates from caries-free (CF) people. Previous studies have found that curcumin can inhibit biofilm formation of S. mutans UA159. The objective of this study is to explore the antibiofilm effect of curcumin on the clinical isolates of S. mutans from severe early childhood caries(SECC). RESULTS The isolates from SECC group had more biomass than CF group (t = 4.296, P < 0.001). The acidogenicity and aciduricity of the strains from two groups showed no significant difference. After treatment with curcumin, the viability of biofilm was reduced to 61.865% ± 7.108% in SECC and to 84.059% ± 10.227% in CF group at 24 h (P < 0.05). The net reduction of live bacteria and total bacteria in the SECC group was significantly higher than that of the CF group (live bacteria t = 3.305, P = 0.016; total bacteria t = 2.378, P = 0.045) at 5 min. For 24 h, the net reduction of live bacteria and total bacteria in the SECC group was significantly higher than that of the CF group (live bacteria t = 3.305, P = 0.016; total bacteria t = 2.378, P = 0.045). The reduction of biofilm thickness reduced significantly in 5 min (t = 4.110, P = 0.015) and in 24 h (t = 3.453, P = 0.014). Long-term (24 h) curcumin treatment inhibited the amount of EPS in SECC group from (25.980 ± 1.156) μm3/μm2 to (20.136 ± 1.042) μm3/μm2, the difference was statistically significant (t = 7.510, P < 0.001). The gene of gtfC, gtfD, ftf, gbpB, fruA and srtA in the CF group and the gtfB, gtfC, gtfD, ftf, gbpB, srtA in SECC group were respectively reduced after 5 min curcumin treatment. After 24 h treatment, the gtfB, gtfC, gtfD, ftf, gbpB, fruA and srtA in both two groups were downregulation, all the differences were statistically significant. CONCLUSIONS Curcumin has antibiofilm activity on clinical strains of S. mutans, especially for those isolated from SECC.
Collapse
Affiliation(s)
- Bingchun Li
- Department of Preventive Dentistry, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, 56 Ling Yuan Road West, Guangzhou, 510055, China.,Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-Sen University, Guangzhou, China
| | - Ting Pan
- Department of Preventive Dentistry, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, 56 Ling Yuan Road West, Guangzhou, 510055, China.,Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-Sen University, Guangzhou, China
| | - Huancai Lin
- Department of Preventive Dentistry, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, 56 Ling Yuan Road West, Guangzhou, 510055, China. .,Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-Sen University, Guangzhou, China.
| | - Yan Zhou
- Department of Preventive Dentistry, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, 56 Ling Yuan Road West, Guangzhou, 510055, China. .,Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
35
|
Naradasu D, Guionet A, Miran W, Okamoto A. Microbial current production from Streptococcus mutans correlates with biofilm metabolic activity. Biosens Bioelectron 2020; 162:112236. [DOI: 10.1016/j.bios.2020.112236] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Revised: 04/10/2020] [Accepted: 04/22/2020] [Indexed: 11/25/2022]
|
36
|
Shanmugam K, Sarveswari HB, Udayashankar A, Swamy SS, Pudipeddi A, Shanmugam T, Solomon AP, Neelakantan P. Guardian genes ensuring subsistence of oral Streptococcus mutans. Crit Rev Microbiol 2020; 46:475-491. [PMID: 32720594 DOI: 10.1080/1040841x.2020.1796579] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Despite the substantial research advancements on oral diseases, dental caries remains a major healthcare burden. A disease of microbial dysbiosis, dental caries is characterised by the formation of biofilms that assist demineralisation and destruction of the dental hard tissues. While it is well understood that this is a multi-kingdom biofilm-mediated disease, it has been elucidated that acid producing and acid tolerant bacteria play pioneering roles in the process. Specifically, Streptococcus mutans houses major virulence pathways that enable it to thrive in the oral cavity and cause caries. This pathogen adheres to the tooth substrate, forms biofilms, resists external stress, produces acids, kills closely related species, and survives the acid as well as the host clearance mechanisms. For an organism to be able to confer such virulence, it requires a large and complex gene network which synergise to establish disease. In this review, we have charted how these multi-faceted genes control several caries-related functions of Streptococcus mutans. In a futuristic thinking approach, we also briefly discuss the potential roles of omics and machine learning, to ease the study of non-functional genes that may play a major role and enable the integration of experimental data.
Collapse
Affiliation(s)
- Karthi Shanmugam
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - Hema Bhagavathi Sarveswari
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - Akshaya Udayashankar
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - Shogan Sugumar Swamy
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - Akhila Pudipeddi
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - Tamilarasi Shanmugam
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - Adline Princy Solomon
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical and Biotechnology, SASTRA Deemed to be University, Thanjavur, India
| | - Prasanna Neelakantan
- Division of Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong, Hong Kong
| |
Collapse
|
37
|
Complete Genome Sequences of Two Mutacin-Producing Streptococcus mutans Strains, T8 and UA140. Microbiol Resour Announc 2020; 9:9/24/e00469-20. [PMID: 32527777 PMCID: PMC7291102 DOI: 10.1128/mra.00469-20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Streptococcus mutans is known to produce various antimicrobial peptides called mutacins. Two clinical isolates, T8 and UA140, are well characterized regarding their mutacin production, but genome sequence information was previously unavailable. Complete genome sequences of these two mutacin-producing strains are reported here. Streptococcus mutans is known to produce various antimicrobial peptides called mutacins. Two clinical isolates, T8 and UA140, are well characterized regarding their mutacin production, but genome sequence information was previously unavailable. Complete genome sequences of these two mutacin-producing strains are reported here.
Collapse
|
38
|
Karygianni L, Ren Z, Koo H, Thurnheer T. Biofilm Matrixome: Extracellular Components in Structured Microbial Communities. Trends Microbiol 2020; 28:668-681. [PMID: 32663461 DOI: 10.1016/j.tim.2020.03.016] [Citation(s) in RCA: 709] [Impact Index Per Article: 141.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 03/16/2020] [Accepted: 03/25/2020] [Indexed: 02/04/2023]
Abstract
Biofilms consist of microbial communities embedded in a 3D extracellular matrix. The matrix is composed of a complex array of extracellular polymeric substances (EPS) that contribute to the unique attributes of biofilm lifestyle and virulence. This ensemble of chemically and functionally diverse biomolecules is termed the 'matrixome'. The composition and mechanisms of EPS matrix formation, and its role in biofilm biology, function, and microenvironment are being revealed. This perspective article highlights recent advances about the multifaceted role of the 'matrixome' in the development, physical-chemical properties, and virulence of biofilms. We emphasize that targeting biofilm-specific conditions such as the matrixome could lead to precise and effective antibiofilm approaches. We also discuss the limited knowledge in the context of polymicrobial biofilms, and the need for more in-depth analyses of the EPS matrix in mixed communities that are associated with many human infectious diseases.
Collapse
Affiliation(s)
- L Karygianni
- Clinic of Conservative and Preventive Dentistry, Center of Dental Medicine University of Zurich, Zurich, Switzerland
| | - Z Ren
- Department of Orthodontics, Divisions of Pediatric Dentistry and Community of Oral Health, University of Pennsylvania School of Dental Medicine, Philadelphia, PA, USA
| | - H Koo
- Department of Orthodontics, Divisions of Pediatric Dentistry and Community of Oral Health, University of Pennsylvania School of Dental Medicine, Philadelphia, PA, USA; Center for Innovation and Precision Dentistry, University of Pennsylvania School of Dental Medicine, School of Engineering and Applied Sciences, Philadelphia, PA, USA
| | - T Thurnheer
- Clinic of Conservative and Preventive Dentistry, Center of Dental Medicine University of Zurich, Zurich, Switzerland.
| |
Collapse
|
39
|
Abstract
Technological advancements have revolutionized our understanding of the complexity and importance of the human microbiome. This progress has also emphasized the need for precision therapeutics, as it has underscored the dilemmas, such as dysbiosis and increasing antibiotic resistance, associated with current, broad-spectrum treatment modalities. Dental caries remains the most common chronic disease worldwide, accompanied by a tremendous financial and social burden, despite widespread and efficacious fluoride and hygienic regimens. Over the past several decades, various precision approaches to combat dental caries, including vaccines, probiotics, and antimicrobial compounds, have been pursued. Despite the distinct overall conceptual strengths of each approach, for various reasons, there are currently no approved precision antibiotic therapeutics to prevent dental caries. Specifically targeted antimicrobial peptides (STAMPs) are synthetic molecules that combine the antibiotic moiety of a traditional antimicrobial peptide with a targeting domain to provide specificity against a particular organism. Conjoining the killing domain from the antimicrobial, novispirin G10, and a targeting domain derived from the Streptococcus mutans pheromone, CSP, the STAMP C16G2 was designed to provide targeted killing of S. mutans, widely considered the keystone species in dental caries pathogenesis. C16G2 was able to selectively eliminate S. mutans from complex ecosystems while leaving closely related, yet health-associated, oral species unharmed. This remodeling of the dental plaque community is expected to have significant advantages compared to conventional broad-spectrum mouthwashes, as the intact, surviving community is apt to prevent reinfection by pathogens. Following successful phase I clinical trials that evaluated the safety and basic microbiology of C16G2 treatments, the phase II trials of several C16G2 formulations are currently in progress. C16G2 represents an exciting advance in precision therapeutics, and the STAMP platform provides vast opportunities for both the development of additional therapeutics and the overall study of microbial ecology.
Collapse
Affiliation(s)
- J L Baker
- Genomic Medicine Group, J. Craig Venter Institute, La Jolla, CA, USA
| | - X He
- The Forsyth Institute, Cambridge, MA, USA
| | - W Shi
- The Forsyth Institute, Cambridge, MA, USA
| |
Collapse
|
40
|
Chen Y, Agnello M, Dinis M, Chien KC, Wang J, Hu W, Shi W, He X, Zou J. Lollipop containing Glycyrrhiza uralensis extract reduces Streptococcus mutans colonization and maintains oral microbial diversity in Chinese preschool children. PLoS One 2019; 14:e0221756. [PMID: 31442287 PMCID: PMC6707631 DOI: 10.1371/journal.pone.0221756] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 08/14/2019] [Indexed: 02/05/2023] Open
Abstract
The anticariogenic activity of the extract of Glycyrrhiza uralensis (licorice) has been well documented. We recently developed an herbal lollipop containing licorice extracts with Glycyrrhizol A, the compound displaying strong antimicrobial activity against Streptococcus mutans. Preliminary testing showed that the herbal lollipop reduced salivary S. mutans counts in vivo. In this study, we aimed to further test the efficacy of this herbal lollipop for reducing salivary S. mutans levels, and investigate its impact on salivary microbiome. Using a well-established in vitro oral microbiome model, we showed that licorice extract displays targeted killing against S. mutans without affecting the biodiversity of the community. In vivo study corroborated in vitro findings, showing for high caries-risk children aged 3–6 with salivary S. mutans levels >5x105 cells/ml, daily use of 2 licorice-containing lollipops for 3 weeks significantly reduced salivary S. mutans levels compared to the control group. Salivary microbiome analysis showed either no change or even increase in phylogenetic diversity of the oral community following herbal lollipop usage. Although further study with longer term observation is needed, these results suggest that use of licorice extract-containing lollipops can be as a simple and effective way to reduce the risk of dental caries in children.
Collapse
Affiliation(s)
- Yandi Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Melissa Agnello
- School of Dentistry, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Márcia Dinis
- School of Dentistry, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Kenneth C. Chien
- School of Dentistry, University of California, San Francisco, San Francisco, California, United States of America
| | - Jing Wang
- College of Pharmaceutical Science, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wei Hu
- State Key Lab of Microbial Technology, Microbial Technology Institute, Shandong University, Qingdao, China
| | - Wenyuan Shi
- The Forsyth Institute, Cambridge, Massachusetts, United States of America
| | - Xuesong He
- The Forsyth Institute, Cambridge, Massachusetts, United States of America
- * E-mail: (JZ); (XH)
| | - Jing Zou
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- * E-mail: (JZ); (XH)
| |
Collapse
|
41
|
Chen X, Liu C, Peng X, He Y, Liu H, Song Y, Xiong K, Zou L. Sortase A‐mediated modification of the
Streptococcus mutans
transcriptome and virulence traits. Mol Oral Microbiol 2019; 34:219-233. [PMID: 31342653 DOI: 10.1111/omi.12266] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 07/16/2019] [Accepted: 07/18/2019] [Indexed: 02/05/2023]
Affiliation(s)
- Xuan Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases Sichuan University Chengdu China
| | - Chengcheng Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases Sichuan University Chengdu China
- Department of Periodontics West China Hospital of Stomatology, Sichuan University Chengdu China
| | - Xian Peng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases Sichuan University Chengdu China
| | - Yuanli He
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases Sichuan University Chengdu China
| | - Haixia Liu
- Stomatological Hospital of Chongqing Medical University Chongqing China
| | - Ying Song
- Stomatological Hospital of Chongqing Medical University Chongqing China
| | - Kaixin Xiong
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases Sichuan University Chengdu China
| | - Ling Zou
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases Sichuan University Chengdu China
- Department of Conservation Dentistry and Endodontics West China Hospital of Stomatology, Sichuan University Chengdu China
| |
Collapse
|
42
|
Park M, Sutherland JB, Rafii F. Effects of nano-hydroxyapatite on the formation of biofilms by Streptococcus mutans in two different media. Arch Oral Biol 2019; 107:104484. [PMID: 31382161 DOI: 10.1016/j.archoralbio.2019.104484] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 06/20/2019] [Accepted: 07/16/2019] [Indexed: 10/26/2022]
Abstract
OBJECTIVES The aim of this study was to examine the effect of nano-hydroxyapatite (nHA) on biofilm formation by Streptococcus mutans, which is actively involved in the initiation of dental caries. DESIGN The effects of nHA on growth and biofilm formation by S. mutans were investigated in two media: a saliva analog medium, basal medium mucin (BMM); and a nutrient-rich medium, brain heart infusion (BHI); in the presence and absence of sucrose. RESULTS Sucrose enhanced the growth of S. mutans in both media. In the presence of sucrose, nHA enhanced bacterial growth and biofilm formation more in BMM medium than in BHI. nHA also affected the transcription of glucosyltransferase (gtf) genes and production of polysaccharide differently in the two media. In BHI medium, the transcription of all three gtf genes, coding for enzymes that synthesize soluble and insoluble glucans from sucrose, was increased more than 3-fold by nHA. However, in BMM medium, only the transcription of gtfB and gtfC, coding for insoluble glucans, was substantially enhanced by nHA. CONCLUSIONS nHA appeared to enhance biofilm formation by increasing glucosyltransferase transcription, which resulted in an increase in production of insoluble glucans. This effect was influenced by the growth conditions.
Collapse
Affiliation(s)
- Miseon Park
- Division of Microbiology, National Center for Toxicological Research, FDA, Jefferson, AR 72079, USA
| | - John B Sutherland
- Division of Microbiology, National Center for Toxicological Research, FDA, Jefferson, AR 72079, USA
| | - Fatemeh Rafii
- Division of Microbiology, National Center for Toxicological Research, FDA, Jefferson, AR 72079, USA.
| |
Collapse
|
43
|
Ikono R, Vibriani A, Wibowo I, Saputro KE, Muliawan W, Bachtiar BM, Mardliyati E, Bachtiar EW, Rochman NT, Kagami H, Xianqi L, Nagamura-Inoue T, Tojo A. Nanochitosan antimicrobial activity against Streptococcus mutans and Candida albicans dual-species biofilms. BMC Res Notes 2019; 12:383. [PMID: 31287001 PMCID: PMC6613267 DOI: 10.1186/s13104-019-4422-x] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 06/29/2019] [Indexed: 01/13/2023] Open
Abstract
OBJECTIVE Chitosan nanoparticle (nanochitosan) has a broad antimicrobial spectrum against diverse pathogenic microorganisms. However, its effect on dental caries-associated microorganisms, such as Streptococcus mutans and Candida albicans is yet to be explored. These microorganisms are known for causing early childhood caries. Therefore, this study was aimed at investigating nanochitosan inhibition capacity against dual-species biofilms of S. mutans and C. albicans. In this study, nanochitosan antimicrobial activity is reported against mono and dual biofilm species of S. mutans and/or C. albicans at 3 and 18 h incubation time. Nanochitosan inhibition capacity was observed through biofilm mass quantity and cell viability. RESULTS The present study successfully synthesized nanochitosan with average diameter of approximately 20-30 nm, and also established dual-species biofilms of S. mutans and C. albicans in vitro. With nanochitosan treatment, the cell viability of both microorganisms significantly decreased with the increasing concentration of nanochitosan. There was no significant decrease in biofilm mass both in the dual and single-species biofilms after 3 h of incubation. However, greater inhibition of biofilm was observed at 18 h incubation.
Collapse
Affiliation(s)
- Radyum Ikono
- Division of Bionanotechnology, Nano Center Indonesia, Tangerang Selatan, Indonesia
- Department of Metallurgical Engineering, Sumbawa University of Technology, Sumbawa Besar, Indonesia
- Division of Molecular Therapy, The Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Agnia Vibriani
- School of Life Science and Technology, Bandung Institute of Technology, Bandung, Indonesia
| | - Indra Wibowo
- School of Life Science and Technology, Bandung Institute of Technology, Bandung, Indonesia
| | | | - Wibias Muliawan
- Division of Bionanotechnology, Nano Center Indonesia, Tangerang Selatan, Indonesia
| | - Boy Muchlis Bachtiar
- Oral Science Laboratory, Department of Oral Biology, Faculty of Dentistry, Universitas Indonesia, Jakarta, Indonesia
| | - Etik Mardliyati
- Center for Pharmaceutical and Medical Technology, Agency for the Assessment and Application of Technology [BPPT], Tangerang Selatan, Indonesia
| | - Endang Winiati Bachtiar
- Oral Science Laboratory, Department of Oral Biology, Faculty of Dentistry, Universitas Indonesia, Jakarta, Indonesia
| | - Nurul Taufiqu Rochman
- Research Center for Physics, Indonesian Institute of Science [LIPI], Tangerang Selatan, Indonesia
| | - Hideaki Kagami
- Department of Oral and Maxillofacial Surgery, Matsumoto Dental University, Shiojiri, Japan
- Department of General Medicine, IMSUT Hospital, The Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Li Xianqi
- Department of Oral and Maxillofacial Surgery, Matsumoto Dental University, Shiojiri, Japan
| | - Tokiko Nagamura-Inoue
- Department of Cell Processing and Transfusion, The Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Arinobu Tojo
- Division of Molecular Therapy, The Institute of Medical Science, University of Tokyo, Tokyo, Japan
| |
Collapse
|
44
|
Castillo Pedraza MC, Rosalen PL, de Castilho ARF, Freires IDA, de Sales Leite L, Faustoferri RC, Quivey RG, Klein MI. Inactivation of Streptococcus mutans genes lytST and dltAD impairs its pathogenicity in vivo. J Oral Microbiol 2019; 11:1607505. [PMID: 31143407 PMCID: PMC6522913 DOI: 10.1080/20002297.2019.1607505] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 04/08/2019] [Accepted: 04/09/2019] [Indexed: 12/19/2022] Open
Abstract
Background: Streptococcus mutans orchestrates the development of a biofilm that causes dental caries in the presence of dietary sucrose, and, in the bloodstream, S. mutans can cause systemic infections. The development of a cariogenic biofilm is dependent on the formation of an extracellular matrix rich in exopolysaccharides, which contains extracellular DNA (eDNA) and lipoteichoic acids (LTAs). While the exopolysaccharides are virulence markers, the involvement of genes linked to eDNA and LTAs metabolism in the pathogenicity of S. mutans remains unclear. Objective and Design: In this study, a parental strain S. mutans UA159 and derivative strains carrying single gene deletions were used to investigate the role of eDNA (ΔlytS and ΔlytT), LTA (ΔdltA and ΔdltD), and insoluble exopolysaccharides (ΔgtfB) in virulence in a rodent model of dental caries (rats) and a systemic infection model (Galleria mellonella larvae). Results: Fewer carious lesions were observed on smooth and sulcal surfaces of enamel and dentin of the rats infected with ∆lytS, ∆dltD, and ΔgtfB (vs. the parental strain). Moreover, strains carrying gene deletions prevented the killing of larvae (vs. the parental strain). Conclusions: Altogether, these findings indicate that inactivation of lytST and dltAD impaired S. mutans cariogenicity and virulence in vivo.
Collapse
Affiliation(s)
- Midian C Castillo Pedraza
- Department of Dental Materials and Prosthodontics, Sao Paulo State University (Unesp), School of Dentistry, Araraquara, Brazil
| | - Pedro L Rosalen
- Department of Physiological Sciences, Piracicaba Dental School, University of Campinas - UNICAMP, Piracicaba, Brazil
| | - Aline Rogéria Freire de Castilho
- Department of Physiological Sciences, Piracicaba Dental School, University of Campinas - UNICAMP, Piracicaba, Brazil.,Department of Pediatric Dentistry, Piracicaba Dental School, University of Campinas - UNICAMP, Piracicaba, Brazil
| | - Irlan de Almeida Freires
- Department of Physiological Sciences, Piracicaba Dental School, University of Campinas - UNICAMP, Piracicaba, Brazil
| | - Luana de Sales Leite
- Department of Dental Materials and Prosthodontics, Sao Paulo State University (Unesp), School of Dentistry, Araraquara, Brazil
| | | | - Robert G Quivey
- Center for Oral Biology, University of Rochester, Rochester, NY, USA
| | - Marlise I Klein
- Department of Dental Materials and Prosthodontics, Sao Paulo State University (Unesp), School of Dentistry, Araraquara, Brazil
| |
Collapse
|
45
|
Liu Y, Ren Z, Hwang G, Koo H. Therapeutic Strategies Targeting Cariogenic Biofilm Microenvironment. Adv Dent Res 2018; 29:86-92. [PMID: 29355421 DOI: 10.1177/0022034517736497] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cariogenic biofilms are highly structured microbial communities embedded in an extracellular matrix, a multifunctional scaffold that is essential for the existence of the biofilm lifestyle and full expression of virulence. The extracellular matrix provides the physical and biological properties that enhance biofilm adhesion and cohesion, as well as create a diffusion-modulating milieu, protecting the resident microbes and facilitating the formation of localized acidic pH niches. These biochemical properties pose significant challenges for the development of effective antibiofilm therapeutics to control dental caries. Conventional approaches focusing solely on antimicrobial activity or enhancing remineralization may not achieve maximal efficacy within the complex biofilm microenvironment. Recent approaches disrupting the biofilm microbial community and the microenvironment have emerged, including specific targeting of cariogenic pathogens, modulation of biofilm pH, and synergistic combination of bacterial killing and matrix degradation. Furthermore, new "smart" nanotechnologies that trigger drug release or activation in response to acidic pH are being developed that could enhance the efficacy of current and prospective chemical modalities. Therapeutic strategies that can locally disrupt the pathogenic niche by targeting the biofilm structure and its microenvironment to eliminate the embedded microorganism and facilitate the action of remineralizing agents may lead to enhanced and precise anticaries approaches.
Collapse
Affiliation(s)
- Y Liu
- 1 Biofilm Research Labs, Levy Center for Oral Health, Department of Orthodontics, Divisions of Pediatric Dentistry and Community Oral Health, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Z Ren
- 1 Biofilm Research Labs, Levy Center for Oral Health, Department of Orthodontics, Divisions of Pediatric Dentistry and Community Oral Health, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA.,2 State Key Laboratory of Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - G Hwang
- 1 Biofilm Research Labs, Levy Center for Oral Health, Department of Orthodontics, Divisions of Pediatric Dentistry and Community Oral Health, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - H Koo
- 1 Biofilm Research Labs, Levy Center for Oral Health, Department of Orthodontics, Divisions of Pediatric Dentistry and Community Oral Health, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
46
|
Xu RR, Yang WD, Niu KX, Wang B, Wang WM. An Update on the Evolution of Glucosyltransferase ( Gtf) Genes in Streptococcus. Front Microbiol 2018; 9:2979. [PMID: 30568640 PMCID: PMC6290343 DOI: 10.3389/fmicb.2018.02979] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 11/19/2018] [Indexed: 11/13/2022] Open
Abstract
In many caries-promoting Streptococcus species, glucosyltransferases (Gtfs) are recognized as key enzymes contributing to the modification of biofilm structures, disruption of homeostasis of healthy microbiota community and induction of caries development. It is therefore of great interest to investigate how Gtf genes have evolved in Streptococcus. In this study, we conducted a comprehensive survey of Gtf genes among 872 streptococci genomes of 37 species and identified Gtf genes from 364 genomes of 18 species. To clarify the relationships of these Gtf genes, 45 representative sequences were used for phylogenic analysis, which revealed two clear clades. Clade I included 12 Gtf genes from nine caries-promoting species of the Mutans and Downei groups, which produce enzymes known to synthesize sticky, water-insoluble glucans (WIG) that are critical for modifying biofilm structures. Clade II primarily contained Gtf genes responsible for synthesizing water-soluble glucans (WSG) from all 18 species, and this clade further diverged into three subclades (IIA, IIB, and IIC). An analysis of 16 pairs of duplicated Gtf genes revealed high divergence levels at the C-terminal repeat regions, with ratios of the non-synonymous substitution rate (dN) to synonymous substitution rate (dS) ranging from 0.60 to 1.03, indicating an overall relaxed constraint in this region. However, among the clade I Gtf genes, some individual repeat units possessed strong functional constraints by the same criterion. Structural variations in the repeat regions were also observed, with detection of deletions or recent duplications of individual repeat units. Overall, by establishing an updated phylogeny and further elucidating their evolutionary patterns, this work enabled us to gain a greater understanding of the origination and divergence of Gtf genes in Streptococcus.
Collapse
Affiliation(s)
- Rong-Rong Xu
- Nanjing Stomatological Hospital, Nanjing University Medical School, Nanjing, China
| | - Wei-Dong Yang
- Nanjing Stomatological Hospital, Nanjing University Medical School, Nanjing, China
| | - Ke-Xin Niu
- Laboratory of Plant Genetics and Molecular Evolution, School of Life Sciences, Nanjing University, Nanjing, China
| | - Bin Wang
- Laboratory of Plant Genetics and Molecular Evolution, School of Life Sciences, Nanjing University, Nanjing, China
| | - Wen-Mei Wang
- Nanjing Stomatological Hospital, Nanjing University Medical School, Nanjing, China
| |
Collapse
|
47
|
Abstract
The dynamic and polymicrobial oral microbiome is a direct precursor of diseases such as dental caries and periodontitis, two of the most prevalent microbially induced disorders worldwide. Distinct microenvironments at oral barriers harbour unique microbial communities, which are regulated through sophisticated signalling systems and by host and environmental factors. The collective function of microbial communities is a major driver of homeostasis or dysbiosis and ultimately health or disease. Despite different aetiologies, periodontitis and caries are each driven by a feedforward loop between the microbiota and host factors (inflammation and dietary sugars, respectively) that favours the emergence and persistence of dysbiosis. In this Review, we discuss current knowledge and emerging mechanisms governing oral polymicrobial synergy and dysbiosis that have both enhanced our understanding of pathogenic mechanisms and aided the design of innovative therapeutic approaches for oral diseases.
Collapse
Affiliation(s)
- Richard J Lamont
- Department of Oral Immunology and Infectious Diseases, School of Dentistry, University of Louisville, Louisville, KY, USA.
| | - Hyun Koo
- Department of Orthodontics and Divisions of Pediatric Dentistry and Community Oral Health, Penn Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - George Hajishengallis
- Department of Microbiology, Penn Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
48
|
Yang Y, Reipa V, Liu G, Meng Y, Wang X, Mineart KP, Prabhu VM, Shi W, Lin NJ, He X, Sun J. pH-Sensitive Compounds for Selective Inhibition of Acid-Producing Bacteria. ACS APPLIED MATERIALS & INTERFACES 2018; 10:8566-8573. [PMID: 29436821 PMCID: PMC6852659 DOI: 10.1021/acsami.8b01089] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Stimuli-responsive compounds that provide on-site, controlled antimicrobial activity promise an effective approach to prevent infections, reducing the need for systemic antibiotics. We present a novel pH-sensitive quaternary pyridinium salt (QPS), whose antibacterial activity is boosted by low pH and controlled by adjusting the pH between 4 and 8. Particularly, this compound selectively inhibits growth of acid-producing bacteria within a multispecies community. The successful antibacterial action of this QPS maintains the environmental pH above 5.5, a threshold pH, below which demineralization/erosion takes place. The design, synthesis, and characterization of this QPS and its short-chain analogue are discussed. In addition, their pH-sensitive physicochemical properties in aqueous and organic solutions are evaluated by UV-vis spectroscopy, dynamic light scattering, and NMR spectroscopy. Furthermore, the mechanism of action reveals a switchable assembly that is triggered by acid-base interaction and formed by tightly stacked π-conjugated systems and base moieties. Finally, a model is proposed to recognize the correlated but different mechanisms of pH sensitivity and acid-induced, pH-controlled antibacterial efficacy. We anticipate that successful application of these QPSs and their derivatives will provide protections against infection and erosion through targeted treatments to acid-producing bacteria and modulation of environmental pH.
Collapse
Affiliation(s)
- Yin Yang
- Volpe Research Center, American Dental Association Foundation, Gaithersburg, MD 20899, USA
| | - Vytas Reipa
- Material Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, MD 20899, USA
| | - Guo Liu
- School of Dentistry, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Yuan Meng
- Volpe Research Center, American Dental Association Foundation, Gaithersburg, MD 20899, USA
| | - Xiaohong Wang
- Volpe Research Center, American Dental Association Foundation, Gaithersburg, MD 20899, USA
| | - Kenneth P. Mineart
- Material Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, MD 20899, USA
| | - Vivek M. Prabhu
- Material Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, MD 20899, USA
| | - Wenyuan Shi
- Forsyth Institute, 245 First Street, Cambridge, Massachusetts 02142, USA
| | - Nancy J. Lin
- Material Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, MD 20899, USA
| | - Xuesong He
- Forsyth Institute, 245 First Street, Cambridge, Massachusetts 02142, USA
- Corresponding Author and
| | - Jirun Sun
- Volpe Research Center, American Dental Association Foundation, Gaithersburg, MD 20899, USA
- Corresponding Author and
| |
Collapse
|
49
|
Bowen WH, Burne RA, Wu H, Koo H. Oral Biofilms: Pathogens, Matrix, and Polymicrobial Interactions in Microenvironments. Trends Microbiol 2018; 26:229-242. [PMID: 29097091 PMCID: PMC5834367 DOI: 10.1016/j.tim.2017.09.008] [Citation(s) in RCA: 629] [Impact Index Per Article: 89.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2017] [Revised: 08/26/2017] [Accepted: 09/25/2017] [Indexed: 02/07/2023]
Abstract
Biofilms are microbial communities embedded within an extracellular matrix, forming a highly organized structure that causes many human infections. Dental caries (tooth decay) is a polymicrobial biofilm disease driven by the diet and microbiota-matrix interactions that occur on a solid surface. Sugars fuel the emergence of pathogens, the assembly of the matrix, and the acidification of the biofilm microenvironment, promoting ecological changes and concerted multispecies efforts that are conducive to acid damage of the mineralized tooth tissue. Here, we discuss recent advances in the role of the biofilm matrix and interactions between opportunistic pathogens and commensals in the pathogenesis of dental caries. In addition, we highlight the importance of matrix-producing organisms in fostering a pathogenic habitat where interspecies competition and synergies occur to drive the disease process, which could have implications to other infections associated with polymicrobial biofilms.
Collapse
Affiliation(s)
- William H Bowen
- Center for Oral Biology, Department of Microbiology & Immunology and Environmental Medicine, University of Rochester Medical Center, Rochester, New York, NY, USA; Deceased (15 November 2016)
| | - Robert A Burne
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, FL, USA
| | - Hui Wu
- Departments of Microbiology and Pediatric Dentistry, School of Dentistry, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hyun Koo
- Levy Center for Oral Health, Department of Orthodontics, Divisions of Pediatric Dentistry and Community of Oral Health, University of Pennsylvania School of Dental Medicine, Philadelphia, PA, USA.
| |
Collapse
|
50
|
Wasfi R, Abd El‐Rahman OA, Zafer MM, Ashour HM. Probiotic Lactobacillus sp. inhibit growth, biofilm formation and gene expression of caries-inducing Streptococcus mutans. J Cell Mol Med 2018; 22:1972-1983. [PMID: 29316223 PMCID: PMC5824418 DOI: 10.1111/jcmm.13496] [Citation(s) in RCA: 154] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 11/16/2017] [Indexed: 01/01/2023] Open
Abstract
Streptococcus mutans contributes significantly to dental caries, which arises from homoeostasic imbalance between host and microbiota. We hypothesized that Lactobacillus sp. inhibits growth, biofilm formation and gene expression of Streptococcus mutans. Antibacterial (agar diffusion method) and antibiofilm (crystal violet assay) characteristics of probiotic Lactobacillus sp. against Streptococcus mutans (ATCC 25175) were evaluated. We investigated whether Lactobacillus casei (ATCC 393), Lactobacillus reuteri (ATCC 23272), Lactobacillus plantarum (ATCC 14917) or Lactobacillus salivarius (ATCC 11741) inhibit expression of Streptococcus mutans genes involved in biofilm formation, quorum sensing or stress survival using quantitative real-time polymerase chain reaction (qPCR). Growth changes (OD600) in the presence of pH-neutralized, catalase-treated or trypsin-treated Lactobacillus sp. supernatants were assessed to identify roles of organic acids, peroxides and bacteriocin. Susceptibility testing indicated antibacterial (pH-dependent) and antibiofilm activities of Lactobacillus sp. against Streptococcus mutans. Scanning electron microscopy revealed reduction in microcolony formation and exopolysaccharide structural changes. Of the oral normal flora, L. salivarius exhibited the highest antibiofilm and peroxide-dependent antimicrobial activities. All biofilm-forming cells treated with Lactobacillus sp. supernatants showed reduced expression of genes involved in exopolysaccharide production, acid tolerance and quorum sensing. Thus, Lactobacillus sp. can inhibit tooth decay by limiting growth and virulence properties of Streptococcus mutans.
Collapse
Affiliation(s)
- Reham Wasfi
- Department of Microbiology and ImmunologyFaculty of PharmacyOctober University for Modern Sciences and Arts (MSA)GizaEgypt
| | - Ola A. Abd El‐Rahman
- Department of Microbiology and ImmunologyFaculty of PharmacyAl‐Azhar University (Girls)CairoEgypt
| | - Mai M. Zafer
- Department of Microbiology and ImmunologyFaculty of PharmacyAhram Canadian University (ACU)GizaEgypt
| | - Hossam M. Ashour
- Department of Biological SciencesCollege of Arts and SciencesUniversity of South Florida St. PetersburgSt. PetersburgFLUSA
- Department of Microbiology and ImmunologyFaculty of PharmacyCairo UniversityCairoEgypt
| |
Collapse
|