1
|
Miranda ML, Salomão KB, Botazzo Delbem AC, Danelon M, Oliveira Barbosa ER, Sampaio C, Campos LA, Brighenti FL. Arginine combination with fluoride and calcium glycerophosphate: effects of concentration and on biofilm fluid. Future Microbiol 2024:1-12. [PMID: 39508342 DOI: 10.1080/17460913.2024.2411921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 09/30/2024] [Indexed: 11/15/2024] Open
Abstract
Aim: To study the influence of varying concentrations of arginine (Arg) combined with fluoride (F) and/or calcium glycerophosphate (CaGP) on biofilms.Materials & methods: Biofilms were analyzed for acidogenicity, microbial viability and Ca, F and inorganic phosphorus (P) concentrations.Results: For total bacteria, the lowest viability was found in F-containing groups, regardless of the arginine concentrations and presence of CaGP. For aciduric bacteria, no significant differences were found among arginine concentrations in the presence of F. For MS, arginine concentrations did not influence MS viability in the presence of fluoride and CaGP only decreased viability at 3.2% Arg concentration. The arginine-treated groups showed the lowest acidogenicity. For ion concentrations in biofilms, CaGP showed the highest values for P; Arg+F for F; and CaGP/Arg+CaGP for Ca.Conclusion: Different concentrations of arginine did not affect the microbial viability or acidogenicity of biofilms. Moreover, 0.8% Arg did not increase ion concentration in biofilm fluid.
Collapse
Affiliation(s)
- Marina Lins Miranda
- Universidade Estadual Paulista (UNESP), Faculdade de Odontologia, Departamento de Morfologia e Clinica Infantil, Araraquara, SP, 14801-903, Brasil
| | - Karina Borges Salomão
- Universidade Estadual Paulista (UNESP), Faculdade de Odontologia, Departamento de Morfologia e Clinica Infantil, Araraquara, SP, 14801-903, Brasil
| | - Alberto Carlos Botazzo Delbem
- Universidade Estadual Paulista (UNESP), Faculdade de Odontologia, Departamento de Odontologia Preventiva e Restauradora, Araçatuba, SP, 16015-050, Brasil
| | - Marcelle Danelon
- Polyclinic of Operative Dentistry, Periodontology and Pediatric Dentistry, Faculty of Medicine Carl Gustav Carus, TU Dresden, Fetscherstraße 74, Dresden, 01307, Germany
| | - Elis Rodrigues Oliveira Barbosa
- Universidade Estadual Paulista (UNESP), Faculdade de Odontologia, Departamento de Morfologia e Clinica Infantil, Araraquara, SP, 14801-903, Brasil
| | - Caio Sampaio
- Universidade Estadual Paulista (UNESP), Faculdade de Odontologia, Departamento de Odontologia Preventiva e Restauradora, Araçatuba, SP, 16015-050, Brasil
| | - Lucas Arrais Campos
- Institute of Dentistry, Faculty of Health Sciences, University of Eastern Finland, Kuopio, FI-70211, Finland
- Dental Sciences Graduate Program, São Paulo State University (UNESP), School of Dentistry, Araraquara, 14801-903,Brazil
- Faculty of Medicine and Health Technology, Tampere University, Tampere, FI-33520, Finland
- Department of Ear & Oral Diseases, Tampere University Hospital, Tampere, FI-33520, Finland
| | - Fernanda Lourenção Brighenti
- Universidade Estadual Paulista (UNESP), Faculdade de Odontologia, Departamento de Morfologia e Clinica Infantil, Araraquara, SP, 14801-903, Brasil
| |
Collapse
|
2
|
Snell AP, Manias DA, Elbehery RR, Dunny GM, Willett JLE. Arginine impacts aggregation, biofilm formation, and antibiotic susceptibility in Enterococcus faecalis. FEMS MICROBES 2024; 5:xtae030. [PMID: 39524554 PMCID: PMC11549559 DOI: 10.1093/femsmc/xtae030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 08/26/2024] [Accepted: 09/24/2024] [Indexed: 11/16/2024] Open
Abstract
Enterococcus faecalis is a commensal bacterium in the gastrointestinal (GI) tract of humans and other organisms. E. faecalis also causes infections in root canals, wounds, the urinary tract, and on heart valves. E. faecalis metabolizes arginine through the arginine deiminase pathway, which converts arginine to ornithine and releases ATP, ammonia, and CO2. E. faecalis arginine metabolism also affects virulence of other pathogens during co-culture. E. faecalis may encounter elevated levels of arginine in the GI tract or the oral cavity, where arginine is used as a dental therapeutic. Little is known about how E. faecalis responds to growth in arginine in the absence of other bacteria. To address this, we used RNAseq and additional assays to measure growth, gene expression, and biofilm formation in E. faecalis OG1RF grown in arginine. We demonstrate that arginine decreases E. faecalis biofilm production and causes widespread differential expression of genes related to metabolism, quorum sensing, and polysaccharide synthesis. Growth in arginine also increases aggregation of E. faecalis and promotes decreased susceptibility to the antibiotics ampicillin and ceftriaxone. This work provides a platform for understanding how the presence of arginine in biological niches affects E. faecalis physiology and virulence of surrounding microbes.
Collapse
Affiliation(s)
- Alex P Snell
- University of Minnesota Medical School, Minneapolis, MN 55455, United States
| | - Dawn A Manias
- University of Minnesota Medical School, Minneapolis, MN 55455, United States
| | - Reham R Elbehery
- University of Minnesota Medical School, Minneapolis, MN 55455, United States
| | - Gary M Dunny
- University of Minnesota Medical School, Minneapolis, MN 55455, United States
| | - Julia L E Willett
- University of Minnesota Medical School, Minneapolis, MN 55455, United States
| |
Collapse
|
3
|
Santos ALC, Correr-Sobrinho L, Tsuzuki FM, Facury Ferraz AG, Neves JG, Sinhoreti MAC, Franco EM, Costa AR. Effect of adding arginine at different concentrations to experimental orthodontic resins: an in vitro study. Braz Oral Res 2024; 38:e078. [PMID: 39258631 PMCID: PMC11376598 DOI: 10.1590/1807-3107bor-2024.vol38.0078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 03/03/2024] [Indexed: 09/12/2024] Open
Abstract
The aim of this study was to assess the effect of adding arginine at different concentrations to commercial and experimental orthodontic resins on shear bond strength (SBS), as well as on the antimicrobial activity of arginine against S. mutans. Metal brackets were bonded onto the surface of 120 bovine incisors using Transbond, OrthoCem, and an experimental resin (ER), adding 0, 2.5, 5, and 7 wt.% of arginine. The SBS test was performed in deionized water at 37 ºC for 24 h, at 0.5 mm/min. SBS test results were subjected to two-way ANOVA and Tukey's test (α = 0.05). CFU/mL data (antimicrobial assessment) were assessed by Kruskal-Wallis and Dunn's tests (α = 0.05). No statistical difference between the resins was observed in untreated groups (p > 0.05). The addition of arginine at 2.5% (27.7 MPa) and 5% (29.0 MPa) increased the SBS of Transbond when compared (p < 0.05) to OrthoCem (18.5 and 15.6 MPa, respectively) and ER (16.3 and 18.1 MPa, respectively). Arginine at 7% improved the SBS of Transbond (24.1 MPa) and ER (21.0 MPa), which was statistically higher (p < 0.05) than OrthoCem (12.6 MPa). OrthoCem did not show a statistically significant difference at the three concentrations of arginine (p > 0.05). The addition of arginine to resins reduced the count of S. mutans (p < 0.05). As for ER, all concentrations of arginine significantly decreased CFU/mL (p < 0.05). Among commercial resins, only 7% of arginine significantly reduced CFU/mL. The addition of arginine did not interfere with the bond strength and demonstrated antibacterial activity against S. mutans.
Collapse
Affiliation(s)
| | - Lourenço Correr-Sobrinho
- Universidade Estadual de Campinas - Unicamp, Piracicaba Dental School, Department of Restorative Dentistry, Piracicaba, SP, Brazil
| | - Fernanda Midori Tsuzuki
- Universidade Estadual de Campinas - Unicamp, Piracicaba Dental School, Department of Restorative Dentistry, Piracicaba, SP, Brazil
| | - Anália Gabriela Facury Ferraz
- Universidade Estadual de Campinas - Unicamp, Piracicaba Dental School, Department of Restorative Dentistry, Piracicaba, SP, Brazil
| | - José Guilherme Neves
- Universidade Estadual de Campinas - Unicamp, Piracicaba Dental School, Department of Restorative Dentistry, Piracicaba, SP, Brazil
| | - Mário Alexandre Coelho Sinhoreti
- Universidade Estadual de Campinas - Unicamp, Piracicaba Dental School, Department of Restorative Dentistry, Piracicaba, SP, Brazil
| | - Eduardo Martinelli Franco
- Universidade Estadual de Campinas - Unicamp, Piracicaba Dental School, Department of Microbiology, Piracicaba, SP, Brazil
| | - Ana Rosa Costa
- Fundação Hermínio Ometto, Department of Orthodontics, Araras, SP, Brazil
| |
Collapse
|
4
|
Miranda ML, Danelon M, Delbem ACB, Kopp W, Nunes GP, Brighenti FL. Enhanced anti-biofilm and anti-caries potential of arginine combined with calcium glycerophosphate and fluoride. J Dent 2024; 146:105039. [PMID: 38714243 DOI: 10.1016/j.jdent.2024.105039] [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: 01/26/2024] [Revised: 05/01/2024] [Accepted: 05/02/2024] [Indexed: 05/09/2024] Open
Abstract
OBJECTIVE The aim of this work was to evaluate the antibiofilm and anticaries properties of the association of arginine (Arg) with calcium glycerophosphate (CaGP) and fluoride (F). METHODS An active attachment, polymicrobial biofilm model obtained from saliva and bovine teeth discs were used. After the initial biofilm growth period, the enamel discs were transferred to culture medium. The treatment solutions were added to the culture media to achieve the desired final concentration. The following groups were used: negative control (Control); F (110 ppm F); CaGP (0.05 %); Arg (0.8 %) and their associations (F + CaGP; Arg + F; Arg + CaGP; Arg +F + CaGP). The following analyses were carried out: bacterial viability (total bacteria, aciduric bacteria and mutans streptococci), pH assessment of the spent culture medium, dry weight quantification, evaluation of surface hardness loss (%SH) and subsurface mineral content. Normality and homoscedasticity were tested (Shapiro-Wilk and Levene's test) and the following tests were applied: two-way ANOVA (acidogenicity), Kruskall-Wallis (microbial viability) and one way ANOVA (dry weight, %SH, mineral content). RESULTS The association Arg + F + CaGP resulted in the lowest surface hardness loss in tooth enamel (-10.9 ± 2.3 %; p < 0.05). Arg +F + CaGP exhibited highest values of subsurface mineral content (10.1 ± 2.9 gHAP/cm3) in comparison to Control and F (p < 0.05). In comparison to Control and F, Arg +F + CaGP promoted the highest reduction in aciduric bacteria and mutans streptococci (5.7 ± 0.4; 4.4 ± 0.5 logCFU/mL, p < 0.05). CONCLUSIONS The Arg-F-Ca association demonstrated to be the most effective combination in protecting the loss of surface hardness and subsurface mineral content, in addition to controlling important virulence factors of the cariogenic biofilm. CLINICAL SIGNIFICANCE Our findings provide evidence that the Arg-F-Ca association showed an additive effect, particularly concerning protection against enamel demineralization. The combination of these compounds may be a strategy for patients at high risk of caries.
Collapse
Affiliation(s)
- Marina Lins Miranda
- São Paulo State University (UNESP), School of Dentistry, Araraquara, Department of Morphology, Orthodontics and Pediatric Dentistry, Araraquara, SP, Brazil
| | - Marcelle Danelon
- São Paulo State University (UNESP), School of Dentistry, Araçatuba, Universidade Estadual Paulista (UNESP), Department of Preventive and Restorative Dentistry, Araçatuba, SP, Brasil
| | - Alberto Carlos Botazzo Delbem
- São Paulo State University (UNESP), School of Dentistry, Araçatuba, Universidade Estadual Paulista (UNESP), Department of Preventive and Restorative Dentistry, Araçatuba, SP, Brasil
| | - Willian Kopp
- São Paulo State University (UNESP), School of Dentistry, Araraquara, Department of Morphology, Orthodontics and Pediatric Dentistry, Araraquara, SP, Brazil
| | - Gabriel Pereira Nunes
- São Paulo State University (UNESP), School of Dentistry, Araçatuba, Universidade Estadual Paulista (UNESP), Department of Preventive and Restorative Dentistry, Araçatuba, SP, Brasil
| | - Fernanda Lourenção Brighenti
- São Paulo State University (UNESP), School of Dentistry, Araraquara, Department of Morphology, Orthodontics and Pediatric Dentistry, Araraquara, SP, Brazil.
| |
Collapse
|
5
|
Sampaio C, Méndez DAC, Buzalaf MAR, Pessan JP, Cruvinel T. Arginine and sodium fluoride affect the microbial composition and reduce biofilm metabolism and enamel mineral loss in an oral microcosm model. J Dent 2024; 145:104997. [PMID: 38621525 DOI: 10.1016/j.jdent.2024.104997] [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: 02/05/2024] [Revised: 04/11/2024] [Accepted: 04/12/2024] [Indexed: 04/17/2024] Open
Abstract
OBJECTIVE To assess the effects of arginine, with or without sodium fluoride (NaF; 1,450 ppm), on saliva-derived microcosm biofilms and enamel demineralization. METHODS Saliva-derived biofilms were grown on bovine enamel blocks in 0.2 % sucrose-containing modified McBain medium, according to six experimental groups: control (McBain 0.2 %); 2.5 % arginine; 8 % arginine; NaF; 2.5 % arginine with NaF; and 8 % arginine with NaF. After 5 days of growth, biofilm viability was assessed by colony-forming units counting, laser scanning confocal microscopy was used to determine biofilm vitality and extracellular polysaccharide (EPS) production, while biofilm metabolism was evaluated using the resazurin assay and lactic acid quantification. Demineralization was evaluated by measuring pH in the culture medium and calcium release. Data were analyzed by Kruskal-Wallis' and Dunn's tests (p < 0.05). RESULTS 8 % arginine with NaF showed the strongest reduction in total streptococci and total microorganism counts, with no significant difference compared to arginine without NaF. Neither 2.5 % arginine alone nor NaF alone significantly reduced microbial counts compared to the control, although in combination, a reduction in all microbial groups was observed. Similar trends were found for biofilm vitality and EPS, and calcium released to the growth medium. CONCLUSIONS 8 % Arginine, with or without NaF, exhibited the strongest antimicrobial activity and reduced enamel calcium loss. Also, NaF enhanced the effects of 2.5 % arginine, yielding similar results to 8 % arginine for most parameters analyzed. CLINICAL SIGNIFICANCE The results provided further evidence on how arginine, with or without NaF, affects oral microcosm biofilms and enamel mineral loss.
Collapse
Affiliation(s)
- Caio Sampaio
- Department of Preventive and Restorative Dentistry, São Paulo State University, School of Dentistry, Araçatuba, Brazil
| | - Daniela Alejandra Cusicanqui Méndez
- Department of Pediatric Dentistry, Orthodontics and Public Health, Bauru School of Dentistry, University of São Paulo, Alameda Dr. Octávio Pinheiro Brisolla, 9-75, Vila Universitária SP, Bauru 17012-901, Brazil
| | | | - Juliano Pelim Pessan
- Department of Preventive and Restorative Dentistry, São Paulo State University, School of Dentistry, Araçatuba, Brazil
| | - Thiago Cruvinel
- Department of Pediatric Dentistry, Orthodontics and Public Health, Bauru School of Dentistry, University of São Paulo, Alameda Dr. Octávio Pinheiro Brisolla, 9-75, Vila Universitária SP, Bauru 17012-901, Brazil.
| |
Collapse
|
6
|
Snell A, Manias DA, Elbehery RR, Dunny GM, Willett JLE. Arginine impacts aggregation, biofilm formation, and antibiotic susceptibility in Enterococcus faecalis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.30.596650. [PMID: 38853917 PMCID: PMC11160706 DOI: 10.1101/2024.05.30.596650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Enterococcus faecalis is a commensal bacterium in the gastrointestinal tract (GIT) of humans and other organisms. E. faecalis also causes infections in root canals, wounds, the urinary tract, and on heart valves. E. faecalis metabolizes arginine through the arginine deiminase (ADI) pathway, which converts arginine to ornithine and releases ATP, ammonia, and CO2. E. faecalis arginine metabolism also affects virulence of other pathogens during co-culture. E. faecalis may encounter elevated levels of arginine in the GIT or the oral cavity, where arginine is used as a dental therapeutic. Little is known about how E. faecalis responds to growth in arginine in the absence of other bacteria. To address this, we used RNAseq and additional assays to measure growth, gene expression, and biofilm formation in E. faecalis OG1RF grown in arginine. We demonstrate that arginine decreases E. faecalis biofilm production and causes widespread differential expression of genes related to metabolism, quorum sensing, and polysaccharide synthesis. Growth in arginine also increases aggregation of E. faecalis and promotes decreased susceptibility to the antibiotics ampicillin and ceftriaxone. This work provides a platform for understanding of how the presence of arginine in biological niches affects E. faecalis physiology and virulence of surrounding microbes.
Collapse
Affiliation(s)
- Alex Snell
- University of Minnesota Medical School, Minneapolis, MN, 55455
| | - Dawn A. Manias
- University of Minnesota Medical School, Minneapolis, MN, 55455
| | | | - Gary M. Dunny
- University of Minnesota Medical School, Minneapolis, MN, 55455
| | | |
Collapse
|
7
|
Willmott T, Serrage HJ, Cottrell EC, Humphreys GJ, Myers J, Campbell PM, McBain AJ. Investigating the association between nitrate dosing and nitrite generation by the human oral microbiota in continuous culture. Appl Environ Microbiol 2024; 90:e0203523. [PMID: 38440981 PMCID: PMC11022587 DOI: 10.1128/aem.02035-23] [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: 11/14/2023] [Accepted: 02/13/2024] [Indexed: 03/06/2024] Open
Abstract
The generation of nitrite by the oral microbiota is believed to contribute to healthy cardiovascular function, with oral nitrate reduction to nitrite associated with systemic blood pressure regulation. There is the potential to manipulate the composition or activities of the oral microbiota to a higher nitrate-reducing state through nitrate supplementation. The current study examined microbial community composition and enzymatic responses to nitrate supplementation in sessile oral microbiota grown in continuous culture. Nitrate reductase (NaR) activity and nitrite concentrations were not significantly different to tongue-derived inocula in model biofilms. These were generally dominated by Streptococcus spp., initially, and a single nitrate supplementation resulted in the increased relative abundance of the nitrate-reducing genera Veillonella, Neisseria, and Proteus spp. Nitrite concentrations increased concomitantly and continued to increase throughout oral microbiota development. Continuous nitrate supplementation, over a 7-day period, was similarly associated with an elevated abundance of nitrate-reducing taxa and increased nitrite concentration in the perfusate. In experiments in which the models were established in continuous low or high nitrate environments, there was an initial elevation in nitrate reductase, and nitrite concentrations reached a relatively constant concentration over time similar to the acute nitrate challenge with a similar expansion of Veillonella and Neisseria. In summary, we have investigated nitrate metabolism in continuous culture oral biofilms, showing that nitrate addition increases nitrate reductase activity and nitrite concentrations in oral microbiota with the expansion of putatively NaR-producing taxa.IMPORTANCEClinical evidence suggests that blood pressure regulation can be promoted by nitrite generated through the reduction of supplemental dietary nitrate by the oral microbiota. We have utilized oral microbiota models to investigate the mechanisms responsible, demonstrating that nitrate addition increases nitrate reductase activity and nitrite concentrations in oral microbiota with the expansion of nitrate-reducing taxa.
Collapse
Affiliation(s)
- Thomas Willmott
- Maternal and Fetal Health Research Centre, Division of Developmental Biology & Medicine, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Hannah J. Serrage
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Elizabeth C. Cottrell
- Maternal and Fetal Health Research Centre, Division of Developmental Biology & Medicine, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Gavin J. Humphreys
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Jenny Myers
- Maternal and Fetal Health Research Centre, Division of Developmental Biology & Medicine, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Paul M. Campbell
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Andrew J. McBain
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
8
|
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
|
9
|
Syahruddin MH, Anggraeni R, Ana ID. A microfluidic organ-on-a-chip: into the next decade of bone tissue engineering applied in dentistry. Future Sci OA 2023; 9:FSO902. [PMID: 37753360 PMCID: PMC10518836 DOI: 10.2144/fsoa-2023-0061] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 08/21/2023] [Indexed: 09/28/2023] Open
Abstract
A comprehensive understanding of the complex physiological and pathological processes associated with alveolar bones, their responses to different therapeutics strategies, and cell interactions with biomaterial becomes necessary in precisely treating patients with severe progressive periodontitis, as a bone-related issue in dentistry. However, existing monolayer cell culture or pre-clinical models have been unable to mimic the complex physiological, pathological and regeneration processes in the bone microenvironment in response to different therapeutic strategies. In this point, 'organ-on-a-chip' (OOAC) technology, specifically 'alveolar-bone-on-a-chip', is expected to resolve the problems by better imitating infection site microenvironment and microphysiology within the oral tissues. The OOAC technology is assessed in this study toward better approaches in disease modeling and better therapeutics strategy for bone tissue engineering applied in dentistry.
Collapse
Affiliation(s)
- Muhammad Hidayat Syahruddin
- Postgraduate Student, Dental Science Doctoral Study Program, Faculty of Dentistry, Universitas Gadjah Mada, Yogyakarta, 55281, Indonesia
| | - Rahmi Anggraeni
- Research Center for Preclinical & Clinical Medicine, National Research & Innovation Agency of the Republic of Indonesia, Cibinong Science Center, Bogor, 16915, Indonesia
- Research Collaboration Center for Biomedical Scaffolds, National Research & Innovation Agency (BRIN) – Universitas Gadjah Mada (UGM), Yogyakarta, 55281, Indonesia
| | - Ika Dewi Ana
- Department of Dental Biomedical Sciences, Faculty of Dentistry, Universitas Gadjah Mada, Yogyakarta, 55281, Indonesia
- Research Collaboration Center for Biomedical Scaffolds, National Research & Innovation Agency (BRIN) – Universitas Gadjah Mada (UGM), Yogyakarta, 55281, Indonesia
| |
Collapse
|
10
|
Ghesquière J, Simoens K, Koos E, Boon N, Teughels W, Bernaerts K. Spatiotemporal monitoring of a periodontal multispecies biofilm model: demonstration of prebiotic treatment responses. Appl Environ Microbiol 2023; 89:e0108123. [PMID: 37768099 PMCID: PMC10617495 DOI: 10.1128/aem.01081-23] [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: 07/01/2023] [Accepted: 07/26/2023] [Indexed: 09/29/2023] Open
Abstract
Biofilms are complex polymicrobial communities which are often associated with human infections such as the oral disease periodontitis. Studying these complex communities under controlled conditions requires in vitro biofilm model systems that mimic the natural environment as close as possible. This study established a multispecies periodontal model in the drip flow biofilm reactor in order to mimic the continuous flow of nutrients at the air-liquid interface in the oral cavity. The design is engineered to enable real-time characterization. A community of five bacteria, Streptococcus gordonii-GFPmut3*, Streptococcus oralis-GFPmut3*, Streptococcus sanguinis-pVMCherry, Fusobacterium nucleatum, and Porphyromonas gingivalis-SNAP26 is visualized using two distinct fluorescent proteins and the SNAP-tag. The biofilm in the reactor develops into a heterogeneous, spatially uniform, dense, and metabolically active biofilm with relative cell abundances similar to those in a healthy individual. Metabolic activity, structural features, and bacterial composition of the biofilm remain stable from 3 to 6 days. As a proof of concept for our periodontal model, the 3 days developed biofilm is exposed to a prebiotic treatment with L-arginine. Multifaceted effects of L-arginine on the oral biofilm were validated by this model setup. L-arginine showed to inhibit growth and incorporation of the pathogenic species and to reduce biofilm thickness and volume. Additionally, L-arginine is metabolized by Streptococcus gordonii-GFPmut3* and Streptococcus sanguinis-pVMCherry, producing high levels of ornithine and ammonium in the biofilm. In conclusion, our drip flow reactor setup is promising in studying spatiotemporal behavior of a multispecies periodontal community.ImportancePeriodontitis is a multifactorial chronic inflammatory disease in the oral cavity associated with the accumulation of microorganisms in a biofilm. Not the presence of the biofilm as such, but changes in the microbiota (i.e., dysbiosis) drive the development of periodontitis, resulting in the destruction of tooth-supporting tissues. In this respect, novel treatment approaches focus on maintaining the health-associated homeostasis of the resident oral microbiota. To get insight in dynamic biofilm responses, our research presents the establishment of a periodontal biofilm model including Streptococcus gordonii, Streptococcus oralis, Streptococcus sanguinis, Fusobacterium nucleatum, and Porphyromonas gingivalis. The added value of the model setup is the combination of simulating continuously changing natural mouth conditions with spatiotemporal biofilm profiling using non-destructive characterization tools. These applications are limited for periodontal biofilm research and would contribute in understanding treatment mechanisms, short- or long-term exposure effects, the adaptation potential of the biofilm and thus treatment strategies.
Collapse
Affiliation(s)
- Justien Ghesquière
- Chemical and Biochemical Reactor Engineering and Safety (CREaS), Department of Chemical Engineering, University of Leuven (KU Leuven), Leuven, Belgium
| | - Kenneth Simoens
- Chemical and Biochemical Reactor Engineering and Safety (CREaS), Department of Chemical Engineering, University of Leuven (KU Leuven), Leuven, Belgium
| | - Erin Koos
- Soft Matter, Rheology and Technology, Department of Chemical Engineering, University of Leuven (KU Leuven), Leuven, Belgium
| | - Nico Boon
- Center for Microbial Ecology and Technology (CMET), Ghent University (UGent), Gent, Belgium
| | - Wim Teughels
- Department of Oral Health Sciences, University of Leuven (KU Leuven) and Dentistry (Periodontology), University Hospitals Leuven, Leuven, Belgium
| | - Kristel Bernaerts
- Chemical and Biochemical Reactor Engineering and Safety (CREaS), Department of Chemical Engineering, University of Leuven (KU Leuven), Leuven, Belgium
| |
Collapse
|
11
|
P Gomes PW, Zuffa S, Bauermeister A, Caraballo-Rodríguez AM, Zhao HN, Mannochio-Russo H, Dogo-Isonagie C, Patel O, Pimenta P, Gronlund J, Lavender S, Pilch S, Maloney V, North M, Dorrestein PC. Ex vivo study of molecular changes of stained teeth following hydrogen peroxide and peroxymonosulfate treatments. Sci Rep 2023; 13:16349. [PMID: 37770593 PMCID: PMC10539445 DOI: 10.1038/s41598-023-43201-y] [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: 04/25/2023] [Accepted: 09/21/2023] [Indexed: 09/30/2023] Open
Abstract
White teeth can give confidence and tend to be associated with a healthier lifestyle in modern society. Therefore, tooth-bleaching strategies have been developed, including the use of hydrogen peroxide. Recently, peroxymonosulfate has been introduced as an alternative bleaching method to hydrogen peroxide. Although both chemicals are oxidizing agents, their effects on the molecular composition of the stained teeth are yet unknown. In this study, the molecular profiles of teeth bleached with hydrogen peroxide and peroxymonosulfate were compared using Liquid Chromatography-Tandem Mass Spectrometry. Statistical analyses were used to assess the samples. In addition, reference spectral libraries and in silico tools were used to perform metabolite annotation. Overall, principal component analysis showed a strong separation between control and hydrogen peroxide and peroxymonosulfate samples (p < 0.001). The analysis of molecular changes revealed amino acids and dipeptides in stained teeth samples after hydrogen peroxide and peroxymonosulfate treatments. Noteworthy, the two bleaching methods led to distinct molecular profiles. For example, diterpenoids were more prevalent after peroxymonosulfate treatment, while a greater abundance of alkaloids was detected after hydrogen peroxide treatment. Whereas non-bleached samples (controls) showed mainly lipids. Therefore, this study shows how two different tooth-whitening peroxides could affect the molecular profiles of human teeth.
Collapse
Affiliation(s)
- Paulo Wender P Gomes
- Skaggs School of Pharmacy and Pharmaceutical Sciences, Collaborative Mass Spectrometry Innovation Center, University of California, San Diego, La Jolla, CA, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Simone Zuffa
- Skaggs School of Pharmacy and Pharmaceutical Sciences, Collaborative Mass Spectrometry Innovation Center, University of California, San Diego, La Jolla, CA, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Anelize Bauermeister
- Skaggs School of Pharmacy and Pharmaceutical Sciences, Collaborative Mass Spectrometry Innovation Center, University of California, San Diego, La Jolla, CA, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Andrés Mauricio Caraballo-Rodríguez
- Skaggs School of Pharmacy and Pharmaceutical Sciences, Collaborative Mass Spectrometry Innovation Center, University of California, San Diego, La Jolla, CA, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Haoqi Nina Zhao
- Skaggs School of Pharmacy and Pharmaceutical Sciences, Collaborative Mass Spectrometry Innovation Center, University of California, San Diego, La Jolla, CA, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Helena Mannochio-Russo
- Skaggs School of Pharmacy and Pharmaceutical Sciences, Collaborative Mass Spectrometry Innovation Center, University of California, San Diego, La Jolla, CA, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA
| | | | - Om Patel
- Colgate-Palmolive, Global Technology Center, Piscataway, NJ, USA
| | - Paloma Pimenta
- Colgate-Palmolive, Global Technology Center, Piscataway, NJ, USA
| | | | - Stacey Lavender
- Colgate-Palmolive, Global Technology Center, Piscataway, NJ, USA
| | - Shira Pilch
- Colgate-Palmolive, Global Technology Center, Piscataway, NJ, USA
| | - Venda Maloney
- Colgate-Palmolive, Global Technology Center, Piscataway, NJ, USA
| | - Michael North
- Colgate-Palmolive, Global Technology Center, Piscataway, NJ, USA
| | - Pieter C Dorrestein
- Skaggs School of Pharmacy and Pharmaceutical Sciences, Collaborative Mass Spectrometry Innovation Center, University of California, San Diego, La Jolla, CA, USA.
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
12
|
Ardila CM, Jiménez-Arbeláez GA, Vivares-Builes AM. Potential Clinical Application of Organs-on-a-Chip in Periodontal Diseases: A Systematic Review of In Vitro Studies. Dent J (Basel) 2023; 11:158. [PMID: 37504224 PMCID: PMC10378380 DOI: 10.3390/dj11070158] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 06/15/2023] [Accepted: 06/23/2023] [Indexed: 07/29/2023] Open
Abstract
The periodontium is a unique organ from the standpoint of building an organ-on-a-chip (OoC) since it is a system that is continually threatened by microorganisms, their noxious compounds, and antigenic components. At the same time, periodontal health depends on a balanced connection between the host and the bacteria in the oral cavity, which is a complex micro-ecological environment. The objective of this systematic review of in vitro studies is to revise the potential clinical application of OoC in periodontal diseases. PRISMA was used to guide this analysis. The review framework made use of several databases, including SCOPUS, PubMed/MEDLINE, SCIELO, and LILACS as well as the gray literature. This systematic review comprised seven studies. The clinical efficacy of OoC in periodontal diseases was observed in models of the gingival crevice for the research of periodontitis, periodontal medication analysis, the interaction of multiple microbial species, pH measurements in in situ-grown biofilm, testing antimicrobial reagents, evaluation of mucosal interactions with microorganisms, and a device for quantitative exploration of microorganisms. OoC has the potential to advance our understanding of periodontal diseases by providing a more accurate representation of the oral microenvironment and enabling the development of new treatments.
Collapse
Affiliation(s)
- Carlos M Ardila
- Basic Studies Department, School of Dentistry, Universidad de Antioquia UdeA, Medellín 050010, Colombia
| | | | | |
Collapse
|
13
|
Malviya J, Alameri AA, Al-Janabi SS, Fawzi OF, Azzawi AL, Obaid RF, Alsudani AA, Alkhayyat AS, Gupta J, Mustafa YF, Karampoor S, Mirzaei R. Metabolomic profiling of bacterial biofilm: trends, challenges, and an emerging antibiofilm target. World J Microbiol Biotechnol 2023; 39:212. [PMID: 37256458 DOI: 10.1007/s11274-023-03651-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 05/17/2023] [Indexed: 06/01/2023]
Abstract
Biofilm-related infections substantially contribute to bacterial illnesses, with estimates indicating that at least 80% of such diseases are linked to biofilms. Biofilms exhibit unique metabolic patterns that set them apart from their planktonic counterparts, resulting in significant metabolic reprogramming during biofilm formation. Differential glycolytic enzymes suggest that central metabolic processes are markedly different in biofilms and planktonic cells. The glycolytic enzyme glyceraldehyde-3-phosphate dehydrogenase (GAPDH) is highly expressed in Staphylococcus aureus biofilm progenitors, indicating that changes in glycolysis activity play a role in biofilm development. Notably, an important consideration is a correlation between elevated cyclic di-guanylate monophosphate (c-di-GMP) activity and biofilm formation in various bacteria. C-di-GMP plays a critical role in maintaining the persistence of Pseudomonas aeruginosa biofilms by regulating alginate production, a significant biofilm matrix component. Furthermore, it has been demonstrated that S. aureus biofilm development is initiated by several tricarboxylic acid (TCA) intermediates in a FnbA-dependent manner. Finally, Glucose 6-phosphatase (G6P) boosts the phosphorylation of histidine-containing protein (HPr) by increasing the activity of HPr kinase, enhancing its interaction with CcpA, and resulting in biofilm development through polysaccharide intercellular adhesion (PIA) accumulation and icaADBC transcription. Therefore, studying the metabolic changes associated with biofilm development is crucial for understanding the complex mechanisms involved in biofilm formation and identifying potential targets for intervention. Accordingly, this review aims to provide a comprehensive overview of recent advances in metabolomic profiling of biofilms, including emerging trends, prevailing challenges, and the identification of potential targets for anti-biofilm strategies.
Collapse
Affiliation(s)
- Jitendra Malviya
- Department of Life Sciences and Biological Sciences, IES University, Bhopal, India
| | - Ameer A Alameri
- Department of Chemistry, College of Science, University of Babylon, Babylon, Iraq
| | - Saif S Al-Janabi
- Medical Laboratory Techniques Department, Al-Maarif University College, Ramadi, Iraq
| | | | | | - Rasha Fadhel Obaid
- Department of Biomedical Engineering, Al-Mustaqbal University College, Babylon, Iraq
| | - Ali A Alsudani
- College of Science, University of Al-Qadisiyah, Al-Diwaniyah, Iraq
| | - Ameer S Alkhayyat
- Medical Laboratory Technology Department, College of Medical Technology, The Islamic University, Najaf, Iraq
| | - Jitendra Gupta
- Institute of Pharmaceutical Research, GLA University, Mathura, 281406, U. P., India
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, 41001, Iraq
| | - Sajad Karampoor
- Gastrointestinal and Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Rasoul Mirzaei
- Venom and Biotherapeutics Molecules Lab, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
14
|
Bijle MN, Abdalla MM, Hung IFN, Yiu CKY. The effect of synbiotic-fluoride therapy on multi-species biofilm. J Dent 2023; 133:104523. [PMID: 37080530 DOI: 10.1016/j.jdent.2023.104523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 04/15/2023] [Accepted: 04/17/2023] [Indexed: 04/22/2023] Open
Abstract
OBJECTIVES The study objective was to examine the effect of synbiotic-fluoride (SF) therapy within a multi-species cariogenic biofilm model system comprising of S. mutans, S. sanguinis, and S. gordonii. METHODS The SF therapy was prepared using 2% L-arginine (Arg), 0.2% NaF and probiotic L. rhamnosus GG (LRG). The 8 treatment groups were: Group 1: No treatment, Group 2: 2% Arg, Group 3: 0.2% NaF, Group 4: LRG, Group 5: 2% Arg+0.2% NaF, Group 6: 2% Arg+LRG, Group 7: 0.2% NaF+LRG, and Group 8: SF therapy (2% Arg+0.2% NaF +LRG). Multi-species biofilm model over 96 h comprising Streptococcus mutans, Streptococcus sanguinis, and Streptococcus gordonii was utilized. The biofilms received cariogenic challenge and SF therapy 2 × /day. The extracellular matrix components were analyzed for carbohydrates, proteins, and extra-cellular DNA (eDNA). The live/dead cells were imaged and quantified using confocal microscopy. The viable/dead bacterial concentrations were estimated using propidium monoazide-quantitative polymerase chain reaction (PMA-qPCR). The gene expressions for gtfB, sagP, arcA, argG, and argH were measured using real-time reverse transcriptase qPCR. RESULTS Carbohydrates and protein content with SF therapy were higher than non-LRG containing groups, while eDNA content was lower than other groups (p<0.05). Live bacterial proportions determined using confocal imaging with SF therapy were the lowest (p<0.05). The 2% Arg+LRG and SF therapy showed higher viable L. rhamnosus GG than 0.2% NaF+LRG (p<0.05). The dead S. mutans with SF therapy were higher than the other groups (p<0.05) with no difference from 2% Arg+0.2% NaF and 2% Arg+LRG (p>0.05). The SF therapy significantly downregulates gtfB and upregulates sagP, arcA, argG, argH gene expression (p<0.05). CONCLUSION Synbiotic-fluoride therapy effectuates multi-fold changes in the multi-species biofilm matrix and cellular components leading to superior ecological homeostasis than its individual contents, prebiotics (arginine), probiotic (L. rhamnosus GG), and fluorides (NaF). CLINICAL SIGNIFICANCE The ecological-based synbiotic-fluoride caries-preventive therapy aids in maintaining biofilm homeostasis to preempt/restore dysbiosis thereby sustaining dynamic-diverse health-associated microbial stability significant as a preventive regimen for high caries-risk patients.
Collapse
Affiliation(s)
- Mohammed Nadeem Bijle
- Assistant Professor in Paediatric Dentistry, Department of Clinical Sciences, College of Dentistry, Ajman University, United Arab Emirates; Center of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates.
| | - Mohamed Mahmoud Abdalla
- Postdoctoral Fellow, Paediatric Dentistry, Faculty of Dentistry, The University of Hong Kong, Hong Kong; Associate Professor, Dental Biomaterials, Faculty of Dental Medicine Al-Azhar University, Cairo, Egypt.
| | - Ivan Fan Ngai Hung
- Ru Chien and Helen Lieh Professor in Health Sciences Pedagogy, Clinical Professor, Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong.
| | - Cynthia Kar Yung Yiu
- Clinical Professor in Paediatric Dentistry, Faculty of Dentistry, The University of Hong Kong, Hong Kong.
| |
Collapse
|
15
|
Gloag ES, Khosravi Y, Masters JG, Wozniak DJ, Amorin Daep C, Stoodley P. A Combination of Zinc and Arginine Disrupt the Mechanical Integrity of Dental Biofilms. Microbiol Spectr 2023; 11:e0335122. [PMID: 36472465 PMCID: PMC9927089 DOI: 10.1128/spectrum.03351-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 11/18/2022] [Indexed: 12/12/2022] Open
Abstract
Mechanical cleaning remains the standard of care for maintaining oral hygiene. However, mechanical cleaning is often augmented with active therapeutics that further promote oral health. A dentifrice, consisting of the "Dual Zinc plus Arginine" (DZA) technology, was found to be effective at controlling bacteria using in vitro laboratory studies, translating to clinical efficacy to deliver plaque and gingivitis reduction benefits. Here, we used biophysical analyses and confocal laser scanning microscopy to understand how a DZA dentifrice impacted the mechanical properties of dental plaque biofilms and determine if changes to biofilm rheology enhanced the removal of dental plaque. Using both uniaxial mechanical indentation and an adapted rotating-disc rheometry assay, it was found that DZA treatment compromised biofilm mechanical integrity, resulting in the biofilm being more susceptible to removal by shear forces compared to treatment with either arginine or zinc alone. Confocal laser scanning microscopy revealed that DZA treatment reduced the amount of extracellular polymeric slime within the biofilm, likely accounting for the reduced mechanical properties. We propose a model where arginine facilitates the entry of zinc into the biofilm, resulting in additive effects of the two activities toward dental plaque biofilms. Together, our results support the use of a dentifrice containing Dual Zinc plus Arginine as part of daily oral hygiene regimens. IMPORTANCE Mechanical removal of dental plaque is augmented with therapeutic compounds to promote oral health. A dentifrice containing the ingredients zinc and arginine has shown efficacy at reducing dental plaque both in vitro and in vivo. However, how these active compounds interact together to facilitate dental plaque removal is unclear. Here, we used a combination of biophysical analyses and microscopy to demonstrate that combined treatment with zinc and arginine targets the matrix of dental plaque biofilms, which destabilized the mechanical integrity of these microbial communities, making them more susceptible to removal by shear forces.
Collapse
Affiliation(s)
- Erin S. Gloag
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, USA
| | - Yalda Khosravi
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, USA
| | - James G. Masters
- Colgate-Palmolive Technology Center, Piscataway, New Jersey, USA
| | - Daniel J. Wozniak
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, USA
- Department of Microbiology, The Ohio State University, Columbus, Ohio, USA
| | | | - Paul Stoodley
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, USA
- Department of Orthopaedics, The Ohio State University, Columbus, Ohio, USA
- National Biofilm Innovation Centre (NBIC), University of Southampton, Southampton, United Kingdom
- National Centre for Advanced Tribology at Southampton (nCATS), Mechanical Engineering, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
16
|
Baraniya D, Do T, Chen T, Albandar JM, Chialastri SM, Devine DA, Marsh PD, Al-Hebshi NN. Optimization of conditions for in vitro modeling of subgingival normobiosis and dysbiosis. Front Microbiol 2022; 13:1031029. [PMID: 36406462 PMCID: PMC9670125 DOI: 10.3389/fmicb.2022.1031029] [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: 08/29/2022] [Accepted: 10/14/2022] [Indexed: 11/06/2022] Open
Abstract
Modeling subgingival microbiome in health and disease is key to identifying the drivers of dysbiosis and to studying microbiome modulation. Here, we optimize growth conditions of our previously described in vitro subgingival microbiome model. Subgingival plaque samples from healthy and periodontitis subjects were used as inocula to grow normobiotic and dysbiotic microbiomes in MBEC assay plates. Saliva supplemented with 1%, 2%, 3.5%, or 5% (v/v) heat-inactivated human serum was used as a growth medium under shaking or non-shaking conditions. The microbiomes were harvested at 4, 7, 10 or 13 days of growth (384 microbiomes in total) and analyzed by 16S rRNA gene sequencing. Biomass significantly increased as a function of serum concentration and incubation period. Independent of growth conditions, the health- and periodontitis-derived microbiomes clustered separately with their respective inocula. Species richness/diversity slightly increased with time but was adversely affected by higher serum concentrations especially in the periodontitis-derived microbiomes. Microbial dysbiosis increased with time and serum concentration. Porphyromonas and Alloprevotella were substantially enriched in higher serum concentrations at the expense of Streptococcus, Fusobacterium and Prevotella. An increase in Porphyromonas, Bacteroides and Mogibacterium accompanied by a decrease in Prevotella, Catonella, and Gemella were the most prominent changes over time. Shaking had only minor effects. Overall, the health-derived microbiomes grown for 4 days in 1% serum, and periodontitis-derived microbiomes grown for 7 days in 3.5%-5% serum were the most similar to the respective inocula. In conclusion, normobiotic and dysbiostic subgingival microbiomes can be grown reproducibly in saliva supplemented with serum, but time and serum concentration need to be adjusted differently for the health and periodontitis-derived microbiomes to maximize similarity to in vivo inocula. The optimized model could be used to identify drivers of dysbiosis, and to evaluate interventions such as microbiome modulators.
Collapse
Affiliation(s)
- Divyashri Baraniya
- Oral Microbiome Research Laboratory, Maurice H. Kornberg School of Dentistry, Temple University, Philadelphia, PA, United States
| | - Thuy Do
- Division of Oral Biology, School of Dentistry, University of Leeds, Leeds, United Kingdom
| | - Tsute Chen
- Department of Microbiology, Forsyth Institute, Cambridge, MA, United States
| | - Jasim M. Albandar
- Department of Periodontology and Oral Implantology, Maurice H. Kornberg School of Dentistry, Temple University, Philadelphia, PA, United States
| | - Susan M. Chialastri
- Department of Periodontology and Oral Implantology, Maurice H. Kornberg School of Dentistry, Temple University, Philadelphia, PA, United States
| | - Deirdre A. Devine
- Division of Oral Biology, School of Dentistry, University of Leeds, Leeds, United Kingdom
| | - Philip D. Marsh
- Division of Oral Biology, School of Dentistry, University of Leeds, Leeds, United Kingdom
| | - Nezar N. Al-Hebshi
- Oral Microbiome Research Laboratory, Maurice H. Kornberg School of Dentistry, Temple University, Philadelphia, PA, United States,*Correspondence: Nezar N. Al-Hebshi,
| |
Collapse
|
17
|
Jayasinghe TN, Harrass S, Erdrich S, King S, Eberhard J. Protein Intake and Oral Health in Older Adults-A Narrative Review. Nutrients 2022; 14:4478. [PMID: 36364741 PMCID: PMC9653899 DOI: 10.3390/nu14214478] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/18/2022] [Accepted: 10/20/2022] [Indexed: 08/31/2023] Open
Abstract
Oral health is vital to general health and well-being for all ages, and as with other chronic conditions, oral health problems increase with age. There is a bi-directional link between nutrition and oral health, in that nutrition affects the health of oral tissues and saliva, and the health of the mouth may affect the foods consumed. Evidence suggests that a healthy diet generally has a positive impact on oral health in older adults. Although studies examining the direct link between oral health and protein intake in older adults are limited, some have explored the relationship via malnutrition, which is also prevalent among older adults. Protein-energy malnutrition (PEM) may be associated with poor oral health, dental caries, enamel hypoplasia, and salivary gland atrophy. This narrative review presents the theoretical evidence on the impact of dietary protein and amino acid composition on oral health, and their combined impact on overall health in older adults.
Collapse
Affiliation(s)
- Thilini N. Jayasinghe
- The Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia
- School of Dentistry, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - Sanaa Harrass
- School of Life Sciences, University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Sharon Erdrich
- School of Pharmacy, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - Shalinie King
- School of Dentistry, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - Joerg Eberhard
- The Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia
- School of Dentistry, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
18
|
Noori A, Hoseinpour M, Kolivand S, Lotfibakhshaiesh N, Azami M, Ai J, Ebrahimi-Barough S. Synergy effects of copper and L-arginine on osteogenic, angiogenic, and antibacterial activities. Tissue Cell 2022; 77:101849. [PMID: 35728334 DOI: 10.1016/j.tice.2022.101849] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 05/29/2022] [Accepted: 05/29/2022] [Indexed: 02/06/2023]
Abstract
Copper (Cu) ions have been found to exert antibacterial and angiogenic effects. However, some studies have indicated that it inhibits osteogenesis at high concentrations. On the other hand, L-arginine (Arg) is a semi-essential amino acid required for various biological processes, including osteogenic and angiogenic activities. As a result, we hypothesized that combining Arg with Cu ions would reduce its inhibitory effects on osteogenesis while increasing its angiogenic and antibacterial capabilities. To assess osteogenic and angiogenic activities, we employed rat bone marrow mesenchymal stem cells (MSCs) and human umbilical vein endothelial cells (HUVECs), respectively. The gram-positive bacteria Staphylococcus epidermidis (S. epidermidis), Staphylococcus aureus (S. aureus), and the gram-negative bacterium Escherichia coli (E. coli) were used to investigate bacterial behaviors. According to ALP activity and calcium deposition outcomes, copper ions inhibited osteogenic development of MSCs at 100 µM; however, Arg supplementation somewhat mitigated the inhibitory effects. Furthermore, Copper and Arg synergistically stimulated migration and tube formation of HUVECs. According to our findings, copper ions and Arg in the range of 1-100 µM had no antibacterial effect on any examined bacteria. However, at a dose of 20 mM, copper demonstrated antibacterial activity, which was boosted by Arg. Overall, these findings suggest that a combination of copper and Arg may be more beneficial for bone regeneration than either copper or Arg alone.
Collapse
Affiliation(s)
- Alireza Noori
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran; Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Mahdieh Hoseinpour
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sedighe Kolivand
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nasrin Lotfibakhshaiesh
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Mahmoud Azami
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Jafar Ai
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Somayeh Ebrahimi-Barough
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
19
|
Strategies to Combat Caries by Maintaining the Integrity of Biofilm and Homeostasis during the Rapid Phase of Supragingival Plaque Formation. Antibiotics (Basel) 2022; 11:antibiotics11070880. [PMID: 35884135 PMCID: PMC9312143 DOI: 10.3390/antibiotics11070880] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/24/2022] [Accepted: 06/28/2022] [Indexed: 02/06/2023] Open
Abstract
Bacteria in the oral cavity, including commensals and opportunistic pathogens, are organized into highly specialized sessile communities, coexisting in homeostasis with the host under healthy conditions. A dysbiotic environment during biofilm evolution, however, allows opportunistic pathogens to become the dominant species at caries-affected sites at the expense of health-associated taxa. Combining tooth brushing with dentifrices or rinses combat the onset of caries by partially removes plaque, but resulting in the biofilm remaining in an immature state with undesirables’ consequences on homeostasis and oral ecosystem. This leads to the need for therapeutic pathways that focus on preserving balance in the oral microbiota and applying strategies to combat caries by maintaining biofilm integrity and homeostasis during the rapid phase of supragingival plaque formation. Adhesion, nutrition, and communication are fundamental in this phase in which the bacteria that have survived these adverse conditions rebuild and reorganize the biofilm, and are considered targets for designing preventive strategies to guide the biofilm towards a composition compatible with health. The present review summarizes the most important advances and future prospects for therapies based on the maintenance of biofilm integrity and homeostasis as a preventive measure of dysbiosis focused on these three key factors during the rapid phase of plaque formation.
Collapse
|
20
|
Yao S, Hao L, Zhou R, Jin Y, Huang J, Wu C. Multispecies biofilms in fermentation: Biofilm formation, microbial interactions, and communication. Compr Rev Food Sci Food Saf 2022; 21:3346-3375. [PMID: 35762651 DOI: 10.1111/1541-4337.12991] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 05/07/2022] [Accepted: 05/15/2022] [Indexed: 02/05/2023]
Abstract
Food fermentation is driven by microorganisms, which usually coexist as multispecies biofilms. The activities and interactions of functional microorganisms and pathogenic bacteria in biofilms have important implications for the quality and safety of fermented foods. It was verified that the biofilm lifestyle benefited the fitness of microorganisms in harsh environments and intensified the cooperation and competition between biofilm members. This review focuses on multispecies biofilm formation, microbial interactions and communication in biofilms, and the application of multispecies biofilms in food fermentation. Microbial aggregation and adhesion are important steps in the early stage of multispecies biofilm formation. Different biofilm-forming abilities and strategies among microorganisms lead to several types of multispecies biofilm formation. The spatial distribution of multispecies biofilms reflects microbial interactions and biofilm function. Then, we discuss the intrinsic factors and external manifestations of multispecies biofilm system succession. Several typical interspecies cooperation and competition modes and mechanisms of microbial communication were reviewed in this review. The main limitations of the studies included in this review are the relatively small number of studies of biofilms formed by functional microorganisms during fermentation and the lack of direct evidence for the formation process of multispecies biofilms and microbial interactions and communication within biofilms. This review aims to provide the food industry with a sufficient understanding of multispecies biofilms in food fermentation. Practical Application: Meanwhile, it offers a reference value for better controlling and utilizing biofilms during food fermentation process, and the improvement of the yield, quality, and safety of fermented products including Chinese Baijiu, cheeese,kefir, soy sauce, kombucha, and fermented olive.
Collapse
Affiliation(s)
- Shangjie Yao
- College of Biomass Science and Engineering, Sichuan University, Chengdu, China.,Key Laboratory of Leather Chemistry and Engineering, Ministry of Education, Sichuan University, Chengdu, China
| | - Liying Hao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Rongqing Zhou
- College of Biomass Science and Engineering, Sichuan University, Chengdu, China.,Key Laboratory of Leather Chemistry and Engineering, Ministry of Education, Sichuan University, Chengdu, China
| | - Yao Jin
- College of Biomass Science and Engineering, Sichuan University, Chengdu, China.,Key Laboratory of Leather Chemistry and Engineering, Ministry of Education, Sichuan University, Chengdu, China
| | - Jun Huang
- College of Biomass Science and Engineering, Sichuan University, Chengdu, China.,Key Laboratory of Leather Chemistry and Engineering, Ministry of Education, Sichuan University, Chengdu, China
| | - Chongde Wu
- College of Biomass Science and Engineering, Sichuan University, Chengdu, China.,Key Laboratory of Leather Chemistry and Engineering, Ministry of Education, Sichuan University, Chengdu, China
| |
Collapse
|
21
|
Khosravi Y, Palmer S, Daep CA, Sambanthamoorthy K, Kumar P, Dusane D, Stoodley P. A commercial SnF 2 toothpaste formulation reduces simulated human plaque biofilm in a dynamic typodont model. J Appl Microbiol 2022; 133:1341-1352. [PMID: 35603698 PMCID: PMC9542754 DOI: 10.1111/jam.15634] [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: 02/16/2022] [Revised: 04/29/2022] [Accepted: 05/19/2022] [Indexed: 11/28/2022]
Abstract
AIMS We present a dynamic typodont biofilm model (DTBM) incorporating 1) human dentition anatomy, 2) fluid flow over intermittently fluid bathed tooth surfaces and 3) an oxic headspace to allow aerobic and anaerobic niches to develop naturally, as a screening tool to assess the effect of stannous fluoride (SnF2 ) toothpaste against a simulated human plaque biofilm (SPB). METHODS AND RESULTS First, hydroxyapatite (HA) coupons were inoculated with human saliva/plaque and cultured at 37o C under air. Selected species representative of common commensal and anaerobic pathogens were quantified for relative abundance changes over 4d by PCR densitometry to confirm the culture conditions allowed the proliferation of these species. A continuous culture DTBM reactor on a rocker table was inoculated with saliva/plaque and incubated at 37°C for 24h. Tooth shear stress was estimated by particle tracking. A SnF2 toothpaste solution, or a sham rise was administered twice daily for 3d to mimic routine oral hygiene. SPB biomass was assessed by total bacterial DNA and methylene blue (MB) staining. Early colonizer aerobes and late colonizer anaerobes species were detected in the HA and DTBM, and the trends in changing abundance were consistent with those seen clinically. CONCLUSIONS Treatment with the SnF2 solution showed significant reductions of 53.05% and 54.4% in the SPB by MB staining and DNA, respectively. SIGNIFICANCE AND IMPACT OF STUDY The model has potential for assessing dentition anatomy and fluid flow on the efficacy of antimicrobial efficacy against localized SPB and may be amenable to the plaque index clinical evaluation.
Collapse
Affiliation(s)
- Yalda Khosravi
- Department of Microbial Infection and Immunity, Ohio State University Columbus, OH, USA
| | - Sara Palmer
- College of Dentistry, The Ohio State University, Columbus, OH, USA
| | | | | | - Purnima Kumar
- College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - Devendra Dusane
- Department of Microbial Infection and Immunity, Ohio State University Columbus, OH, USA
| | - Paul Stoodley
- Department of Microbial Infection and Immunity, Ohio State University Columbus, OH, USA.,Dept. Orthopaedics, Ohio State University, Columbus, OH, USA.,National Centre for Advanced Tribology (nCATS) and National Biofilm Innovation Centre (NBIC), Mechanical Engineering, University of Southampton, UK
| |
Collapse
|
22
|
Bacali C, Vulturar R, Buduru S, Cozma A, Fodor A, Chiș A, Lucaciu O, Damian L, Moldovan ML. Oral Microbiome: Getting to Know and Befriend Neighbors, a Biological Approach. Biomedicines 2022; 10:671. [PMID: 35327473 PMCID: PMC8945538 DOI: 10.3390/biomedicines10030671] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/10/2022] [Accepted: 03/11/2022] [Indexed: 02/01/2023] Open
Abstract
The oral microbiome, forming a biofilm that covers the oral structures, contains a high number of microorganisms. Biofilm formation starts from the salivary pellicle that allows bacterial adhesion-colonization-proliferation, co-aggregation and biofilm maturation in a complex microbial community. There is a constant bidirectional crosstalk between human host and its oral microbiome. The paper presents the fundamentals regarding the oral microbiome and its relationship to modulator factors, oral and systemic health. The modern studies of oral microorganisms and relationships with the host benefits are based on genomics, transcriptomics, proteomics and metabolomics. Pharmaceuticals such as antimicrobials, prebiotics, probiotics, surface active or abrasive agents and plant-derived ingredients may influence the oral microbiome. Many studies found associations between oral dysbiosis and systemic disorders, including autoimmune diseases, cardiovascular, diabetes, cancers and neurodegenerative disorders. We outline the general and individual factors influencing the host-microbial balance and the possibility to use the analysis of the oral microbiome in prevention, diagnosis and treatment in personalized medicine. Future therapies should take in account the restoration of the normal symbiotic relation with the oral microbiome.
Collapse
Affiliation(s)
- Cecilia Bacali
- Department of Prosthodontics and Dental Materials, “Iuliu Hatieganu” University of Medicine and Pharmacy, 32 Clinicilor St., 400006 Cluj-Napoca, Romania; (C.B.); (S.B.)
| | - Romana Vulturar
- Department of Molecular Sciences, “Iuliu Hațieganu” University of Medicine and Pharmacy Cluj-Napoca, 6 Pasteur St., 400349 Cluj-Napoca, Romania;
- Cognitive Neuroscience Laboratory, University Babes-Bolyai, 30 Fântânele St., 400294 Cluj-Napoca, Romania
| | - Smaranda Buduru
- Department of Prosthodontics and Dental Materials, “Iuliu Hatieganu” University of Medicine and Pharmacy, 32 Clinicilor St., 400006 Cluj-Napoca, Romania; (C.B.); (S.B.)
| | - Angela Cozma
- 4th Medical Department, University of Medicine and Pharmacy “Iuliu Hatieganu” Cluj-Napoca, 18 Republicii St., 400015 Cluj-Napoca, Romania;
| | - Adriana Fodor
- Clinical Center of Diabetes, Nutrition and Metabolic Diseases, “Iuliu Hatieganu” University of Medicine and Pharmacy, 2-4 Clinicilor St., 400012 Cluj-Napoca, Romania;
| | - Adina Chiș
- Department of Molecular Sciences, “Iuliu Hațieganu” University of Medicine and Pharmacy Cluj-Napoca, 6 Pasteur St., 400349 Cluj-Napoca, Romania;
- Cognitive Neuroscience Laboratory, University Babes-Bolyai, 30 Fântânele St., 400294 Cluj-Napoca, Romania
| | - Ondine Lucaciu
- Department of Oral Health, University of Medicine and Pharmacy “Iuliu Hatieganu”, 400012 Cluj-Napoca, Romania;
| | - Laura Damian
- Department of Rheumatology, Emergency Clinical County Hospital Cluj, Centre for Rare Autoimmune and Autoinflammatory Diseases, 2-4 Clinicilor St., 400006 Cluj-Napoca, Romania;
- CMI Reumatologie Dr. Damian, 6-8 Petru Maior St., 400002 Cluj-Napoca, Romania
| | - Mirela Liliana Moldovan
- Department of Dermopharmacy and Cosmetics, Faculty of Pharmacy, “Iuliu Hatieganu” University of Medicine and Pharmacy, 12, I. Creanga St., 400010 Cluj-Napoca, Romania;
| |
Collapse
|
23
|
Metal Complexes—A Promising Approach to Target Biofilm Associated Infections. Molecules 2022; 27:molecules27030758. [PMID: 35164021 PMCID: PMC8838073 DOI: 10.3390/molecules27030758] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/19/2022] [Accepted: 01/23/2022] [Indexed: 02/06/2023] Open
Abstract
Microbial biofilms are represented by sessile microbial communities with modified gene expression and phenotype, adhered to a surface and embedded in a matrix of self-produced extracellular polymeric substances (EPS). Microbial biofilms can develop on both prosthetic devices and tissues, generating chronic and persistent infections that cannot be eradicated with classical organic-based antimicrobials, because of their increased tolerance to antimicrobials and the host immune system. Several complexes based mostly on 3D ions have shown promising potential for fighting biofilm-associated infections, due to their large spectrum antimicrobial and anti-biofilm activity. The literature usually reports species containing Mn(II), Ni(II), Co(II), Cu(II) or Zn(II) and a large variety of multidentate ligands with chelating properties such as antibiotics, Schiff bases, biguanides, N-based macrocyclic and fused rings derivatives. This review presents the progress in the development of such species and their anti-biofilm activity, as well as the contribution of biomaterials science to incorporate these complexes in composite platforms for reducing the negative impact of medical biofilms.
Collapse
|
24
|
Paluch E, Okińczyc P, Zwyrzykowska-Wodzińska A, Szperlik J, Żarowska B, Duda-Madej A, Bąbelewski P, Włodarczyk M, Wojtasik W, Kupczyński R, Szumny A. Composition and Antimicrobial Activity of Ilex Leaves Water Extracts. Molecules 2021; 26:molecules26247442. [PMID: 34946528 PMCID: PMC8707412 DOI: 10.3390/molecules26247442] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 12/02/2021] [Accepted: 12/05/2021] [Indexed: 11/16/2022] Open
Abstract
Plants from the Ilex genus are known for properties such as antimicrobial and anti-inflammatory activity, can act as antiobesity agents and thus can be helpful in medicine. Some holly species, such as Ilex paraguariensis (widely known in the form of popular beverage: yerba mate), have been investigated, while others have been partially researched or remain unknown. Therefore, we performed qualitative and quantitative phytochemical analyses and screened antimicrobial properties of lesser-studied species (I. aquifolium L., I. aquifolium ‘Argentea Marginata’ and I. × meserveae ‘Blue Angel’). I. paraguariensis was used as a standard species for comparison purposes. Investigations were performed on water extracts due to their expected activity and composition. Antimicrobial research included evaluating minimal inhibitory, bactericidal (Staphylococcus aureus and Escherichia coli) and fungicidal concentration (Candida albicans, Alternaria alternata, Fusarium oxysporum, and Aspergillus niger) of extracts. The influence of the extracts on the production, eradication, and viability of bacterial biofilms was also analysed. It was established that Ilex paraguariensis possesses the richest profile of hydroxycinnamic acids derivatives in terms of component concentration and diversity. Ilex spp., especially I. × meserveae, contain a slightly higher amount of flavonoids and more different flavonoid derivatives than I. paraguariensis. However, the strongest antibacterial activity was shown by I. aquifolium L. and its cultivar ‘Argentea Marginata’ in terms of minimal inhibitory, bactericidal and fungicidal concentration, and biofilm assays. Extracts from both species significantly reduced the biofilm viability of S. aureus as well, which may be of use in the production of multicomponent lavaseptics, antiseptics, diuretics (supporting urinary tract infection therapy) and, due to their action on fungi, additives to growth media for specific fungi. The significant content of saponins enables Ilex extracts to be used as natural emulsifiers, for example, in cosmetics. Moreover, relatively high chlorogenic acid and rutin content may suggest use of Ilex spp. to treat obesity, digestive problems, in chemoprevention, and as preservatives in the food industry.
Collapse
Affiliation(s)
- Emil Paluch
- Department of Microbiology, Faculty of Medicine, Wroclaw Medical University, Chałubińskiego 4, 50-376 Wrocław, Poland; (E.P.); (A.D.-M.)
| | - Piotr Okińczyc
- Department of Pharmacognosy and Herbal Medicines, Wroclaw Medical University, Borowska 211a, 50-556 Wrocław, Poland;
- Correspondence: (P.O.); (P.B.); Tel.: +48-71-784-02-16 (P.O.)
| | - Anna Zwyrzykowska-Wodzińska
- Department of Environment Hygiene and Animal Welfare, Wrocław University of Environmental and Life Sciences, Chełmońskiego 38C, 51-630 Wrocław, Poland; (A.Z.-W.); (R.K.)
| | - Jakub Szperlik
- Laboratory of Tissue Culture, Botanical Garden, Faculty of Biological Sciences, University of Wrocław, Sienkiewicza 23, 50-525 Wrocław, Poland;
| | - Barbara Żarowska
- Department of Biotechnology and Food Microbiology, Wrocław University of Environmental and Life Sciences, Chełmońskiego 37, 51-630 Wrocław, Poland;
| | - Anna Duda-Madej
- Department of Microbiology, Faculty of Medicine, Wroclaw Medical University, Chałubińskiego 4, 50-376 Wrocław, Poland; (E.P.); (A.D.-M.)
| | - Przemysław Bąbelewski
- Department of Horticulture, Wrocław University of Environmental and Life Sciences, Grunwaldzki 24A, 50-375 Wrocław, Poland
- Correspondence: (P.O.); (P.B.); Tel.: +48-71-784-02-16 (P.O.)
| | - Maciej Włodarczyk
- Department of Pharmacognosy and Herbal Medicines, Wroclaw Medical University, Borowska 211a, 50-556 Wrocław, Poland;
| | - Wioleta Wojtasik
- Faculty of Biotechnology, University of Wrocław, Przybyszewskiego 63/77, 51-148 Wrocław, Poland;
| | - Robert Kupczyński
- Department of Environment Hygiene and Animal Welfare, Wrocław University of Environmental and Life Sciences, Chełmońskiego 38C, 51-630 Wrocław, Poland; (A.Z.-W.); (R.K.)
| | - Antoni Szumny
- Department of Chemistry, Wrocław University of Environmental and Life Sciences, Norwida 25, 50-375 Wrocław, Poland;
| |
Collapse
|
25
|
Gloag ES, Wozniak DJ, Wolf KL, Masters JG, Daep CA, Stoodley P. Arginine Induced Streptococcus gordonii Biofilm Detachment Using a Novel Rotating-Disc Rheometry Method. Front Cell Infect Microbiol 2021; 11:784388. [PMID: 34805002 PMCID: PMC8602906 DOI: 10.3389/fcimb.2021.784388] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 10/21/2021] [Indexed: 11/13/2022] Open
Abstract
Oral diseases are one of the most common pathologies affecting human health. These diseases are typically associated with dental plaque-biofilms, through either build-up of the biofilm or dysbiosis of the microbial community. Arginine can disrupt dental plaque-biofilms, and maintain plaque homeostasis, making it an ideal therapeutic to combat the development of oral disease. Despite our understanding of the actions of arginine towards dental plaque-biofilms, it is still unclear how or if arginine effects the mechanical integrity of the dental plaque-biofilm. Here we adapted a rotating-disc rheometry assay, a method used to quantify marine biofilm fouling, to study how arginine treatment of Streptococcus gordonii biofilms influences biofilm detachment from surfaces. We demonstrate that the assay is highly sensitive at quantifying the presence of biofilm and the detachment or rearrangement of the biofilm structure as a function of shear stress. We demonstrate that arginine treatment leads to earlier detachment of the biofilm, indicating that arginine treatment weakens the biofilm, making it more susceptible to removal by shear stresses. Finally, we demonstrate that the biofilm disrupting affect is specific to arginine, and not a general property of amino acids, as S. gordonii biofilms treated with either glycine or lysine had mechanical properties similar to untreated biofilms. Our results add to the understanding that arginine targets biofilms by multifaceted mechanisms, both metabolic and physical, further promoting the potential of arginine as an active compound in dentifrices to maintain oral health.
Collapse
Affiliation(s)
- Erin S Gloag
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States
| | - Daniel J Wozniak
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States.,Department of Microbiology, The Ohio State University, Columbus, OH, United States
| | - Kevin L Wolf
- Department of Mechanical and Aerospace Engineering, The Ohio State University, Columbus, OH, United States
| | - James G Masters
- Colgate-Palmolive Technology Center, Piscataway, NJ, United States
| | | | - Paul Stoodley
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, United States.,Department of Orthopedics, The Ohio State University, Columbus, OH, United States.,National Biofilm Innovation Centre (NBIC) and National Centre for Advanced Tribology at Southampton (nCATS), University of Southampton, Southampton, United Kingdom
| |
Collapse
|
26
|
Bijle MN, Abdalla MM, Ashraf U, Siu KW, Tsoi J, Yiu CKY. The acid-resistance potential of arginine-fluoride varnish treated enamel. J Mech Behav Biomed Mater 2021; 125:104763. [PMID: 34781227 DOI: 10.1016/j.jmbbm.2021.104763] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 08/05/2021] [Accepted: 08/05/2021] [Indexed: 11/24/2022]
Abstract
The study objective was to examine the acid-resistance potential of enamel carious lesions treated with arginine (Arg)-sodium fluoride (NaF) varnishes using nano-mechanical testing and chemical mapping. L-arginine (at 1%, 2%, & 4%) was incorporated in 5% NaF varnish. The experimental/control groups were: 1% Arg-NaF, 2% Arg-NaF, 4% Arg-NaF, NaF, and no treatment. Enamel specimen blocks were subjected to incipient carious lesion formation. After treatment, the specimens underwent chemical pH-cycling for 8-days and acid challenge for 2 h. The specimens were characterised for surface nano-hardness (SNH) and calcium/phosphate content of the treated lesions to determine enamel solubility reduction (ESR). Post-acid challenge, X-ray diffraction crystallography (XRD), and energy dispersive X-ray spectrophotometry (EDX) were performed. The SNH for 2%/4% Arg-NaF demonstrated a higher resistance to acid challenge with significantly higher SNH recovery than NaF varnish (p<0.05). The ESR potential of 2%/4% Arg-NaF varnish was significantly higher than NaF varnish (p<0.05). The XRD crystalline phases demonstrated that 2%/4% Arg-NaF had intense hydroxyapatite peaks discerning its increased potential to resist demineralization than NaF varnish. The EDX results showed that 2%/4% Arg-NaF demonstrated Ca/P ratio closer to hydroxyapatite (~1.67) post-acid challenge. Incorporating 2%/4% L-arginine in a 5% NaF varnish enhances the acid-resistance potential of NaF varnish.
Collapse
Affiliation(s)
| | - Mohamed Mahmoud Abdalla
- Paediatric Dentistry, Faculty of Dentistry, The University of Hong Kong, Hong Kong; Dental Biomaterials, Faculty of Dental Medicine Al-Azhar University, Cairo, Egypt.
| | - Usman Ashraf
- Paediatric Dentistry, Faculty of Dentistry, The University of Hong Kong, Hong Kong.
| | - Kai Wing Siu
- Mechanical Engineering, Faculty of Engineering, The University of Hong Kong, Hong Kong.
| | - James Tsoi
- Dental Materials Science, Faculty of Dentistry, The University of Hong Kong, Hong Kong.
| | - Cynthia Kar Yung Yiu
- Paediatric Dentistry, Faculty of Dentistry, The University of Hong Kong, Hong Kong.
| |
Collapse
|
27
|
Salivary microbiome profiling reveals a dysbiotic schizophrenia-associated microbiota. NPJ SCHIZOPHRENIA 2021; 7:51. [PMID: 34711862 PMCID: PMC8553823 DOI: 10.1038/s41537-021-00180-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 10/01/2021] [Indexed: 01/01/2023]
Abstract
Schizophrenia is a debilitating mental disorder and often has a prodromal period, referred to as clinical high risk (CHR) for psychosis, prior to the first episode. The etiology and pathogenesis of schizophrenia remain unclear. Despite the human gut microbiome being associated with schizophrenia, the role of the oral microbiome, which is a vital player in the mouth-body connection, is not well understood. To address this, we performed 16S rRNA gene sequencing to investigate the salivary microbiome in 85 patients with drug-naïve first-episode schizophrenia (FES), 43 individuals at CHR, and 80 healthy controls (HCs). The salivary microbiome of FES patients was characterized by higher α-diversity and lower β-diversity heterogeneity than those of CHR subjects and HCs. Proteobacteria, the predominant phylum, was depleted, while Firmicutes and the Firmicutes/Proteobacteria ratio was enriched, in a stepwise manner from HC to CHR to FES. H2S-producing bacteria exhibited disease-stage-specific enrichment and could be potential diagnostic biomarkers for FES and CHR. Certain salivary microbiota exhibited disease-specific correlation patterns with symptomatic severities, peripheral pro-inflammatory cytokines, thioredoxin, and S100B in FES. Furthermore, the metabolic functions from inferred metagenomes of the salivary microbiome were disrupted in FES, especially amino acid metabolism, carbohydrate metabolism, and xenobiotic degradation. This study has established a link between salivary microbiome alterations and disease initiation and provided the hypothesis of how the oral microbiota could influence schizophrenia.
Collapse
|
28
|
Ugya AY, Ari HA, Hua X. Microalgae biofilm formation and antioxidant responses to stress induce by Lemna minor L., Chlorella vulgaris, and Aphanizomenon flos-aquae. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 221:112468. [PMID: 34198191 DOI: 10.1016/j.ecoenv.2021.112468] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 06/23/2021] [Accepted: 06/24/2021] [Indexed: 06/13/2023]
Abstract
The study shows how microalgae biofilm formation and antioxidant responses to the production of reactive oxygen species (ROS) is alter by the presences of Lemna minor L., Chlorella vulgaris, and Aphanizomenon flos-aquae. The study involves the cultivation of the biofilm of Chlorella vulgaris and Aphanizomenon flos-aquae in three bioreactors. The condition of growth for the biofilm formation was varied across the three bioreactors to enable the dominance Chlorella vulgaris and Aphanizomenon flos-aquae in one of the bioreactors. Lemna minor L. was also introduce into one of the bioreactors to determine its effect on the biofilm formation. The result obtained shows that C. vulgaris and A. flos-aquae dominate the biofilm, resulting in a high level of H2O2 and O2- (H2O2 was 0.122 ± 0.052 and 0.183 ± 0.108 mmol/L in C. vulgaris and A. flos-aquae, respectively, and O2- was 0.261 ± 0.039 and 0.251 ± 0.148 mmol/L in C. vulgaris and A. flos-aquae, respectively). The study also revealed that the presence of L. minor L. tend to reduce the oxidative stress to the biofilm leading to low production of ROS (H2O2 was 0.086 ± 0.027 and 0.089 ± 0.045 mmol/L in C. vulgaris and A. flos-aquae respectively, and O2- was 0.185 ± 0.044 and 0.161 ± 0.065 mmol/L in C. vulgaris and A. flos-aquae respectively). The variation in the ability of the biofilm of C. vulgaris and A. flos-aquae to respond via chlorophyll, carotenoid, flavonoid, anthocyanin, superoxide dismutase, peroxidase, catalase, glutathione reductase activities, antioxidant reducing power, phosphomolybdate activity, DPPH reduction activity, H2O2 scavenging activity, lipid content and organic carbon also supports the fact that the presence of biomass of microalgae and aquatic macrophytes tend to affect the process of microalgae biofilm formation and the ability of the biofilm to produce antioxidant. This high nutrient utilization leads to the production of biomass which can be used for biofuel production and other biotechnological products.
Collapse
Affiliation(s)
- Adamu Yunusa Ugya
- Key Lab of Groundwater Resources and Environment of Ministry of Education, Key Lab of Water Resources and Aquatic Environment of Jilin Province, College of New Energy and Environment, Jilin University, Changchun 130012, China; Department of Environmental Management, Kaduna State University, Kaduna, Nigeria
| | - Hadiza Abdullahi Ari
- Key Lab of Groundwater Resources and Environment of Ministry of Education, Key Lab of Water Resources and Aquatic Environment of Jilin Province, College of New Energy and Environment, Jilin University, Changchun 130012, China; Faculty of Sciences, National Open University of Nigeria, Lagos, Nigeria
| | - Xiuyi Hua
- Key Lab of Groundwater Resources and Environment of Ministry of Education, Key Lab of Water Resources and Aquatic Environment of Jilin Province, College of New Energy and Environment, Jilin University, Changchun 130012, China.
| |
Collapse
|
29
|
Lazar V, Holban AM, Curutiu C, Chifiriuc MC. Modulation of Quorum Sensing and Biofilms in Less Investigated Gram-Negative ESKAPE Pathogens. Front Microbiol 2021; 12:676510. [PMID: 34394026 PMCID: PMC8359898 DOI: 10.3389/fmicb.2021.676510] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 06/30/2021] [Indexed: 01/20/2023] Open
Abstract
Pathogenic bacteria have the ability to sense their versatile environment and adapt by behavioral changes both to the external reservoirs and the infected host, which, in response to microbial colonization, mobilizes equally sophisticated anti-infectious strategies. One of the most important adaptive processes is the ability of pathogenic bacteria to turn from the free, floating, or planktonic state to the adherent one and to develop biofilms on alive and inert substrata; this social lifestyle, based on very complex communication networks, namely, the quorum sensing (QS) and response system, confers them an increased phenotypic or behavioral resistance to different stress factors, including host defense mechanisms and antibiotics. As a consequence, biofilm infections can be difficult to diagnose and treat, requiring complex multidrug therapeutic regimens, which often fail to resolve the infection. One of the most promising avenues for discovering novel and efficient antibiofilm strategies is targeting individual cells and their QS mechanisms. A huge amount of data related to the inhibition of QS and biofilm formation in pathogenic bacteria have been obtained using the well-established gram-positive Staphylococcus aureus and gram-negative Pseudomonas aeruginosa models. The purpose of this paper was to revise the progress on the development of antibiofilm and anti-QS strategies in the less investigated gram-negative ESKAPE pathogens Klebsiella pneumoniae, Acinetobacter baumannii, and Enterobacter sp. and identify promising leads for the therapeutic management of these clinically significant and highly resistant opportunistic pathogens.
Collapse
Affiliation(s)
- Veronica Lazar
- Department of Microbiology and Immunology, Faculty of Biology, University of Bucharest, Bucharest, Romania
- The Research Institute of the University of Bucharest, Bucharest, Romania
| | - Alina Maria Holban
- Department of Microbiology and Immunology, Faculty of Biology, University of Bucharest, Bucharest, Romania
- The Research Institute of the University of Bucharest, Bucharest, Romania
| | - Carmen Curutiu
- Department of Microbiology and Immunology, Faculty of Biology, University of Bucharest, Bucharest, Romania
- The Research Institute of the University of Bucharest, Bucharest, Romania
| | - Mariana Carmen Chifiriuc
- Department of Microbiology and Immunology, Faculty of Biology, University of Bucharest, Bucharest, Romania
- The Research Institute of the University of Bucharest, Bucharest, Romania
| |
Collapse
|
30
|
Li S, Wang C, Wu Z. Dietary L-arginine supplementation of tilapia (Oreochromis niloticus) alters the microbial population and activates intestinal fatty acid oxidation. Amino Acids 2021; 54:339-351. [PMID: 34212252 DOI: 10.1007/s00726-021-03018-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Accepted: 06/08/2021] [Indexed: 12/15/2022]
Abstract
Currently, little is known about the function of L-arginine in the homeostasis of intestinal lipid metabolism. This study was conducted to test the hypothesis that dietary L-arginine supplementation may alter intestinal microbiota and lipid metabolism in tilapia. Tilapia were fed a basal diet (containing 16.9 g L-arginine per kilogram diets) or the basal diet supplemented with 1% or 2% L-arginine for 8 wks. In the present study, we found that dietary supplementation with 1% or 2% L-arginine induced a shift in the community structure of gut microbiota, as showed by increased (p < 0.05) α-diversity, altered (p < 0.05) β-diversity and function profile. This finding coincided with decreased lipid accretion in the intestine of tilapia, which was associated with an enhancement in mRNA levels for peroxisome proliferator-activated receptor α (Pparα), acyl-coenzyme a oxidase 1 (Acox1), and peroxisome proliferator-activated receptor γ coactivator-1α (Pgc-1α). Using intestinal epithelial cell culture, we demonstrated that the lipid-lowering effect of L-arginine was mainly mediated by activating the AMP-activated protein kinase (AMPK) signaling pathway, carnitine palmitoyltransferase 1 (CPT1), and PPARα, as well as mRNA levels for Acox1 and Acox2. Collectively, our results suggest that dietary L-arginine supplementation of tilapia changed the intestinal microbiota and activated intestinal fatty acid oxidation. However, future studies are warranted to determine the relationship between microbiota and lipid metabolism in the intestine.
Collapse
Affiliation(s)
- Senlin Li
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing, 100193, China
| | - Chao Wang
- College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Zhenlong Wu
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition and Feed Science, China Agricultural University, Beijing, 100193, China. .,Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
31
|
Muras A, Mallo N, Otero-Casal P, Pose-Rodríguez JM, Otero A. Quorum sensing systems as a new target to prevent biofilm-related oral diseases. Oral Dis 2020; 28:307-313. [PMID: 33080080 DOI: 10.1111/odi.13689] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 09/23/2020] [Accepted: 10/11/2020] [Indexed: 12/13/2022]
Abstract
OBJECTIVE The present study summarizes the current knowledge on the role of bacterial extracellular signaling systems, known as quorum sensing (QS), in oral biofilm formation, and on the possibility of blocking these microbial communication systems as a potential approach to prevent and treat oral infectious diseases. METHODS A detailed literature review of the current knowledge of QS in the oral cavity was performed, using the databases MEDLINE (through PubMed) and Web of Science. RESULTS Accumulating direct and indirect evidence indicates an important role of QS molecules in the oral microbial ecosystem. CONCLUSIONS The mechanisms regulating gene expression through bacterial communication systems constitute a promising target to control oral biofilm formation. Although cell-to-cell communication is pivotal for biofilm formation of many pathogenic bacteria, knowledge concerning microbial interactions and signaling processes within multispecies biofilms in the oral cavity is still limited.
Collapse
Affiliation(s)
- Andrea Muras
- Department of Microbiology and Parasitology, Faculty of Biology-CIBUS, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Natalia Mallo
- Department of Microbiology and Parasitology, Faculty of Biology-CIBUS, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Paz Otero-Casal
- Department of Surgery and Medical-Surgical Specialty, Faculty of Medicine and Dentistry, Universidade de Santiago de Compostela, Santiago de Compostela, Spain.,Unit of Oral Health, C.S. Santa Comba-Negreira, SERGAS, Santa Comba, Spain
| | - José M Pose-Rodríguez
- Department of Surgery and Medical-Surgical Specialty, Faculty of Medicine and Dentistry, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Ana Otero
- Department of Microbiology and Parasitology, Faculty of Biology-CIBUS, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| |
Collapse
|
32
|
Zeng L, Burne RA. Subpopulation behaviors in lactose metabolism by Streptococcus mutans. Mol Microbiol 2020; 115:58-69. [PMID: 32881164 DOI: 10.1111/mmi.14596] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 08/17/2020] [Accepted: 08/24/2020] [Indexed: 01/08/2023]
Abstract
When Streptococcus mutans is transferred from a preferred carbohydrate (glucose or fructose) to lactose, initiation of growth can take several hours, and substantial amounts of glucose are released during growth. Here, S. mutans strains UA159 and GS-5 were examined for stochastic behaviors in transcription of the lac operon. Using a gfp reporter fusion, we demonstrated that induction of the lac operon occurs in only a fraction of the population, with prior exposure to carbohydrate source and strain influencing the magniture of the sub-population response. Lower glucokinase activity in GS-5 was associated with release of substantially more glucose than UA159 and significantly lower lac expression. Mutants unable to use lactose grew on lactose as the sole carbohydrate when strains with an intact lac operon were also present in the cultures, indicative of the potential for population cheating. Utilizing a set of engineered obligate cheating and non-cheating strains, we confirmed that cheating can sustain a heterogeneous population. Futher, obligate cheaters of GS-5 competed well with the non-cheaters and showed a high degree of competitive fitness in a human-derived consortium biofilm model. The results show that bet-hedging behaviors in carbohydrate metabolism may substantially influence the composition and pathogenic potential of oral biofilms.
Collapse
Affiliation(s)
- Lin Zeng
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, FL, USA
| | - Robert A Burne
- Department of Oral Biology, University of Florida College of Dentistry, Gainesville, FL, USA
| |
Collapse
|
33
|
Bijle MN, Ekambaram M, Yiu CKY. A Scoping Review on Arginine in Caries Prevention. J Evid Based Dent Pract 2020; 20:101470. [PMID: 32921383 DOI: 10.1016/j.jebdp.2020.101470] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 05/04/2020] [Accepted: 06/20/2020] [Indexed: 10/23/2022]
Abstract
OBJECTIVES Emerging science on arginine or arginine formulations has driven the need to examine the research in the field. The scoping study objectives were (1) to identify the extent, range, and type of evidence on the role of arginine or arginine formulations in caries prevention and (2) to explore the future scope of research on arginine-containing caries-preventive agents. METHODS A systematic search was performed in PubMed, Scopus, and Web of Science. In vitro studies, clinical trials, narrative reviews, systematic reviews and or meta-analysis, and umbrella reviews or meta-evaluation examining arginine or arginine formulations for caries prevention were included. The data-charting process involved extracting variables followed by evidence synthesis. Arginine variants investigated up to date were discussed to explore future scope of research. RESULTS Thirty-nine articles were included for review from 105 identified citations comprising of in vitro studies, clinical trials, and reviews. Most articles studied 1.5% arginine-fluoride toothpaste. Most studies were from Asia, followed by North America, with fewest studies from Europe and South America. Arginine or arginine formulations demonstrated a superior caries-preventive effect compared with their matched controls (including fluorides); however, the evidence is with high risk of bias. Until now, three arginine variants have been investigated with l-arginine monohydrochloride as the least explored variant. CONCLUSIONS The evidence on the caries-preventive effect of arginine or arginine formulations has a high risk of bias. High-quality clinical trials are needed to assess the caries-preventive potential of arginine in commercial formulations. The role of l-arginine monohydrochloride in caries prevention can further be explored by incorporating in self-applied and professionally applied caries-preventive agents.
Collapse
Affiliation(s)
- Mohammed Nadeem Bijle
- PhD Candidate, Paediatric Dentistry, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China
| | - Manikandan Ekambaram
- Senior Lecturer, Paediatric Dentistry, Faculty of Dentistry, University of Otago, Dunedin, New Zealand
| | - Cynthia Kar Yung Yiu
- Clinical Professor, Paediatric Dentistry, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
34
|
Raju SC, Viljakainen H, Figueiredo RAO, Neuvonen PJ, Eriksson JG, Weiderpass E, Rounge TB. Antimicrobial drug use in the first decade of life influences saliva microbiota diversity and composition. MICROBIOME 2020; 8:121. [PMID: 32825849 PMCID: PMC7441731 DOI: 10.1186/s40168-020-00893-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 07/13/2020] [Indexed: 05/06/2023]
Abstract
BACKGROUND The human microbiota contributes to health and well-being. Antimicrobials (AM) have an immediate effect on microbial diversity and composition in the gut, but next to nothing is known about their long-term contribution to saliva microbiota. Our objectives were to investigate the long-term impact of AM use on saliva microbiota diversity and composition in preadolescents. We compared the lifetime effects by gender and AMs. We used data from 808 randomly selected children in the Finnish Health In Teens (Fin-HIT) cohort with register-based data on AM purchases from the Social Insurance Institution of Finland. Saliva microbiota was assessed with 16S rRNA (V3-V4) sequencing. The sequences were aligned to the SILVA ribosomal RNA database and classified and counted using the mothur pipeline. Associations between AM use and alpha-diversity (Shannon index) were identified with linear regression, while associations between beta-diversity (Bray-Curtis dissimilarity) and low, medium or high AM use were identified with PERMANOVA. RESULTS Of the children, 53.6% were girls and their mean age was 11.7 (0.4) years. On average, the children had 7.4 (ranging from 0 to 41) AM prescriptions during their lifespan. The four most commonly used AMs were amoxicillin (n = 2622, 43.7%), azithromycin (n = 1495, 24.9%), amoxicillin-clavulanate (n = 1123, 18.7%) and phenoxymethylpenicillin (n = 408, 6.8%). A linear inverse association was observed between the use of azithromycin and Shannon index (b - 0.015, p value = 0.002) in all children, the effect was driven by girls (b - 0.032, p value = 0.001), while not present in boys. Dissimilarities were marked between high, medium and low users of all AMs combined, in azithromycin users specifically, and in boys with amoxicillin use. Amoxicillin and amoxicillin-clavulanate use was associated with the largest decrease in abundance of Rikenellaceae. AM use in general and phenoxymethylpenicillin specifically were associated with a decrease of Paludibacter and pathways related to amino acid degradations differed in proportion between high and low AM users. CONCLUSIONS A systematic approach utilising reliable registry data on lifetime use of AMs demonstrated long-term effects on saliva microbiota diversity and composition. These effects are gender- and AM-dependent. We found that frequent lifelong use of AMs shifts bacterial profiles years later, which might have unforeseen health impacts in the future. Our findings emphasise a concern for high azithromycin use, which substantially decreases bacterial diversity and affects composition as well. Further studies are needed to determine the clinical implications of our findings. Video Abstract.
Collapse
Affiliation(s)
- Sajan C Raju
- Folkhälsan Research Center, Topeliuksenkatu 20, 00250, Helsinki, Finland
- Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Heli Viljakainen
- Folkhälsan Research Center, Topeliuksenkatu 20, 00250, Helsinki, Finland
- Department of Food and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Rejane A O Figueiredo
- Folkhälsan Research Center, Topeliuksenkatu 20, 00250, Helsinki, Finland
- Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Pertti J Neuvonen
- Department of Clinical Pharmacology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Johan G Eriksson
- Folkhälsan Research Center, Topeliuksenkatu 20, 00250, Helsinki, Finland
- Department of General Practice and Primary Health Care, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Elisabete Weiderpass
- International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Trine B Rounge
- Folkhälsan Research Center, Topeliuksenkatu 20, 00250, Helsinki, Finland.
- Faculty of Medicine, University of Helsinki, Helsinki, Finland.
- Department of Research, Cancer Registry of Norway, Oslo, Norway.
- Department of Informatics, University of Oslo, Oslo, Norway.
| |
Collapse
|
35
|
Jiang Y, Geng M, Bai L. Targeting Biofilms Therapy: Current Research Strategies and Development Hurdles. Microorganisms 2020; 8:microorganisms8081222. [PMID: 32796745 PMCID: PMC7465149 DOI: 10.3390/microorganisms8081222] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 07/31/2020] [Accepted: 08/07/2020] [Indexed: 01/05/2023] Open
Abstract
Biofilms are aggregate of microorganisms in which cells are frequently embedded within a self-produced matrix of extracellular polymeric substance (EPS) and adhere to each other and/or to a surface. The development of biofilm affords pathogens significantly increased tolerances to antibiotics and antimicrobials. Up to 80% of human bacterial infections are biofilm-associated. Dispersal of biofilms can turn microbial cells into their more vulnerable planktonic phenotype and improve the therapeutic effect of antimicrobials. In this review, we focus on multiple therapeutic strategies that are currently being developed to target important structural and functional characteristics and drug resistance mechanisms of biofilms. We thoroughly discuss the current biofilm targeting strategies from four major aspects—targeting EPS, dispersal molecules, targeting quorum sensing, and targeting dormant cells. We explain each aspect with examples and discuss the main hurdles in the development of biofilm dispersal agents in order to provide a rationale for multi-targeted therapy strategies that target the complicated biofilms. Biofilm dispersal is a promising research direction to treat biofilm-associated infections in the future, and more in vivo experiments should be performed to ensure the efficacy of these therapeutic agents before being used in clinic.
Collapse
|
36
|
Dynamic mucus penetrating microspheres for efficient pulmonary delivery and enhanced efficacy of host defence peptide (HDP) in experimental tuberculosis. J Control Release 2020; 324:17-33. [DOI: 10.1016/j.jconrel.2020.05.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 04/22/2020] [Accepted: 05/06/2020] [Indexed: 12/14/2022]
|
37
|
Miranda ML, Silva BNS, Salomão KB, de Oliveira AB, Gabbai-Armelin PR, Brighenti FL. Effect of arginine on microorganisms involved in dental caries: a systematic literature review of in vitro studies. BIOFOULING 2020; 36:696-709. [PMID: 32752889 DOI: 10.1080/08927014.2020.1802587] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 07/24/2020] [Accepted: 07/25/2020] [Indexed: 06/11/2023]
Abstract
This systematic review aimed to discuss the effects of arginine on caries-related microorganisms in different in vitro biofilm models. The eligibility criteria were in vitro studies that evaluated the effect of arginine at different concentrations on caries-related microorganisms using biofilm models. Eighteen studies published between 2012 and 2019 were included. Different bacterial species were studied. Seventeen studies (94.4%) achieved a low risk of bias and only one showed a medium risk of bias. The studies showed that arginine is a promising approach for the ecological management of dental caries. The focus of this review was to evaluate the effects of arginine on microorganisms involved in the mechanism of dental caries.
Collapse
Affiliation(s)
- Marina Lins Miranda
- Department of Morphology, Genetics, Orthodontics and Pediatric Dentistry, Araraquara Dental School, São Paulo State University (UNESP), Araraquara, Brazil
| | - Bianca Núbia Souza Silva
- Department of Morphology, Genetics, Orthodontics and Pediatric Dentistry, Araraquara Dental School, São Paulo State University (UNESP), Araraquara, Brazil
| | - Karina Borges Salomão
- Department of Morphology, Genetics, Orthodontics and Pediatric Dentistry, Araraquara Dental School, São Paulo State University (UNESP), Araraquara, Brazil
| | - Analú Barros de Oliveira
- Department of Morphology, Genetics, Orthodontics and Pediatric Dentistry, Araraquara Dental School, São Paulo State University (UNESP), Araraquara, Brazil
| | - Paulo Roberto Gabbai-Armelin
- Department of Morphology, Genetics, Orthodontics and Pediatric Dentistry, Araraquara Dental School, São Paulo State University (UNESP), Araraquara, Brazil
| | - Fernanda Lourenção Brighenti
- Department of Morphology, Genetics, Orthodontics and Pediatric Dentistry, Araraquara Dental School, São Paulo State University (UNESP), Araraquara, Brazil
| |
Collapse
|
38
|
Muras A, Otero-Casal P, Blanc V, Otero A. Acyl homoserine lactone-mediated quorum sensing in the oral cavity: a paradigm revisited. Sci Rep 2020; 10:9800. [PMID: 32555242 PMCID: PMC7300016 DOI: 10.1038/s41598-020-66704-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 05/27/2020] [Indexed: 01/08/2023] Open
Abstract
Acyl homoserine lactones (AHLs), the quorum sensing (QS) signals produced by Gram-negative bacteria, are currently considered to play a minor role in the development of oral biofilm since their production by oral pathogens has not been ascertained thus far. However, we report the presence of AHLs in different oral samples and their production by the oral pathogen Porphyromonas gingivalis. The importance of AHLs is further supported by a very high prevalence of AHL-degradation capability, up to 60%, among bacteria isolated from dental plaque and saliva samples. Furthermore, the wide-spectrum AHL-lactonase Aii20J significantly inhibited oral biofilm formation in different in vitro biofilm models and caused important changes in bacterial composition. Besides, the inhibitory effect of Aii20J on a mixed biofilm of 6 oral pathogens was verified using confocal microscopy. Much more research is needed in order to be able to associate specific AHLs with oral pathologies and to individuate the key actors in AHL-mediated QS processes in dental plaque formation. However, these results indicate a higher relevance of the AHLs in the oral cavity than generally accepted thus far and suggest the potential use of inhibitory strategies against these signals for the prevention and treatment of oral diseases.
Collapse
Affiliation(s)
- Andrea Muras
- Departamento de Microbioloxía e Parasitoloxía, Facultade de Bioloxía-CIBUS, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Paz Otero-Casal
- Departamento de Ciruxía e Especialidade Médico-Cirúrxica, Facultade de Medicina e Odontoloxía, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
- Unit of Oral Health, C.S. Santa Comba-Negreira, SERGAS, Spain
| | - Vanessa Blanc
- Department of Microbiology, Dentaid Research Center, Dentaid S.L., Barcelona, Spain
| | - Ana Otero
- Departamento de Microbioloxía e Parasitoloxía, Facultade de Bioloxía-CIBUS, Universidade de Santiago de Compostela, Santiago de Compostela, Spain.
| |
Collapse
|
39
|
Bijle MN, Neelakantan P, Ekambaram M, Lo ECM, Yiu CKY. Effect of a novel synbiotic on Streptococcus mutans. Sci Rep 2020; 10:7951. [PMID: 32409686 PMCID: PMC7224275 DOI: 10.1038/s41598-020-64956-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 04/22/2020] [Indexed: 12/21/2022] Open
Abstract
We examined the effect of L-arginine - (i) on the growth of L. rhamnosus GG (LrG) and (ii) combined LrG synbiotic on the growth of cariogenic S. mutans. Viability of LrG was assessed using MTT/XTT assays, confocal imaging with ADS activity measurement. The effect of L-arginine (0.5%/1%/2%) (2×/24 h) with LrG on S. mutans was evaluated by measuring the colony forming units, biofilm biomass, real-time qPCR and confocal imaging. The pH of the spent media was measured immediately and 24 h post-treatment with assessment of lactic acid. The LrG viability was highest with 2% L-arginine (p < 0.001). Confocal imaging showed that 2% L-arginine increased biofilm thickness of LrG. The 2% L-arginine and LrG synbiotic significantly inhibited the growth of S. mutans (p < 0.001) reducing the viable counts (p = 0.002) and biofilm biomass (p < 0.001). The pH of spent media was the highest when treated with 2% L-arginine and LrG synbiotic (p < 0.001) with no difference between post-treatment and 24 h post-treatment (p > 0.05). Conversely, the 2% L-arginine and LrG synbiotic showed the lowest lactic acid production (p < 0.001). This study demonstrated that L-arginine enhanced the growth of LrG. The 2% L-arginine and LrG synbiotic synergistically inhibits the growth of S. mutans with significant potential to develop as an anti-caries regimen.
Collapse
Affiliation(s)
- Mohammed Nadeem Bijle
- Paediatric Dentistry, Faculty of Dentistry, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Prasanna Neelakantan
- Endodontology, Faculty of Dentistry, The University of Hong Kong, Pok Fu Lam, Hong Kong.
| | - Manikandan Ekambaram
- Paediatric Dentistry, Faculty of Dentistry, University of Otago, Dunedin, New Zealand
| | - Edward C M Lo
- Dental Public Health, Faculty of Dentistry, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Cynthia Kar Yung Yiu
- Paediatric Dentistry, Faculty of Dentistry, The University of Hong Kong, Pok Fu Lam, Hong Kong.
| |
Collapse
|
40
|
Baraniya D, Naginyte M, Chen T, Albandar JM, Chialastri SM, Devine DA, Marsh PD, Al-Hebshi NN. Modeling Normal and Dysbiotic Subgingival Microbiomes: Effect of Nutrients. J Dent Res 2020; 99:695-702. [PMID: 31999932 DOI: 10.1177/0022034520902452] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Screening for microbiome modulators requires availability of a high-throughput in vitro model that replicates subgingival dysbiosis and normobiosis, with a tool to measure microbial dysbiosis. Here, we tested various formulations to grow health- and periodontitis-associated subgingival microbiomes in parallel, and we describe a new subgingival dysbiosis index. Subgingival plaque samples pooled from 5 healthy subjects and, separately, 5 subjects with periodontitis were used to inoculate a Calgary Biofilm Device containing saliva-conditioned, hydroxyapatite-coated pegs. Microbiomes were grown for 7 d on either nutrient-rich media-including a modification of SHI medium, brain-heart infusion (BHI) supplemented with hemin and vitamin K, and a blend of SHI and BHI, each at 3 sucrose concentrations (0%, 0.05% and 0.1%)-or nutrient-limited media (saliva with 5%, 10%, or 20% inactivated human serum). The microbiomes were assessed for biomass, viability, and 16S rRNA profiles. In addition to richness and diversity, a dysbiosis index was calculated as the ratio of the sum of relative abundances of disease-associated species to that of health-associated species. The supplemented BHI and blend of SHI and BHI resulted in the highest biomass, whereas saliva-serum maximized viability. Distinct groups of bacteria were enriched in the different media. Regardless of medium type, the periodontitis-derived microbiomes showed higher species richness and alpha diversity and clustered with their inoculum separate from the health-derived microbiomes. Microbiomes grown in saliva-serum showed the highest species richness and the highest similarity to the clinical inocula in both health and disease. However, inclusion of serum reduced alpha diversity and increased dysbiosis in healthy microbiomes in a dose-dependent manner, mainly due to overenrichment of Porphyromonas species. The modification of SHI stood second in terms of species richness and diversity but resulted in low biomass and viability and significantly worsened dysbiosis in the periodontitis-derived microbiomes. Overall, saliva with 5% human serum was optimal for replicating subgingival microbiomes from health and disease.
Collapse
Affiliation(s)
- D Baraniya
- Oral Microbiome Research Laboratory, Department of Oral Health Sciences, Maurice H. Kornberg School of Dentistry, Temple University, Philadelphia, PA, USA
| | - M Naginyte
- Division of Oral Biology, School of Dentistry, University of Leeds, Leeds, UK
| | - T Chen
- Department of Microbiology, Forsyth Institute, Cambridge, MA, USA
| | - J M Albandar
- Department of Periodontology and Oral Implantology, Maurice H. Kornberg School of Dentistry, Temple University, Philadelphia, PA, USA
| | - S M Chialastri
- Department of Periodontology and Oral Implantology, Maurice H. Kornberg School of Dentistry, Temple University, Philadelphia, PA, USA
| | - D A Devine
- Division of Oral Biology, School of Dentistry, University of Leeds, Leeds, UK
| | - P D Marsh
- Division of Oral Biology, School of Dentistry, University of Leeds, Leeds, UK
| | - N N Al-Hebshi
- Oral Microbiome Research Laboratory, Department of Oral Health Sciences, Maurice H. Kornberg School of Dentistry, Temple University, Philadelphia, PA, USA
| |
Collapse
|
41
|
Zhao M, Qu Y, Liu J, Mai S, Gu L. A universal adhesive incorporating antimicrobial peptide nisin: effects on Streptococcus mutans and saliva-derived multispecies biofilms. Odontology 2020; 108:376-385. [PMID: 31912370 DOI: 10.1007/s10266-019-00478-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 11/17/2019] [Indexed: 12/13/2022]
Abstract
For purpose of enhancing the antibacterial activity of a universal adhesive, the antimicrobial peptide nisin was incorporated into Single Bond Universal and its antibacterial effect on Streptococcus mutans monospecific biofilms and saliva-derived multispecies biofilms was studied. Nisin was incorporated into Single Bond Universal and the antibacterial activity was examined by confocal laser scanning microscopy (CLSM), reverse transcription-quantitative polymerase chain reaction (qRT-PCR), phenol-sulfuric acid method and lactate dehydrogenase enzymatic method. The bonding properties were tested by microtensile bond strength (μTBS) and degree of conversion (DC). Data were analyzed by one-way analysis of variance (ANOVA) and least significant difference multiple comparison tests (P < 0.05). The Single Bond Universal incorporated with 3% (w/v) nisin could significantly inhibit the growth of the S. mutans monospecific biofilms (P< 0.01) and decrease the expression of genes related to extracellular polysaccharide (EPS) synthesis (gtfB, gtfC, gtfD and spaP) and acidogenicity (ldh) (P < 0.05). 3% (w/v) nisin-incorporated Single Bond Universal could also inhibit the growth of saliva-derived multispecies biofilms and decrease the excretion of EPS and lactic acid ( P< 0.05). μTBS and DC of 3% (w/v) nisin-incorporated Single Bond Universal did not deteriorate obviously (P > 0.05). In conclusion, 3% (w/v) nisin-incorporated Single Bond Universal substantially inhibited the growth of both S. mutans monospecific and saliva-derived multispecies biofilms without compromising the bonding properties.
Collapse
Affiliation(s)
- Manduo Zhao
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, 56 Lingyuanxi Road, Yuexiu District, Guangzhou, Guangdong Province, China
| | - Yang Qu
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, 56 Lingyuanxi Road, Yuexiu District, Guangzhou, Guangdong Province, China
| | - Jia Liu
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, 56 Lingyuanxi Road, Yuexiu District, Guangzhou, Guangdong Province, China
| | - Sui Mai
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, 56 Lingyuanxi Road, Yuexiu District, Guangzhou, Guangdong Province, China.
| | - Lisha Gu
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Hospital of Stomatology, Sun Yat-sen University, 56 Lingyuanxi Road, Yuexiu District, Guangzhou, Guangdong Province, China.
| |
Collapse
|
42
|
Cendra MDM, Blanco-Cabra N, Pedraz L, Torrents E. Optimal environmental and culture conditions allow the in vitro coexistence of Pseudomonas aeruginosa and Staphylococcus aureus in stable biofilms. Sci Rep 2019; 9:16284. [PMID: 31705015 PMCID: PMC6841682 DOI: 10.1038/s41598-019-52726-0] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 10/22/2019] [Indexed: 12/15/2022] Open
Abstract
The coexistence between species that occurs in some infections remains hard to achieve in vitro since bacterial fitness differences eventually lead to a single organism dominating the mixed culture. Pseudomonas aeruginosa and Staphylococcus aureus are major pathogens found growing together in biofilms in disease-affected lungs or wounds. Herein, we tested and analyzed different culture media, additives and environmental conditions to support P. aeruginosa and S. aureus coexistence in vitro. We have unraveled the potential of DMEM to support the growth of these two organisms in mature cocultured biofilms (three days old) in an environment that dampens the pH rise. Our conditions use equal initial inoculation ratios of both strains and allow the stable formation of separate S. aureus microcolonies that grow embedded in a P. aeruginosa biofilm, as well as S. aureus biofilm overgrowth when bovine serum albumin is added to the system. Remarkably, we also found that S. aureus survival is strictly dependent on a well-characterized phenomenon of oxygen stratification present in the coculture biofilm. An analysis of differential tolerance to gentamicin and ciprofloxacin treatment, depending on whether P. aeruginosa and S. aureus were growing in mono- or coculture biofilms, was used to validate our in vitro coculture conditions.
Collapse
Affiliation(s)
- Maria Del Mar Cendra
- Bacterial Infections and Antimicrobial Therapies Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Baldiri Reixac 15-21, 08028, Barcelona, Spain.
| | - Núria Blanco-Cabra
- Bacterial Infections and Antimicrobial Therapies Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Baldiri Reixac 15-21, 08028, Barcelona, Spain
| | - Lucas Pedraz
- Bacterial Infections and Antimicrobial Therapies Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Baldiri Reixac 15-21, 08028, Barcelona, Spain
| | - Eduard Torrents
- Bacterial Infections and Antimicrobial Therapies Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Baldiri Reixac 15-21, 08028, Barcelona, Spain.
| |
Collapse
|
43
|
Fanesi A, Paule A, Bernard O, Briandet R, Lopes F. The Architecture of Monospecific Microalgae Biofilms. Microorganisms 2019; 7:microorganisms7090352. [PMID: 31540235 PMCID: PMC6780892 DOI: 10.3390/microorganisms7090352] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Revised: 09/03/2019] [Accepted: 09/06/2019] [Indexed: 11/16/2022] Open
Abstract
Microalgae biofilms have been proposed as an alternative to suspended cultures in commercial and biotechnological fields. However, little is known about their architecture that may strongly impact biofilm behavior, bioprocess stability, and productivity. In order to unravel the architecture of microalgae biofilms, four species of commercial interest were cultivated in microplates and characterized using a combination of confocal laser scanning microscopy and FTIR spectroscopy. In all the species, the biofilm biovolume and thickness increased over time and reached a plateau after seven days; however, the final biomass reached was very different. The roughness decreased during maturation, reflecting cell division and voids filling. The extracellular polymeric substances content of the matrix remained constant in some species, and increased over time in some others. Vertical profiles showed that young biofilms presented a maximum cell density at 20 μm above the substratum co-localized with matrix components. In mature biofilms, the maximum density of cells moved at a greater distance from the substratum (30–40 μm), whereas the maximum coverage of matrix components remained in a deeper layer. Carbohydrates and lipids were the main macromolecules changing during biofilm maturation. Our results revealed that the architecture of microalgae biofilms is species-specific. However, time similarly affects the structural and biochemical parameters.
Collapse
Affiliation(s)
- Andrea Fanesi
- Laboratoire Génie des Procédés et Matériaux (LGPM), CentraleSupélec, Université Paris-Saclay, 91190 Gif-sur-Yvette, France.
| | - Armelle Paule
- Laboratoire Génie des Procédés et Matériaux (LGPM), CentraleSupélec, Université Paris-Saclay, 91190 Gif-sur-Yvette, France.
| | - Olivier Bernard
- Université Côte d'Azur, Inria, BIOCORE, BP 93, 06902 Sophia Antipolis Cedex, France.
| | - Romain Briandet
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350 Jouy-en-Josas, France.
| | - Filipa Lopes
- Laboratoire Génie des Procédés et Matériaux (LGPM), CentraleSupélec, Université Paris-Saclay, 91190 Gif-sur-Yvette, France.
| |
Collapse
|
44
|
|
45
|
Spontaneously Arising Streptococcus mutans Variants with Reduced Susceptibility to Chlorhexidine Display Genetic Defects and Diminished Fitness. Antimicrob Agents Chemother 2019; 63:AAC.00161-19. [PMID: 31036688 DOI: 10.1128/aac.00161-19] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 04/20/2019] [Indexed: 01/10/2023] Open
Abstract
Chlorhexidine (CHX) has been used to control dental caries caused by acid-tolerant bacteria such as Streptococcus mutans since the 1970s. Repeat CHX exposure for other bacterial species results in the development of variants with reduced susceptibility that also become more resistant to other antimicrobials. It has not been tested if such variants arise when streptococci are exposed to CHX. Here, we passaged S. mutans in increasing concentrations of CHX and isolated spontaneously arising reduced susceptibility variants (RSVs) from separate lineages that have MICs that are up to 3-fold greater than the parental strain. The RSVs have increased growth rates at neutral pH and under acidic conditions in the presence of CHX but accumulate less biomass in biofilms. RSVs display higher MICs for daptomycin and clindamycin but increased sensitivity to dental-relevant antimicrobials triclosan and sodium fluoride. Plate-based assays for competition with health-associated oral streptococci revealed decreased bacteriocin production by the RSVs, increased sensitivity to hydrogen peroxide, and diminished competitive fitness in a human-derived ex vivo biofilm consortium. Whole-genome sequencing identified common single nucleotide polymorphisms (SNPs) within a diacylglycerol kinase homolog and a glycolipid synthesis enzyme, which could alter the accumulation of lipoteichoic acids and other envelope constituents, as well as a variety of mutations in other genes. Collectively, these findings confirm that S. mutans and likely other streptococci can develop tolerance to CHX but that increased tolerance comes at a fitness cost, such that CHX-induced variants that spontaneously arise in the human oral cavity may not persist.
Collapse
|
46
|
Amino Sugars Modify Antagonistic Interactions between Commensal Oral Streptococci and Streptococcus mutans. Appl Environ Microbiol 2019; 85:AEM.00370-19. [PMID: 30877119 DOI: 10.1128/aem.00370-19] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 03/12/2019] [Indexed: 02/05/2023] Open
Abstract
N-Acetylglucosamine (GlcNAc) and glucosamine (GlcN) enhance the competitiveness of the laboratory strain DL1 of Streptococcus gordonii against the caries pathogen Streptococcus mutans Here, we examine how amino sugars affect the interaction of five low-passage-number clinical isolates of abundant commensal streptococci with S. mutans by utilizing a dual-species biofilm model. Compared to that for glucose, growth on GlcN or GlcNAc significantly reduced the viability of S. mutans in cocultures with most commensals, shifting the proportions of species. Consistent with these results, production of H2O2 was increased in most commensals when growing on amino sugars, and inhibition of S. mutans by Streptococcus cristatus, Streptococcus oralis, or S. gordonii was enhanced by amino sugars on agar plates. All commensals except S. oralis had higher arginine deiminase activities when grown on GlcN and, in some cases, GlcNAc. In ex vivo biofilms formed using pooled cell-containing saliva (CCS), the proportions of S. mutans were drastically diminished when GlcNAc was the primary carbohydrate. Increased production of H2O2 could account in large part for the inhibitory effects of CCS biofilms. Surprisingly, amino sugars appeared to improve mutacin production by S. mutans on agar plates, suggesting that the commensals have mechanisms to actively subvert antagonism by S. mutans in cocultures. Collectively, these findings demonstrate that amino sugars can enhance the beneficial properties of low-passage-number commensal oral streptococci and highlight their potential for moderating the cariogenicity of oral biofilms.IMPORTANCE Dental caries is driven by dysbiosis of oral biofilms in which dominance by acid-producing and acid-tolerant bacteria results in loss of tooth mineral. Our previous work demonstrated the beneficial effects of amino sugars GlcNAc and GlcN in promoting the antagonistic properties of a health-associated oral bacterium, Streptococcus gordonii, in competition with the major caries pathogen Streptococcus mutans Here, we investigated 5 low-passage-number clinical isolates of the most common streptococcal species to establish how amino sugars may influence the ecology and virulence of oral biofilms. Using multiple in vitro models, including a human saliva-derived microcosm biofilm, experiments showed significant enhancement by at least one amino sugar in the ability of most of these bacteria to suppress the caries pathogen. Therefore, our findings demonstrated the mechanism of action by which amino sugars may affect human oral biofilms to promote health.
Collapse
|
47
|
Ikeda K, Ejima D, Arakawa T, Koyama AH. Protein aggregation suppressor arginine as an effective mouth cleaning agent. Int J Biol Macromol 2019; 122:224-227. [PMID: 30393137 DOI: 10.1016/j.ijbiomac.2018.10.158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Revised: 10/23/2018] [Accepted: 10/23/2018] [Indexed: 11/28/2022]
Abstract
We have tested here whether or not arginine, a well-known aggregation suppressor, is effective in removing bacterial cells, which may present a potential risk of accidental pneumonia infection in aged individuals, from the oral mucosal membranes. This is based on the ability of arginine to suppress protein-protein interaction and surface adsorption and increase the solubility of organic compounds. Twelve student volunteers were subjected to mouthwashes with saline, citrate buffer (pH 3.5), arginine (pH 3.5) and a commercial Listerine. Insignificant effects were observed with saline and citrate buffer, whereas arginine and Listerine mouthwashes led to significant reduction of bacterial cells from the dorsal side of the volunteer's tongue. Arginine also appeared to disrupt biofilms present in the mouth.
Collapse
Affiliation(s)
- Keiko Ikeda
- Adult Nursing, School of Health and Nursing Science, Wakayama Medical University, Wakayama 641-0011, Wakayama, Japan
| | - Daisuke Ejima
- Technology Development, Sysmex Corporation, 4-4-4 Takatsukadai, Nishi-ku, Kobe 651-2271, Hyogo, Japan
| | - Tsutomu Arakawa
- Alliance Protein Laboratories, San Diego, CA 92121, United States of America.
| | - A Hajime Koyama
- Graduate School of Medicine, Wakayama Medical University, Wakayama 641-0011, Wakayama, Japan
| |
Collapse
|
48
|
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
|
49
|
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
|
50
|
Dutta K, Shityakov S, Khalifa I, Mal A, Moulik SP, Panda AK, Ghosh C. Effects of secondary carbon supplement on biofilm-mediated biodegradation of naphthalene by mutated naphthalene 1, 2-dioxygenase encoded by Pseudomonas putida strain KD9. JOURNAL OF HAZARDOUS MATERIALS 2018; 357:187-197. [PMID: 29886364 DOI: 10.1016/j.jhazmat.2018.05.024] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Revised: 05/11/2018] [Accepted: 05/12/2018] [Indexed: 06/08/2023]
Abstract
Polycyclic aromatic hydrocarbons (PAHs) belong to a diverse group of environmental pollutants distributed ubiquitously in the environment. The carcinogenic properties of PAHs are the main causes of harm to human health. The green technology, biodegradation have become convenient options to address the environmental pollution. In this study, we analyzed the biodegradation potential of naphthalene with secondary carbon supplements (SCSs) in carbon deficient media (CSM) by Pseudomonas putida strain KD9 isolated from oil refinerary waste. The rigid-flexible molecular docking method revealed that the mutated naphthalene 1,2-dioxygenase had lower affinity for naphthalene than that found in wild type strain. Moreover, analytical methods (HPLC, qRT-PCR) and soft agar chemotaxis suggest sucrose (0.5 wt%) to be the best chemo-attractant and it unequivocally caused enhanced biodegradation of naphthalene (500 mg L-1) in both biofilm-mediated and shake-flask biodegradation methods. In addition, the morphological analysis detected from microscopy clearly showed KD9 to change its size and shape (rod to pointed) during biodegradation of naphthalene in CSM as sole source of carbon and energy. The forward versus side light scatter plot of the singlet cells obtained from flow cytometry suggests smaller cell size in CSM and lower florescence intensity of the total DNA content of cells. This study concludes that sucrose may be used as potential bio-stimulation agent.
Collapse
Affiliation(s)
- Kunal Dutta
- Microbiology and Immunology Laboratory, Department of Human Physiology with Community Health, Vidyasagar University, Midnapore-721102, West Bengal, India; Department of Chemistry and Chemical Technology, Vidyasagar University, Midnapore-721102, West Bengal, India
| | - Sergey Shityakov
- Department of Anaesthesia and Critical Care, University of Würzburg-97080, Würzburg, Germany
| | - Ibrahim Khalifa
- Food Technology Department, Faculty of Agriculture, 13736, Moshtohor, Benha University, Egypt; College of Food Science and Technology, Huazhong Agricultural University, Wuhan-430070, China
| | - Arpan Mal
- Center for Surface Science, Department of Chemistry, Jadavpur University, Kolkata 700032, India
| | - Satya Priya Moulik
- Center for Surface Science, Department of Chemistry, Jadavpur University, Kolkata 700032, India
| | - Amiya Kumar Panda
- Department of Chemistry and Chemical Technology, Vidyasagar University, Midnapore-721102, West Bengal, India
| | - Chandradipa Ghosh
- Microbiology and Immunology Laboratory, Department of Human Physiology with Community Health, Vidyasagar University, Midnapore-721102, West Bengal, India.
| |
Collapse
|