1
|
Sangavi R, Jothi R, Malligarjunan N, Raja V, Pandian SK, Gowrishankar S. Cetyltrimethylammonium Chloride (CTAC) and Its Formulated Mouthwash Reduce the Infectivity of Streptococcus mutans and Candida albicans in Mono and Dual State. Appl Biochem Biotechnol 2024:10.1007/s12010-024-05119-7. [PMID: 39731640 DOI: 10.1007/s12010-024-05119-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/19/2024] [Indexed: 12/30/2024]
Abstract
Early childhood caries (ECC), a severe form of dental caries, is exacerbated by the synergistic interaction between Streptococcus mutans and Candida albicans, leading to greater disease severity than their individual effects. This underscores the need for more targeted and potent therapeutic alternatives. Given the promising anti-infective properties of quaternary ammonium surfactants (QAS), this study explores the microbicidal properties of one such QAS, cetyltrimethylammonium chloride (CTAC), against both individual- and dual-species cultures of S. mutans and C. albicans for effective ECC treatment. Initially, the minimal inhibitory concentrations (MICs) of CTAC were determined to range from 4 to 8 µg/mL against S. mutans, C. albicans, and dual-species cultures. Time-kill kinetics, assessed via spot assays and spectrometry, demonstrated that CTAC completely eradicated both individual- and dual-species cultures within 30 min of exposure. Furthermore, at sub-MIC concentrations, CTAC effectively reduced biofilm formation and virulence traits in S. mutans (including acidogenicity and aciduricity) and C. albicans (including yeast-to-hyphal transition and filamentation). To explore therapeutic application, a mouthwash containing CTAC was formulated. The results showed that the formulated CTAC mouthwash was as effective at eradicating pathogens as a commercially available mouthwash containing 0.075% cetylpyridinium chloride (CPC). Moreover, the CTAC mouthwash maintained stable physicochemical characteristics and antimicrobial activity over 4 weeks. It exhibited rapid killing activity against pathogens, achieving efficacy within just 2 min of exposure. Fluorescence microscopy and SEM micrographs confirmed the strong biofilm eradication potential of the CTAC mouthwash. The non-toxic nature of the formulated mouthwash was validated using human buccal epithelial cells, and in vivo studies further demonstrated that CTAC mouthwash significantly reduced bacterial and fungal loads in Galleria mellonella. Overall, the findings of this study highlight the potential application of QAS-CTAC in the treatment of ECC.
Collapse
Affiliation(s)
- Ravichellam Sangavi
- Department of Biotechnology, Science Campus, Alagappa University, Karaikudi, India
| | - Ravi Jothi
- Department of Biotechnology, Science Campus, Alagappa University, Karaikudi, India
| | | | - Veerapandian Raja
- Department of Molecular and Translational Medicine, Center of Emphasis in Infectious Diseases, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, 79905, USA
| | | | | |
Collapse
|
2
|
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
|
3
|
Campbell PM, Willmott T, Summers A, Knight CG, Humphreys GJ, Konkel JE, Augustine T, McBain AJ. Investigating oral microbiome dynamics in chronic kidney disease and post-transplantation in continuous culture. Microbiol Spectr 2024; 12:e0059824. [PMID: 39382278 PMCID: PMC11537021 DOI: 10.1128/spectrum.00598-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 08/02/2024] [Indexed: 10/10/2024] Open
Abstract
The oral microbiome is influenced by environmental factors in chronic kidney disease and following kidney transplantation affecting microbial composition, which may have implications for health and recovery. A major driver of oral microbiome perturbation is the accumulation of urea in saliva. We have modelled increased salivary urea concentrations associated with CKD and subsequent reductions that may occur post-transplantation. Oral microbiota were established in constant-depth film fermenters by inoculation with saliva. Duplicate validation runs were maintained with artificial saliva with baseline urea concentrations (0.205 mg/mL) for 21 days. Triplicate treatment runs were then done with baseline urea for 10 days (healthy phase) before urea was increased for 10 days to reflect CKD concentrations (0.92 mg/mL) (CKD phase). This was followed by reversion to baseline urea concentrations (post-transplant phase). Biofilms in primary validation runs reached dynamic stability within 5 days according to viable counting. DNA sequence data indicated minimal taxonomic variation over time and between low and high urea treatments despite background noise indicating changes in bacteria belonging to the family Gemellaceae and the genera TG5 and Leptotrichia. Significant differences in alpha and beta diversity occurred between low and high urea states but not following reversion to a low urea environment. Increased abundance of the TG5 was detected in late model phases, despite apparent count stability, and independent of changes in urea concentrations. IMPORTANCE This study investigates dynamic changes in the oral microbiome associated with changes in salivary urea concentration, an important factor in chronic kidney disease (CKD). The in vitro system modeled increased urea concentrations and subsequent reductions post-transplantation. The study provides insight into the oral microbial shifts during different simulated clinical phases. Understanding these dynamics is crucial for advancing our comprehension of CKD-associated oral microbiome variations and their potential impact on patient well-being and recovery.
Collapse
Affiliation(s)
- Paul M. Campbell
- School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Thomas Willmott
- School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Angela Summers
- Department of Renal and Pancreatic Transplantation, Manchester Academic Health Science Centre, Manchester University Hospitals NHS Foundation Trust, Manchester, United Kingdom
| | - Christopher G. Knight
- School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester, Manchester, United Kingdom
| | - Gavin J. Humphreys
- School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Joanne E. Konkel
- School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Titus Augustine
- Department of Renal and Pancreatic Transplantation, Manchester Academic Health Science Centre, Manchester University Hospitals NHS Foundation Trust, Manchester, United Kingdom
| | - Andrew J. McBain
- School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
4
|
Gonzalez Agurto M, Olivares N, Canedo-Marroquin G, Espinoza D, Tortora SC. The Intersection of the Oral Microbiome and Salivary Metabolites in Head and Neck Cancer: From Diagnosis to Treatment. Cancers (Basel) 2024; 16:3545. [PMID: 39456639 PMCID: PMC11506592 DOI: 10.3390/cancers16203545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/11/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
Head and neck cancers (HNCs) are the seventh most common cancer worldwide, accounting for 4-5% of all malignancies. Salivary metabolites, which serve as key metabolic intermediates and cell-signalling molecules, are emerging as potential diagnostic biomarkers for HNC. While current research has largely concentrated on these metabolites as biomarkers, a critical gap remains in understanding their fluctuations before and after treatment, as well as their involvement in oral side effects. Recent studies emphasise the role of the oral microbiome and its metabolic activity in cancer progression and treatment efficacy by bacterial metabolites and virulence factors. Oral bacteria, such as P. gingivalis and F. nucleatum, contribute to a pro-inflammatory environment that promotes tumour growth. Additionally, F. nucleatum enhances its virulence through flagellar assembly and iron transport mechanisms, facilitating tumour invasion and survival. Moreover, alterations in the oral microbiome can influence chemotherapy efficacy and toxicity through the microbiota-host irinotecan axis, highlighting the complex interplay between microbial communities and therapeutic outcomes. Salivary metabolite profiles are influenced by factors such as gender, methods, and patient habits like smoking-a major risk factor for HNC. Radiotherapy (RT), a key treatment for HNC, often causes side effects such as xerostomia, oral mucositis, and swallowing difficulties which impact survivors' quality of life. Intensity-modulated radiotherapy (IMRT) aims to improve treatment outcomes and minimise side effects but can still lead to significant salivary gland dysfunction and associated complications. This review underscores the microbial and host interactions affecting salivary metabolites and their implications for cancer treatment and patient outcomes.
Collapse
Affiliation(s)
| | - Nicolas Olivares
- Faculty of Dentistry, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile;
| | - Gisela Canedo-Marroquin
- Faculty of Dentistry, Universidad de los Andes, Santiago 7620086, Chile;
- Faculty of Dentistry, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile;
- Millennium Institute on Immunology and Immunotherapy (MIII), Santiago 8331150, Chile
| | - Daniela Espinoza
- Faculty of Dentistry, Universidad Mayor, Santiago 8580745, Chile
| | - Sofia C. Tortora
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|
5
|
Siddiqui DA, Lakkasetter Chandrashekar B, Natarajan SG, Palmer KL, Rodrigues DC. Development of a Coculture Model for Assessing Competing Host Mammalian Cell and Bacterial Attachment on Zirconia versus Titanium. ACS Biomater Sci Eng 2024; 10:6218-6229. [PMID: 39312708 DOI: 10.1021/acsbiomaterials.4c01075] [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] [Indexed: 09/25/2024]
Abstract
Objectives: Coculture models are limited by bacteria rapidly outcompeting host mammalian cells for nutrients in vitro, resulting in mammalian cell death. The goal of this study was to develop a coculture model enabling survival of mammalian cells and oral bacterial species to assess their competition for growth on dental implant materials. Methods: Two early colonizing oral bacterial species, Streptococcus mutans or Actinomyces naeslundii, were grown in coculture with primary human macrophages or human gingival fibroblasts for up to 7 days on tissue-culture treated polystyrene or polished titanium and zirconia disks. Chloramphenicol was supplemented in cell culture medium at bacteriostatic concentrations to maintain stable bacterial inoculum size. Planktonic and adherent bacterial growth was assessed via spot plating while mammalian cell growth and attachment were evaluated using colorimetric metabolic assay and confocal fluorescence microscopy, respectively. Results: Macrophages and fibroblasts proliferated in the presence of S. mutans and maintained viability above 70% during coculture for up to 7 days on tissue-culture treated polystyrene and polished titanium and zirconia. In contrast, both mammalian cell types exhibited decreasing proliferation and surface coverage on titanium and zirconia over time in coculture with A. naeslundii versus control. S. mutans and A. naeslundii were maintained within an order of magnitude of seeding inoculum sizes throughout coculture. Significance: Cell culture medium supplemented with antibiotics at bacteriostatic concentrations can suppress bacterial overgrowth and facilitate mammalian cell viability in coculture model systems. Within the study's limitations, oral bacteria and mammalian cell growth in coculture are comparable on polished titanium and zirconia surfaces.
Collapse
Affiliation(s)
- Danyal A Siddiqui
- Department of Bioengineering, The University of Texas at Dallas, 800 W. Campbell Road, Richardson, Texas 75080, United States
| | | | - Smriti G Natarajan
- Texas A&M University School of Dentistry, 3302 Gaston Avenue, Dallas, Texas 75246, United States
| | - Kelli L Palmer
- Department of Biological Sciences, The University of Texas at Dallas, 800 W. Campbell Road, Richardson, Texas 75080, United States
| | - Danieli C Rodrigues
- Department of Bioengineering, The University of Texas at Dallas, 800 W. Campbell Road, Richardson, Texas 75080, United States
| |
Collapse
|
6
|
Kuriki N, Asahi Y, Okamoto M, Noiri Y, Ebisu S, Machi H, Suzuki M, Hayashi M. Synergistic effects of arginine and fluoride on human dental biofilm control. J Dent 2024; 149:105307. [PMID: 39178800 PMCID: PMC11391429 DOI: 10.1016/j.jdent.2024.105307] [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: 07/11/2024] [Revised: 08/07/2024] [Accepted: 08/10/2024] [Indexed: 08/26/2024] Open
Abstract
OBJECTIVES The aim of this study was to quantitatively and comprehensively investigate the combined effects of arginine and fluoride on the suppression of pathogenicity using an in situ biofilm model and next-generation sequencing (NGS). METHODS Using the in situ model, dental biofilms were formed and the viable bacterial counts and arginine activity in the arginine- and fluoride-containing dentifrice and control groups were measured. We also compared their effects on the bacterial microbiota and predictive functional factors in the control, arginine (arg), and arginine + fluoride (argF) groups using NGS analysis. RESULTS Compared to the control treatment, the use of 8 % arginine and 1450 ppm fluoride toothpaste resulted in significantly high oral NH4+ concentrations without affecting the number of viable bacteria (P < 0.05). NGS analysis revealed that the oral microbiota of the control, arg, and argF groups were significantly different. Heat map analysis of the predicted functional factors revealed that the arg group had different properties from the other groups and activated specific substrate metabolic pathways; contrastingly, argF treatment inhibited the activity of these pathways and prevented an increase in the abundance of bacterial genera that utilize substrates such as sucrose, suggesting the synergistic effect of arginine and fluoride. CONCLUSIONS This study indicates that the combination of arginine and fluoride has a synergistic effect on the bacterial microbiota and pathogenicity of dental biofilms compared with arginine alone. CLINICAL SIGNIFICANCE Our findings suggest that the combination of arginine and fluoride could be used as an effective prebiotic and may inhibit the growth of bacteria associated with dental diseases.
Collapse
Affiliation(s)
- Nanako Kuriki
- Department of Restorative Dentistry and Endodontology, Osaka University Graduate School of Dentistry, Osaka, Japan; Department of Oral Science and Translational Research, College of Dental Medicine, Nova Southeastern University, Fort Lauderdale, FL, USA.
| | - Yoko Asahi
- Department of Restorative Dentistry and Endodontology, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Motoki Okamoto
- Department of Oral Science and Translational Research, College of Dental Medicine, Nova Southeastern University, Fort Lauderdale, FL, USA
| | - Yuichiro Noiri
- Division of Cariology, Operative Dentistry and Endodontics, Department of Oral Health Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Shigeyuki Ebisu
- Department of Restorative Dentistry and Endodontology, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Hiroyuki Machi
- Osaka University Dental Technology Institute, Osaka, Japan
| | - Maiko Suzuki
- Department of Oral Science and Translational Research, College of Dental Medicine, Nova Southeastern University, Fort Lauderdale, FL, USA
| | - Mikako Hayashi
- Department of Restorative Dentistry and Endodontology, Osaka University Graduate School of Dentistry, Osaka, Japan
| |
Collapse
|
7
|
Bostanghadiri N, Kouhzad M, Taki E, Elahi Z, Khoshbayan A, Navidifar T, Darban-Sarokhalil D. Oral microbiota and metabolites: key players in oral health and disorder, and microbiota-based therapies. Front Microbiol 2024; 15:1431785. [PMID: 39228377 PMCID: PMC11368800 DOI: 10.3389/fmicb.2024.1431785] [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: 05/14/2024] [Accepted: 08/02/2024] [Indexed: 09/05/2024] Open
Abstract
The review aimed to investigate the diversity of oral microbiota and its influencing factors, as well as the association of oral microbiota with oral health and the possible effects of dysbiosis and oral disorder. The oral cavity harbors a substantial microbial burden, which is particularly notable compared to other organs within the human body. In usual situations, the microbiota exists in a state of equilibrium; however, when this balance is disturbed, a multitude of complications arise. Dental caries, a prevalent issue in the oral cavity, is primarily caused by the colonization and activity of bacteria, particularly streptococci. Furthermore, this environment also houses other pathogenic bacteria that are associated with the onset of gingival, periapical, and periodontal diseases, as well as oral cancer. Various strategies have been employed to prevent, control, and treat these disorders. Recently, techniques utilizing microbiota, like probiotics, microbiota transplantation, and the replacement of oral pathogens, have caught the eye. This extensive examination seeks to offer a general view of the oral microbiota and their metabolites concerning oral health and disease, and also the resilience of the microbiota, and the techniques used for the prevention, control, and treatment of disorders in this specific area.
Collapse
Affiliation(s)
- Narjess Bostanghadiri
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mobina Kouhzad
- Department of Genetics, Faculty of Science, Islamic Azad University North Tehran Branch, Tehran, Iran
| | - Elahe Taki
- Department of Microbiology, School of Medicine, Kermanshah University of Medical Science, Kermanshah, Iran
| | - Zahra Elahi
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Amin Khoshbayan
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Tahereh Navidifar
- Department of Basic Sciences, Shoushtar Faculty of Medical Sciences, Shoushtar, Iran
| | - Davood Darban-Sarokhalil
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
8
|
Ye D, Liu Y, Li J, Zhou J, Cao J, Wu Y, Wang X, Fang Y, Ye X, Zou J, Ma Q. Competitive dynamics and balance between Streptococcus mutans and commensal streptococci in oral microecology. Crit Rev Microbiol 2024:1-12. [PMID: 39132685 DOI: 10.1080/1040841x.2024.2389386] [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: 03/22/2024] [Revised: 06/01/2024] [Accepted: 08/02/2024] [Indexed: 08/13/2024]
Abstract
Dental caries, as a biofilm-related disease, is closely linked to dysbiosis in microbial ecology within dental biofilms. Beyond its impact on oral health, bacteria within the oral cavity pose systemic health risks by potentially entering the bloodstream, thereby increasing susceptibility to bacterial endocarditis, among other related diseases. Streptococcus mutans, a principal cariogenic bacterium, possesses virulence factors crucial to the pathogenesis of dental caries. Its ability to adhere to tooth surfaces, produce glucans for biofilm formation, and metabolize sugars into lactic acid contributes to enamel demineralization and the initiation of carious lesions. Its aciduricity and ability to produce bacteriocins enable a competitive advantage, allowing it to thrive in acidic environments and dominate in changing oral microenvironments. In contrast, commensal streptococci, such as Streptococcus sanguinis, Streptococcus gordonii, and Streptococcus salivarius, act as primary colonizers and compete with S. mutans for adherence sites and nutrients during biofilm formation. This competition involves the production of alkali, peroxides, and antibacterial substances, thereby inhibiting S. mutans growth and maintaining microbial balance. This dynamic interaction influences the balance of oral microbiota, with disruptions leading to shifts in microbial composition that are marked by rapid increases in S. mutans abundance, contributing to the onset of dental caries. Thus, understanding the dynamic interactions between commensal and pathogenic bacteria in oral microecology is important for developing effective strategies to promote oral health and prevent dental caries. This review highlights the roles and competitive interactions of commensal bacteria and S. mutans in oral microecology, emphasizing the importance of maintaining oral microbial balance for health, and discusses the pathological implications of perturbations in this balance.
Collapse
Affiliation(s)
- Dingwei Ye
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yaqi Liu
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jing Li
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jing Zhou
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jingwei Cao
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yumeng Wu
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xinyue Wang
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yuwen Fang
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xingchen Ye
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jing Zou
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Qizhao Ma
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
9
|
Nadaf R, Kumbar VM, Ghagane S. Unravelling the intricacies of Porphyromonas gingivalis: virulence factors, lifecycle dynamics and phytochemical interventions for periodontal disease management. APMIS 2024. [PMID: 39030947 DOI: 10.1111/apm.13440] [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: 01/09/2024] [Accepted: 05/14/2024] [Indexed: 07/22/2024]
Abstract
Porphyromonas gingivalis is a gram-negative anaerobic bacterium recognized for its pivotal role in the pathogenesis of periodontal diseases. This review covers an overview of the virulence factors and lifecycle stages of P. gingivalis, with a specific focus on attachment and colonization, biofilm formation, growth and multiplication, dormancy survival and dissemination. Additionally, we explore the significance of inter-bacterial cross-feeding within biofilms. Furthermore, we discuss potential phytochemical-based strategies to target P. gingivalis, including the use of curcumin, apigenin, quercetin and resveratrol. Understanding the virulence factors and lifecycle stages of P. gingivalis, along with the promising phytochemical-based interventions, holds promise for advancing strategies in periodontal disease management and oral health promotion.
Collapse
Affiliation(s)
- Rubeen Nadaf
- Dr. Prabhakar Kore Basic Science Research Centre, KLE Academy of Higher Education (KLE University), Belagavi, Karnataka, India
| | - Vijay M Kumbar
- Dr. Prabhakar Kore Basic Science Research Centre, KLE Academy of Higher Education (KLE University), Belagavi, Karnataka, India
| | - Shridhar Ghagane
- Dr. Prabhakar Kore Basic Science Research Centre, KLE Academy of Higher Education (KLE University), Belagavi, Karnataka, India
| |
Collapse
|
10
|
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
|
11
|
Lo K, Liman AN, Zhang Y, Ye W. Tongue coating metabolic profiles of intra-oral halitosis patients. Oral Dis 2024. [PMID: 38852162 DOI: 10.1111/odi.15034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 05/01/2024] [Accepted: 05/23/2024] [Indexed: 06/11/2024]
Abstract
OBJECTIVE This study focused on the metabolic characteristics of tongue coating in patients with intra-oral halitosis (IOH) to investigate potential diagnostic biomarkers for IOH. METHODS Oral healthy participants were enrolled in this study. Halitosis was evaluated with an organoleptic assessment, a Halimeter®, and an OralChroma™. Tongue coating samples were collected from 18 halitosis patients and 18 healthy controls. Liquid chromatography-mass spectrometry was conducted to reveal the IOH-related metabolic variations in tongue coating. RESULTS A total of 2214 metabolites were obtained. Most metabolites were shared between the two groups. A total of 274 upregulated metabolites, such as paramethasone acetate and indole-3-acetic acid, and 43 downregulated metabolites, including deoxyadenosine and valyl-arginine, were detected in the halitosis group. Functional analysis indicated that several metabolic pathways, including arginine biosynthesis, arginine and proline metabolism, histidine metabolism, and lysine degradation were significantly enriched in the IOH group. The least absolute shrinkage and selection operator logistic regression analysis revealed that paramethasone acetate, {1-[2-(4-carbamimidoyl-benzoylamino)-propionyl]-piperidin-4-yloxy}-acetic acid, indole-3-acetic acid, and valyl-arginine were remarkably associated with IOH. CONCLUSIONS This study revealed the metabolites present in tongue coating and identified effective biomarkers, providing essential insights into the prediction, pathogenesis, and diagnosis of IOH.
Collapse
Affiliation(s)
- Kalam Lo
- Department of Preventive Dentistry, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, Shanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai, China
- Shanghai Research Institute of Stomatology, Shanghai, China
| | - Aviella Nathania Liman
- Department of Preventive Dentistry, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, Shanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai, China
- Shanghai Research Institute of Stomatology, Shanghai, China
| | - Yu Zhang
- Department of Preventive Dentistry, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, Shanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai, China
- Shanghai Research Institute of Stomatology, Shanghai, China
| | - Wei Ye
- Department of Preventive Dentistry, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, Shanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai, China
- Shanghai Research Institute of Stomatology, Shanghai, China
| |
Collapse
|
12
|
Dahlquist-Axe G, Standeven FJ, Speller CF, Tedder A, Meehan CJ. Inferring diet, disease and antibiotic resistance from ancient human oral microbiomes. Microb Genom 2024; 10:001251. [PMID: 38739117 PMCID: PMC11165619 DOI: 10.1099/mgen.0.001251] [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: 11/20/2023] [Accepted: 04/24/2024] [Indexed: 05/14/2024] Open
Abstract
The interaction between a host and its microbiome is an area of intense study. For the human host, it is known that the various body-site-associated microbiomes impact heavily on health and disease states. For instance, the oral microbiome is a source of various pathogens and potential antibiotic resistance gene pools. The effect of historical changes to the human host and environment to the associated microbiome, however, has been less well explored. In this review, we characterize several historical and prehistoric events which are considered to have impacted the oral environment and therefore the bacterial communities residing within it. The link between evolutionary changes to the oral microbiota and the significant societal and behavioural changes occurring during the pre-Neolithic, Agricultural Revolution, Industrial Revolution and Antibiotic Era is outlined. While previous studies suggest the functional profile of these communities may have shifted over the centuries, there is currently a gap in knowledge that needs to be filled. Biomolecular archaeological evidence of innate antimicrobial resistance within the oral microbiome shows an increase in the abundance of antimicrobial resistance genes since the advent and widespread use of antibiotics in the modern era. Nevertheless, a lack of research into the prevalence and evolution of antimicrobial resistance within the oral microbiome throughout history hinders our ability to combat antimicrobial resistance in the modern era.
Collapse
Affiliation(s)
- Gwyn Dahlquist-Axe
- School of Chemistry and Biosciences, University of Bradford, Bradford, UK
| | | | - Camilla F. Speller
- Department of Anthropology, University of British Columbia, Vancouver, Canada
| | - Andrew Tedder
- School of Chemistry and Biosciences, University of Bradford, Bradford, UK
| | - Conor J. Meehan
- Department of Biosciences, Nottingham Trent University, Nottingham, UK
| |
Collapse
|
13
|
Zhang Y, Lo KL, Liman AN, Feng XP, Ye W. Tongue-Coating Microbial and Metabolic Characteristics in Halitosis. J Dent Res 2024; 103:484-493. [PMID: 38623900 DOI: 10.1177/00220345241230067] [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] [Indexed: 04/17/2024] Open
Abstract
Halitosis is a common oral condition, which leads to social embarrassment and affects quality of life. Cumulative evidence has suggested the association of tongue-coating microbiome with the development of intraoral halitosis. The dynamic variations of tongue-coating microbiota and metabolites in halitosis have not been fully elucidated. Therefore, the present study aimed to determine the tongue-coating microbial and metabolic characteristics in halitosis subjects without other oral diseases using metagenomics and metabolomics analysis. The participants underwent oral examination, halitosis assessment, and tongue-coating sample collection for the microbiome and metabolome analysis. It was found that the microbiota richness and diversity were significantly elevated in the halitosis group. Furthermore, species from Actinomyces, Prevotella, Veillonella, and Solobacterium were significantly more abundant in the halitosis group. However, the Rothia and Streptococcus species exhibited opposite tendencies. Eleven Kyoto Encyclopedia of Genes and Genomes pathways were significantly enriched in the halitosis tongue coatings, including cysteine and methionine metabolism. Functional genes related to sulfur, indole, skatole, and cadaverine metabolic processes (such as serA, metH, metK and dsrAB) were identified to be more abundant in the halitosis samples. The metabolome analysis revealed that indole-3-acetic, ornithine, and L-tryptophan were significantly elevated in the halitosis samples. Furthermore, it was observed that the values of volatile sulfur compounds and indole-3-acetic abundances were positively correlated. The multiomics analysis identified the metagenomic and metabolomic characteristics to differentiate halitosis from healthy individuals using the least absolute shrinkage and selection operator logistic regression and random forest classifier. A total of 19 species and 39 metabolites were identified as features in halitosis patients, which included indole-3-acetic acid, Bacillus altitudinis, Candidatus Saccharibacteria, and Actinomyces species. In conclusion, an evident shift in microbiome and metabolome characteristics was observed in the halitosis tongue coating, which may have a potential etiological significance and provide novel insights into the mechanism for halitosis.
Collapse
Affiliation(s)
- Y Zhang
- Department of Preventive Dentistry, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, Shanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai, China
- Shanghai Research Institute of Stomatology, Shanghai, China
| | - K L Lo
- Department of Preventive Dentistry, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, Shanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai, China
- Shanghai Research Institute of Stomatology, Shanghai, China
| | - A N Liman
- Department of Preventive Dentistry, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, Shanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai, China
- Shanghai Research Institute of Stomatology, Shanghai, China
| | - X P Feng
- Department of Preventive Dentistry, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, Shanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai, China
- Shanghai Research Institute of Stomatology, Shanghai, China
| | - W Ye
- Department of Preventive Dentistry, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, Shanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai, China
- Shanghai Research Institute of Stomatology, Shanghai, China
| |
Collapse
|
14
|
Luo SC, Wei SM, Luo XT, Yang QQ, Wong KH, Cheung PCK, Zhang BB. How probiotics, prebiotics, synbiotics, and postbiotics prevent dental caries: an oral microbiota perspective. NPJ Biofilms Microbiomes 2024; 10:14. [PMID: 38402294 PMCID: PMC10894247 DOI: 10.1038/s41522-024-00488-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 02/12/2024] [Indexed: 02/26/2024] Open
Abstract
Dental caries, a highly prevalent oral disease, impacts a significant portion of the global population. Conventional approaches that indiscriminately eradicate microbes disrupt the natural equilibrium of the oral microbiota. In contrast, biointervention strategies aim to restore this balance by introducing beneficial microorganisms or inhibiting cariogenic ones. Over the past three decades, microbial preparations have garnered considerable attention in dental research for the prevention and treatment of dental caries. However, unlike related pathologies in the gastrointestinal, vaginal, and respiratory tracts, dental caries occurs on hard tissues such as tooth enamel and is closely associated with localized acid overproduction facilitated by cariogenic biofilms. Therefore, it is insufficient to rely solely on previous mechanisms to delineate the role of microbial preparations in the oral cavity. A more comprehensive perspective should involve considering the concepts of cariogenic biofilms. This review elucidates the latest research progress, mechanisms of action, challenges, and future research directions regarding probiotics, prebiotics, synbiotics, and postbiotics for the prevention and treatment of dental caries, taking into account the unique pathogenic mechanisms of dental caries. With an enhanced understanding of oral microbiota, personalized microbial therapy will emerge as a critical future research trend.
Collapse
Affiliation(s)
- Si-Chen Luo
- Guangdong Provincial Key Laboratory of Marine Biology, Department of Biology, College of Science, Shantou University, Shantou, 515063, Guangdong, PR China
| | - Si-Min Wei
- Guangdong Provincial Key Laboratory of Marine Biology, Department of Biology, College of Science, Shantou University, Shantou, 515063, Guangdong, PR China
| | - Xin-Tao Luo
- Guangdong Provincial Key Laboratory of Marine Biology, Department of Biology, College of Science, Shantou University, Shantou, 515063, Guangdong, PR China
| | - Qiong-Qiong Yang
- Guangdong Provincial Key Laboratory of Marine Biology, Department of Biology, College of Science, Shantou University, Shantou, 515063, Guangdong, PR China
| | - Ka-Hing Wong
- Research Institute for Future Food, Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hong Kong, PR China
| | - Peter C K Cheung
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, PR China
| | - Bo-Bo Zhang
- Guangdong Provincial Key Laboratory of Marine Biology, Department of Biology, College of Science, Shantou University, Shantou, 515063, Guangdong, PR China.
| |
Collapse
|
15
|
Lopes PC, Gomes ATPC, Mendes K, Blanco L, Correia MJ. Unlocking the potential of probiotic administration in caries management: a systematic review. BMC Oral Health 2024; 24:216. [PMID: 38341538 PMCID: PMC10859023 DOI: 10.1186/s12903-024-03893-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 01/14/2024] [Indexed: 02/12/2024] Open
Abstract
BACKGROUND The use of prebiotics and/or probiotic bacteria with the potential to modulate the oral ecosystem may play an important role in the prevention and management of dental caries. To assess the evidence of the potential of pre/probiotics both in the prevention and treatment of dental caries, we focused on the PICO question "In individuals with caries, after probiotic administration, is there an improvement in outcomes directly related to caries risk and development?". METHODS An extensive systematic search was conducted in electronic databases PubMed, Web of Science, Scopus and Cochrane, to identify articles with relevant data. This systematic review included trials performed in Humans; published in English; including the observation of patients with caries, with clear indication of the probiotic used and measuring the outcomes directly involved with the cariogenic process, including the quantification of bacteria with cariogenic potential. To evaluate the methodological quality of the studies, the critical assessment tool from the Joanna Briggs Institute was used. RESULTS Eight hundred and fifty articles, potentially relevant, were identified. Following PRISMA guidelines 14 articles were included in this systematic review. Outcomes such as reduction of cariogenic microorganism counts, salivary pH, buffer capacity, and caries activity were assessed. The probiotic most often referred with beneficial results in dental caries outcomes is Lacticaseibacillus rhamnosus. Regarding the most used administration vehicle, in studies with positive effects on the caries management, probiotic supplemented milk could be considered the best administration vehicle. CONCLUSIONS Evidence suggests a beneficial effect of probiotic supplemented milk (Lacticaseibacillus rhamnosus) as an adjuvant for caries prevention and management. However, comparable evidence is scarce and better designed and comparable studies are needed.
Collapse
Affiliation(s)
- Pedro C Lopes
- Faculty of Dental Medicine, Center for Interdisciplinary Research in Health, Universidade Católica Portuguesa, Viseu, 3504-505, Portugal
| | - Ana T P C Gomes
- Faculty of Dental Medicine, Center for Interdisciplinary Research in Health, Universidade Católica Portuguesa, Viseu, 3504-505, Portugal
| | - Karina Mendes
- Faculty of Dental Medicine, Center for Interdisciplinary Research in Health, Universidade Católica Portuguesa, Viseu, 3504-505, Portugal
| | - Letícia Blanco
- Department of Surgery, Universidad de Salamanca, Salamanca, Spain
| | - Maria J Correia
- Faculty of Dental Medicine, Center for Interdisciplinary Research in Health, Universidade Católica Portuguesa, Viseu, 3504-505, Portugal.
| |
Collapse
|
16
|
Zhang JS, Huang S, Chen Z, Chu CH, Takahashi N, Yu OY. Application of omics technologies in cariology research: A critical review with bibliometric analysis. J Dent 2024; 141:104801. [PMID: 38097035 DOI: 10.1016/j.jdent.2023.104801] [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: 09/19/2023] [Revised: 12/06/2023] [Accepted: 12/07/2023] [Indexed: 12/19/2023] Open
Abstract
OBJECTIVES To review the application of omics technologies in the field of cariology research and provide critical insights into the emerging opportunities and challenges. DATA & SOURCES Publications on the application of omics technologies in cariology research up to December 2022 were sourced from online databases, including PubMed, Web of Science and Scopus. Two independent reviewers assessed the relevance of the publications to the objective of this review. STUDY SELECTION Studies that employed omics technologies to investigate dental caries were selected from the initial pool of identified publications. A total of 922 publications with one or more omics technologies adopted were included for comprehensive bibliographic analysis. (Meta)genomics (676/922, 73 %) is the predominant omics technology applied for cariology research in the included studies. Other applied omics technologies are metabolomics (108/922, 12 %), proteomics (105/922, 11 %), and transcriptomics (76/922, 8 %). CONCLUSION This study identified an emerging trend in the application of multiple omics technologies in cariology research. Omics technologies possess significant potential in developing strategies for the detection, staging evaluation, risk assessment, prevention, and management of dental caries. Despite the numerous challenges that lie ahead, the integration of multi-omics data obtained from individual biological samples, in conjunction with artificial intelligence technology, may offer potential avenues for further exploration in caries research. CLINICAL SIGNIFICANCE This review presented a comprehensive overview of the application of omics technologies in cariology research and discussed the advantages and challenges of using these methods to detect, assess, predict, prevent, and treat dental caries. It contributes to steering research for improved understanding of dental caries and advancing clinical translation of cariology research outcomes.
Collapse
Affiliation(s)
| | - Shi Huang
- Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, PR China
| | - Zigui Chen
- Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, PR China; Department of Microbiology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, PR China
| | - Chun-Hung Chu
- Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, PR China
| | - Nobuhiro Takahashi
- Division of Oral Ecology and Biochemistry, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | - Ollie Yiru Yu
- Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, PR China.
| |
Collapse
|
17
|
Yu X, Devine D, Vernon J. Manipulating the diseased oral microbiome: the power of probiotics and prebiotics. J Oral Microbiol 2024; 16:2307416. [PMID: 38304119 PMCID: PMC10833113 DOI: 10.1080/20002297.2024.2307416] [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: 10/20/2023] [Accepted: 01/14/2024] [Indexed: 02/03/2024] Open
Abstract
Dental caries and periodontal disease are amongst the most prevalent global disorders. Their aetiology is rooted in microbial activity within the oral cavity, through the generation of detrimental metabolites and the instigation of potentially adverse host immune responses. Due to the increasing threat of antimicrobial resistance, alternative approaches to readdress the balance are necessary. Advances in sequencing technologies have established relationships between disease and oral dysbiosis, and commercial enterprises seek to identify probiotic and prebiotic formulations to tackle preventable oral disorders through colonisation with, or promotion of, beneficial microbes. It is the metabolic characteristics and immunomodulatory capabilities of resident species which underlie health status. Research emphasis on the metabolic environment of the oral cavity has elucidated relationships between commensal and pathogenic organisms, for example, the sequential metabolism of fermentable carbohydrates deemed central to acid production in cariogenicity. Therefore, a focus on the preservation of an ecological homeostasis in the oral environment may be the most appropriate approach to health conservation. In this review we discuss an ecological approach to the maintenance of a healthy oral environment and debate the potential use of probiotic and prebiotic supplementation, specifically targeted at sustaining oral niches to preserve the delicately balanced microbiome.
Collapse
Affiliation(s)
- X. Yu
- Division of Oral Biology, School of Dentistry, University of Leeds, Leeds, UK
| | - D.A. Devine
- Division of Oral Biology, School of Dentistry, University of Leeds, Leeds, UK
| | - J.J. Vernon
- Division of Oral Biology, School of Dentistry, University of Leeds, Leeds, UK
| |
Collapse
|
18
|
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
|
19
|
Goyal V, Damle S, Puranik MP, Nuvvula S, Kakanur M, Marwah N, Asokan S, Suprabha BS, Sreenivasan P, Wadgave U, Shyam S, Thakur D. Arginine: A New Paradigm in Preventive Oral Care. Int J Clin Pediatr Dent 2023; 16:698-706. [PMID: 38162235 PMCID: PMC10753110 DOI: 10.5005/jp-journals-10005-2693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2024] Open
Abstract
Dental caries is ubiquitous and one of the most prevalent oral diseases and the foremost cause of dental pain and poor quality of life (QoL). Fluoride is an effective caries preventive agent; however, despite its use, there remain some gaps in prevention of dental caries. Arginine, an amino acid, helps to maintain a noncariogenic plaque. It shows synergistic effects with fluoride in dental caries and can help nourish the tooth by enhancing the remineralization effect of fluoride. Supplementing fluoride dentifrices with arginine can bridge the gap in caries prevention. Several clinical studies with 1.5% arginine + fluoride provide evidence for its use in prevention of dental caries. This report throws light on the benefits of arginine in dental caries and guides on its use with fluoride. Recommendations given in the report will help in effective prevention of dental caries. How to cite this article Goyal V, Damle S, Puranik MP, et al. Arginine: A New Paradigm in Preventive Oral Care. Int J Clin Pediatr Dent 2023;16(5):698-706.
Collapse
Affiliation(s)
- Varinder Goyal
- Department of Pediatric and Preventive Dentistry, Guru Nanak Dev Dental College and Research Institute, Sunam, Punjab, India
| | - Satyawan Damle
- Department of Paediatric Dentistry, M.M. College of Dental Sciences and Research, Maharishi Markandeshwar University, Ambala, Haryana, India
| | - Manjunath P Puranik
- Department of Public Health Dentistry, Government Dental College and Research Institute, Bengaluru, Karnataka, India
| | - Sivakumar Nuvvula
- Department of Pediatric Dentistry, Narayana Dental College and Hospital, Nellore, Andhra Pradesh, India
| | - Madhu Kakanur
- Department of Pediatric and Preventive Dentistry, KLE Society's Institute of Dental Sciences, Bengaluru, Karnataka, India
| | - Nikhil Marwah
- Department of Pediatric and Preventive Dentistry, Mahatma Gandhi Dental College & Hospital, Jaipur, Rajasthan, India
| | - Sharath Asokan
- Department of Pediatric and Preventive Dentistry, KSR Institute of Dental Science and Research, Tiruchengode, Tamil Nadu, India
| | - Baranya S Suprabha
- Department of Pediatric and Preventive Dentistry, Manipal College of Dental Sciences, Manipal, Mangaluru, India
| | - Prathima Sreenivasan
- Department of Oral Medicine and Radiology, Kannur Dental College, Kannur, Kerala, India
| | - Umesh Wadgave
- Department of Public Health Dentistry, Wadgave's Dental Home, Bidar, Karnataka, India
| | - S Shyam
- Department of Public Health Dentistry, Meenakshi Ammal Dental College and Hospital, Meenakshi Academy of Higher Education and Research, Chennai, Tamil Nadu, India
| | - Dewakshi Thakur
- Department of Pediatric and Preventive Dentistry, Guru Nanak Dev Dental College and Research Institute, Sunam, Punjab, India
| |
Collapse
|
20
|
Demkovych A, Kalashnikov D, Hasiuk P, Zubchenko S, Vorobets A. The influence of microbiota on the development and course of inflammatory diseases of periodontal tissues. FRONTIERS IN ORAL HEALTH 2023; 4:1237448. [PMID: 37609105 PMCID: PMC10440822 DOI: 10.3389/froh.2023.1237448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 07/28/2023] [Indexed: 08/24/2023] Open
Abstract
An important feature of the functioning of the organs and tissues of the oral cavity is the fact that all processes that take place in it are carried out in the constant presence of various microorganisms that cause the development of pathological processes in the body or are associated with them. In the pathogenesis of chronic generalized periodontitis, dental plaque penetrates the bottom of the gingival sulcus, penetrating under the epithelium into the stroma of the connective tissue, causing its inflammation. Bacteria produce a number of toxic substances that have a toxic effect on surrounding tissues. Most bacteria produce chain fatty acids that inhibit chemotaxis of leukocytes and phagocytes. Anaerobes and spirochetes secrete a number of substances (propionic acid and indole) that are extremely toxic to most tissues. Inflammation in the periodontal tissues is caused by the microbiota of the dental plaque biofilm. As periodontitis develops, an increase in the number of P. gingivalis, P. intermedia and T. forsythia was found in it, more than 100 times. Therefore, the given data prove that in the development and course of the inflammatory process in the periodontal tissues, complex dysbiotic and tissue-cellular interactions are involved, the dynamic balance of which depends on its outcome.
Collapse
Affiliation(s)
- Andrii Demkovych
- Department of Orthopedic Dentistry, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| | - Dmytro Kalashnikov
- Department of Propaedeutics of Prosthetic Dentistry, Poltava State Medical University, Poltava, Ukraine
| | - Petro Hasiuk
- Department of Orthopedic Dentistry, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| | - Sergiy Zubchenko
- Department of Propaedeutics of Prosthetic Dentistry, Poltava State Medical University, Poltava, Ukraine
| | - Anna Vorobets
- Department of Orthopedic Dentistry, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| |
Collapse
|
21
|
Farva K, Sattar H, Ullah H, Raziq A, Mehmood MD, Tareen AK, Sultan IN, Zohra Q, Khan MW. Phenotypic Analysis, Molecular Characterization, and Antibiogram of Caries-Causing Bacteria Isolated from Dental Patients. Microorganisms 2023; 11:1952. [PMID: 37630520 PMCID: PMC10457851 DOI: 10.3390/microorganisms11081952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/23/2023] [Accepted: 07/26/2023] [Indexed: 08/27/2023] Open
Abstract
Dental caries is a biofilm-mediated, sugar-driven, multifactorial, dynamic disease that results in the phasic demineralization and remineralization of dental hard tissues. Despite scientific advances in cariology, dental caries remains a severe global concern. The aim of this study was to determine the optimization of microbial and molecular techniques for the detection of cariogenic pathogens in dental caries patients, the prevalence of cariogenic bacteria on the basis of socioeconomic, climatological, and hygienic factors, and in vitro evaluation of the antimicrobial activity of selected synthetic antibiotics and herbal extracts. In this study, oral samples were collected from 900 patients for bacterial strain screening on a biochemical and molecular basis. Plant extracts, such as ginger, garlic, neem, tulsi, amla, and aloe vera, were used to check the antimicrobial activity against the isolated strains. Synthetic antimicrobial agents, such as penicillin, amoxicillin, erythromycin, clindamycin, metronidazole, doxycycline, ceftazidime, levofloxacin, and ciprofloxacin, were also used to access the antimicrobial activity. Among 900 patients, 63% were males and 37% were females, patients aged between 36 and 58 (45.7%) years were prone to disease, and the most common symptom was toothache (61%). For oral diseases, 21% used herbs, 36% used antibiotics, and 48% were self-medicated, owing to sweets consumption (60.66%) and fizzy drinks and fast food (51.56%). Staphylococcus mutans (29.11%) and Streptococcus sobrinus (28.11%) were found as the most abundant strains. Seven bacterial strains were successfully screened and predicted to be closely related to genera S. sobrinus, S. mutans, Actinomyces naeslundii, Lactobacillus acidophilus, Eubacterium nodatum, Propionibacterium acidifaciens, and Treponema Pallidum. Among plant extracts, the maximum zone of inhibition was recorded by ginger (22.36 mm) and amla (20.01 mm), while among synthetic antibiotics, ciprofloxacin and levofloxacin were most effective against all microbes. This study concluded that phyto extracts of ginger and amla were considered suitable alternatives to synthetic antibiotics to treat dental diseases.
Collapse
Affiliation(s)
- Khushbu Farva
- Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore 54000, Pakistan
| | - Huma Sattar
- Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore 54000, Pakistan
| | - Hayat Ullah
- Metabolic Engineering Lab, Department of Biological Engineering, Utah State University, Logan, UT 84322, USA
| | - Abdur Raziq
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, School of Bioengineering, Dalian University of Technology, Dalian 116024, China
| | - Muhammad Danish Mehmood
- Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore 54000, Pakistan
| | - Afrasiab Khan Tareen
- Department of Biotechnology, Balochistan University of Information Technology Engineering and Management Sciences, Quetta 87300, Pakistan
| | - Imrana Niaz Sultan
- Department of Biotechnology, Balochistan University of Information Technology Engineering and Management Sciences, Quetta 87300, Pakistan
| | - Quratulaain Zohra
- Department of Biotechnology, Project of Sahara for Life Trust, The Sahara College Narowal, Punjab 51601, Pakistan
| | - Muhammad Waseem Khan
- Department of Biotechnology, Balochistan University of Information Technology Engineering and Management Sciences, Quetta 87300, Pakistan
| |
Collapse
|
22
|
Basilicata M, Pieri M, Marrone G, Nicolai E, Di Lauro M, Paolino V, Tomassetti F, Vivarini I, Bollero P, Bernardini S, Noce A. Saliva as Biomarker for Oral and Chronic Degenerative Non-Communicable Diseases. Metabolites 2023; 13:889. [PMID: 37623833 PMCID: PMC10456419 DOI: 10.3390/metabo13080889] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 07/20/2023] [Accepted: 07/21/2023] [Indexed: 08/26/2023] Open
Abstract
Saliva is a very complex fluid and it is essential to maintain several physiological processes and functions, including oral health, taste, digestion and immunological defenses. Saliva composition and the oral microbiome can be influenced by several factors, like diet and smoking habits, and their alteration can represent an important access point for pathogens and, thus, for systemic illness onset. In this review, we explore the potentiality of saliva as a new tool for the early detection of some pathological conditions, such as oral diseases, chronic degenerative non-communicable diseases, among these chronic kidney disease (CKD). We also examined the possible correlation between oral and systemic diseases and oral and gut microbiota dysbiosis. In particular, we deeply analyzed the relationship between oral diseases and CKD. In this context, some salivary parameters can represent a new device to detect either oral or systemic pathologies. Moreover, the positive modulation of oral and gut microbiota induced by prebiotics, postbiotics, or symbiotics could represent a new possible adjuvant therapy in the clinical management of oral diseases and CKD.
Collapse
Affiliation(s)
- Michele Basilicata
- UOSD Special Care Dentistry, Policlinico Tor Vergata, 00133 Rome, Italy
- Department of Experimental Medicine and Surgery, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Massimo Pieri
- Department of Experimental Medicine and Surgery, University of Rome Tor Vergata, 00133 Rome, Italy
- Department of Laboratory Medicine, “Tor Vergata” University Hospital, Viale Oxford 81, 00133 Rome, Italy
| | - Giulia Marrone
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Eleonora Nicolai
- Department of Experimental Medicine and Surgery, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Manuela Di Lauro
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Vincenza Paolino
- UOSD Special Care Dentistry, Policlinico Tor Vergata, 00133 Rome, Italy
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Flaminia Tomassetti
- Department of Experimental Medicine and Surgery, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Ilaria Vivarini
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Patrizio Bollero
- UOSD Special Care Dentistry, Policlinico Tor Vergata, 00133 Rome, Italy
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Sergio Bernardini
- Department of Experimental Medicine and Surgery, University of Rome Tor Vergata, 00133 Rome, Italy
- Department of Laboratory Medicine, “Tor Vergata” University Hospital, Viale Oxford 81, 00133 Rome, Italy
| | - Annalisa Noce
- Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
- UOSD Nephrology and Dialysis, Policlinico Tor Vergata, 00133 Rome, Italy
| |
Collapse
|
23
|
Berger V, Green Buzhor M, Evstafeva D, Mügeli L, Leroux JC. 3D printing of a controlled urea delivery device for the prevention of tooth decay. Int J Pharm 2023; 631:122528. [PMID: 36563799 DOI: 10.1016/j.ijpharm.2022.122528] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/15/2022] [Accepted: 12/17/2022] [Indexed: 12/24/2022]
Abstract
Dental caries is one of the most widespread chronic infectious diseases in the world. It is mainly caused by the production of acid in the biofilm from the bacterial metabolism of carbohydrates. Nowadays, the prevention of caries is mainly based on the use of topical formulations containing fluoride. However, effective fluoride supplementation may not be sufficient in high-risk individuals, leading to the exploration of alternative strategies such as the neutralization of acid in the oral cavity. Urea is hydrolyzed into ammonia by oral bacteria, leading to a local alkalization that may counteract tooth decay. Herein, we report the fabrication of 3D printed personalized dental trays with a local and prolonged release of urea. Composite filaments with tunable urea release kinetics were produced by hot melt extrusion of poly(ε-caprolactone) and poly(vinyl alcohol) or poly(ethylene glycol) blends mixed with urea. The filaments were further used to 3D print by fused deposition modeling objects capable of releasing urea in a sustained and spatially controlled manner. In vitro studies performed in the presence of Streptococcus salivarius demonstrated the ability of urea released from a 3D printed model toothguards to reduce the pH drop induced by carbohydrates. This study showed the potential of urea-loaded devices to reduce cariogenic acidification of the environment surrounding the enamel by delivering urea directly to the tooth surface.
Collapse
Affiliation(s)
- Valentine Berger
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Vladimir-Prelog-Weg 3, 8093 Zurich, Switzerland
| | - Marina Green Buzhor
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Vladimir-Prelog-Weg 3, 8093 Zurich, Switzerland
| | - Diana Evstafeva
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Vladimir-Prelog-Weg 3, 8093 Zurich, Switzerland
| | - Lena Mügeli
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Vladimir-Prelog-Weg 3, 8093 Zurich, Switzerland
| | - Jean-Christophe Leroux
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Vladimir-Prelog-Weg 3, 8093 Zurich, Switzerland.
| |
Collapse
|
24
|
Tonelli A, Lumngwena EN, Ntusi NAB. The oral microbiome in the pathophysiology of cardiovascular disease. Nat Rev Cardiol 2023; 20:386-403. [PMID: 36624275 DOI: 10.1038/s41569-022-00825-3] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/01/2022] [Indexed: 01/11/2023]
Abstract
Despite advances in our understanding of the pathophysiology of many cardiovascular diseases (CVDs) and expansion of available therapies, the global burden of CVD-associated morbidity and mortality remains unacceptably high. Important gaps remain in our understanding of the mechanisms of CVD and determinants of disease progression. In the past decade, much research has been conducted on the human microbiome and its potential role in modulating CVD. With the advent of high-throughput technologies and multiomics analyses, the complex and dynamic relationship between the microbiota, their 'theatre of activity' and the host is gradually being elucidated. The relationship between the gut microbiome and CVD is well established. Much less is known about the role of disruption (dysbiosis) of the oral microbiome; however, interest in the field is growing, as is the body of literature from basic science and animal and human investigations. In this Review, we examine the link between the oral microbiome and CVD, specifically coronary artery disease, stroke, peripheral artery disease, heart failure, infective endocarditis and rheumatic heart disease. We discuss the various mechanisms by which oral dysbiosis contributes to CVD pathogenesis and potential strategies for prevention and treatment.
Collapse
Affiliation(s)
- Andrea Tonelli
- Division of Cardiology, Department of Medicine, University of Cape Town and Groote Schuur Hospital, Cape Town, South Africa.,Cardiovascular Research Unit, Christiaan Barnard Division of Cardiothoracic Surgery, Department of Surgery, University of Cape Town and Groote Schuur Hospital, Cape Town, South Africa.,Cape Heart Institute, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.,Extramural Research Unit on the Intersection of Noncommunicable Diseases and Infectious Disease, South African Medical Research Council, Cape Town, South Africa
| | - Evelyn N Lumngwena
- Cape Heart Institute, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.,School of Clinical Medicine, Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa.,Centre for the Study of Emerging and Re-emerging Infections, Institute for Medical Research and Medicinal Plant Studies, Ministry of Scientific Research and Innovation, Yaoundé, Cameroon
| | - Ntobeko A B Ntusi
- Division of Cardiology, Department of Medicine, University of Cape Town and Groote Schuur Hospital, Cape Town, South Africa. .,Cape Heart Institute, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa. .,Extramural Research Unit on the Intersection of Noncommunicable Diseases and Infectious Disease, South African Medical Research Council, Cape Town, South Africa. .,Cape Universities Body Imaging Centre, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa. .,Wellcome Centre for Infectious Disease Research, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.
| |
Collapse
|
25
|
Wang X, Li J, Zhang S, Zhou W, Zhang L, Huang X. pH-activated antibiofilm strategies for controlling dental caries. Front Cell Infect Microbiol 2023; 13:1130506. [PMID: 36949812 PMCID: PMC10025512 DOI: 10.3389/fcimb.2023.1130506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 02/20/2023] [Indexed: 03/08/2023] Open
Abstract
Dental biofilms are highly assembled microbial communities surrounded by an extracellular matrix, which protects the resident microbes. The microbes, including commensal bacteria and opportunistic pathogens, coexist with each other to maintain relative balance under healthy conditions. However, under hostile conditions such as sugar intake and poor oral care, biofilms can generate excessive acids. Prolonged low pH in biofilm increases proportions of acidogenic and aciduric microbes, which breaks the ecological equilibrium and finally causes dental caries. Given the complexity of oral microenvironment, controlling the acidic biofilms using antimicrobials that are activated at low pH could be a desirable approach to control dental caries. Therefore, recent researches have focused on designing novel kinds of pH-activated strategies, including pH-responsive antimicrobial agents and pH-sensitive drug delivery systems. These agents exert antibacterial properties only under low pH conditions, so they are able to disrupt acidic biofilms without breaking the neutral microenvironment and biodiversity in the mouth. The mechanisms of low pH activation are mainly based on protonation and deprotonation reactions, acids labile linkages, and H+-triggered reactive oxygen species production. This review summarized pH-activated antibiofilm strategies to control dental caries, concentrating on their effect, mechanisms of action, and biocompatibility, as well as the limitation of current research and the prospects for future study.
Collapse
Affiliation(s)
- Xiuqing Wang
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Jingling Li
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Shujun Zhang
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Wen Zhou
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Linglin Zhang
- State Key Laboratory of Oral Diseases, Department of Cariology and Endodontics, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xiaojing Huang
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- *Correspondence: Xiaojing Huang,
| |
Collapse
|
26
|
Bijle MN, Abdalla MM, Chu CH, Yiu CKY. Synbiotic-fluoride synergism on enamel remineralization, cytotoxicity and genotoxicity. J Dent 2023; 128:104356. [PMID: 36370897 DOI: 10.1016/j.jdent.2022.104356] [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: 07/13/2022] [Revised: 11/01/2022] [Accepted: 11/04/2022] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVE(S) The objectives of the present study were to examine the - a) enamel remineralization potential of synbiotic-fluoride (SF) therapy using a multi-species bacterial pH-cycling model; and b) cytotoxic and genotoxic effects of SF therapy extracts. MATERIALS AND METHODS The SF therapy group comprised of 2% arginine (Arg), 0.2% NaF, and a probiotic Lactobacillus rhamnosus GG (LRG). The intervention groups studied were: 1) No treatment; 2) 2% Arg; 3) 0.2% NaF; 4) LRG; 5) 2% Arg+0.2% NaF; 6) 2% Arg+LRG; 7) 0.2% NaF+LRG; and 8) 2% Arg+0.2% NaF+LRG (SF therapy). The enamel remineralization potential of SF therapy was investigated under cariogenic biofilm challenge; while the cytotoxicity and genotoxicity of SF therapy extracts were examined on HGF-1 and Chinese hamster fibroblast V79, respectively. To determine the remineralization effect, the specimens were subjected to mineral density (MD) assessment using micro-CT, Ca/P molar ratio with SEM-EDX, and enamel fluoride uptake (EFU) estimates. The HGF-1 proliferation assessment was quantified using MTT/CCK-8 assays with qualitative analysis by nuclei staining Hoechst-based fluorescence imaging. The genotoxicity was determined by micronuclei formation test. RESULTS Mineral gain and %remineralization derived from MD assessment for the SF therapy were significantly higher than the other groups (p<0.05). The %ΔCa/P for the SF and 2% Arg+0.2% NaF were significantly higher than the other groups (p<0.05). The SF and 2% Arg+0.2% NaF groups had the highest EFU compared to the other groups (p<0.05). No significant difference in the %viable HGF-1 cells were observed between the treatment interventions and no treatment group (p>0.05). Compared to the EMS-positive control, the micronuclei formation for all the intervention groups was significantly lower (p<0.05), with no significant difference among the treatment groups (p>0.05). CONCLUSION The SF therapy enhanced enamel remineralization with no biocompatibility concerns. CLINICAL SIGNIFICANCE With the enhanced enamel remineralization potential discerned in the present study, the SF therapy can be used as a promising caries-preventive agent targeted for high caries-risk individuals.
Collapse
Affiliation(s)
- Mohammed Nadeem Bijle
- 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
- Paediatric Dentistry, Faculty of Dentistry, The University of Hong Kong, Hong Kong; Dental Biomaterials, Faculty of Dental Medicine Al-Azhar University, Cairo, Egypt.
| | - Chun Hung Chu
- Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong.
| | | |
Collapse
|
27
|
Kunath BJ, Hickl O, Queirós P, Martin-Gallausiaux C, Lebrun LA, Halder R, Laczny CC, Schmidt TSB, Hayward MR, Becher D, Heintz-Buschart A, de Beaufort C, Bork P, May P, Wilmes P. Alterations of oral microbiota and impact on the gut microbiome in type 1 diabetes mellitus revealed by integrated multi-omic analyses. MICROBIOME 2022; 10:243. [PMID: 36578059 PMCID: PMC9795701 DOI: 10.1186/s40168-022-01435-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 10/04/2022] [Indexed: 05/29/2023]
Abstract
BACKGROUND Alterations to the gut microbiome have been linked to multiple chronic diseases. However, the drivers of such changes remain largely unknown. The oral cavity acts as a major route of exposure to exogenous factors including pathogens, and processes therein may affect the communities in the subsequent compartments of the gastrointestinal tract. Here, we perform strain-resolved, integrated meta-genomic, transcriptomic, and proteomic analyses of paired saliva and stool samples collected from 35 individuals from eight families with multiple cases of type 1 diabetes mellitus (T1DM). RESULTS We identified distinct oral microbiota mostly reflecting competition between streptococcal species. More specifically, we found a decreased abundance of the commensal Streptococcus salivarius in the oral cavity of T1DM individuals, which is linked to its apparent competition with the pathobiont Streptococcus mutans. The decrease in S. salivarius in the oral cavity was also associated with its decrease in the gut as well as higher abundances in facultative anaerobes including Enterobacteria. In addition, we found evidence of gut inflammation in T1DM as reflected in the expression profiles of the Enterobacteria as well as in the human gut proteome. Finally, we were able to follow transmitted strain-variants from the oral cavity to the gut at the individual omic levels, highlighting not only the transfer, but also the activity of the transmitted taxa along the gastrointestinal tract. CONCLUSIONS Alterations of the oral microbiome in the context of T1DM impact the microbial communities in the lower gut, in particular through the reduction of "mouth-to-gut" transfer of Streptococcus salivarius. Our results indicate that the observed oral-cavity-driven gut microbiome changes may contribute towards the inflammatory processes involved in T1DM. Through the integration of multi-omic analyses, we resolve strain-variant "mouth-to-gut" transfer in a disease context. Video Abstract.
Collapse
Affiliation(s)
- B J Kunath
- Luxembourg Centre for Systems Biomedicine, Esch-sur-Alzette, Luxembourg.
| | - O Hickl
- Luxembourg Centre for Systems Biomedicine, Esch-sur-Alzette, Luxembourg
| | - P Queirós
- Luxembourg Centre for Systems Biomedicine, Esch-sur-Alzette, Luxembourg
| | | | - L A Lebrun
- Luxembourg Centre for Systems Biomedicine, Esch-sur-Alzette, Luxembourg
| | - R Halder
- Luxembourg Centre for Systems Biomedicine, Esch-sur-Alzette, Luxembourg
| | - C C Laczny
- Luxembourg Centre for Systems Biomedicine, Esch-sur-Alzette, Luxembourg
| | - T S B Schmidt
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - M R Hayward
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, 02139, USA
| | - D Becher
- Institute of Microbiology, University of Greifswald, Greifswald, Germany
| | - A Heintz-Buschart
- Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, Netherlands
| | - C de Beaufort
- Luxembourg Centre for Systems Biomedicine, Esch-sur-Alzette, Luxembourg
- Clinique Pédiatrique, Centre Hospitalier de Luxembourg, Luxembourg, Luxembourg
| | - P Bork
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- Max Delbrück Centre for Molecular Medicine, Berlin, Germany
- Yonsei Frontier Lab (YFL), Yonsei University, Seoul, 03722, South Korea
- Department of Bioinformatics, Biocenter, University of Würzburg, Würzburg, Germany
| | - P May
- Luxembourg Centre for Systems Biomedicine, Esch-sur-Alzette, Luxembourg
| | - P Wilmes
- Luxembourg Centre for Systems Biomedicine, Esch-sur-Alzette, Luxembourg.
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Belvaux, Luxembourg.
| |
Collapse
|
28
|
Rostami N, Shields RC, Serrage HJ, Lawler C, Brittan JL, Yassin S, Ahmed H, Treumann A, Thompson P, Waldron KJ, Nobbs AH, Jakubovics NS. Interspecies competition in oral biofilms mediated by Streptococcus gordonii extracellular deoxyribonuclease SsnA. NPJ Biofilms Microbiomes 2022; 8:96. [PMID: 36509765 PMCID: PMC9744736 DOI: 10.1038/s41522-022-00359-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 11/21/2022] [Indexed: 12/14/2022] Open
Abstract
Extracellular DNA (eDNA) is a key component of many microbial biofilms including dental plaque. However, the roles of extracellular deoxyribonuclease (DNase) enzymes within biofilms are poorly understood. Streptococcus gordonii is a pioneer colonizer of dental plaque. Here, we identified and characterised SsnA, a cell wall-associated protein responsible for extracellular DNase activity of S. gordonii. The SsnA-mediated extracellular DNase activity of S. gordonii was suppressed following growth in sugars. SsnA was purified as a recombinant protein and shown to be inactive below pH 6.5. SsnA inhibited biofilm formation by Streptococcus mutans in a pH-dependent manner. Further, SsnA inhibited the growth of oral microcosm biofilms in human saliva. However, inhibition was ameliorated by the addition of sucrose. Together, these data indicate that S. gordonii SsnA plays a key role in interspecies competition within oral biofilms. Acidification of the medium through sugar catabolism could be a strategy for cariogenic species such as S. mutans to prevent SsnA-mediated exclusion from biofilms.
Collapse
Affiliation(s)
- Nadia Rostami
- grid.1006.70000 0001 0462 7212School of Dental Sciences, Faculty of Medical Sciences, Newcastle University, Newcastle, UK
| | - Robert C. Shields
- grid.1006.70000 0001 0462 7212School of Dental Sciences, Faculty of Medical Sciences, Newcastle University, Newcastle, UK ,grid.252381.f0000 0001 2169 5989Department of Biological Sciences, Arkansas State University, Jonesboro, AR USA
| | - Hannah J. Serrage
- grid.5337.20000 0004 1936 7603Bristol Dental School, University of Bristol, Bristol, UK
| | - Catherine Lawler
- grid.5337.20000 0004 1936 7603Bristol Dental School, University of Bristol, Bristol, UK
| | - Jane L. Brittan
- grid.5337.20000 0004 1936 7603Bristol Dental School, University of Bristol, Bristol, UK
| | - Sufian Yassin
- grid.1006.70000 0001 0462 7212School of Dental Sciences, Faculty of Medical Sciences, Newcastle University, Newcastle, UK ,grid.265892.20000000106344187Department of Restorative Sciences, University of Alabama at Birmingham, Birmingham, AL USA
| | - Halah Ahmed
- grid.1006.70000 0001 0462 7212School of Dental Sciences, Faculty of Medical Sciences, Newcastle University, Newcastle, UK
| | - Achim Treumann
- grid.1006.70000 0001 0462 7212Protein and Proteome Analysis Facility, Faculty of Medical Sciences, Newcastle University, Newcastle, UK ,KBI Biopharma BV, Leuven, Belgium
| | - Paul Thompson
- grid.1006.70000 0001 0462 7212Protein and Proteome Analysis Facility, Faculty of Medical Sciences, Newcastle University, Newcastle, UK
| | - Kevin J. Waldron
- grid.1006.70000 0001 0462 7212Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle, UK
| | - Angela H. Nobbs
- grid.5337.20000 0004 1936 7603Bristol Dental School, University of Bristol, Bristol, UK
| | - Nicholas S. Jakubovics
- grid.1006.70000 0001 0462 7212School of Dental Sciences, Faculty of Medical Sciences, Newcastle University, Newcastle, UK
| |
Collapse
|
29
|
Balaganesh S, Balasubramaniam A, Indiran MA, Rathinavelu PK, Kumar MPS. Determination of salivary cortisol and salivary pH level in gaming teenagers - A cross-sectional study. J Oral Biol Craniofac Res 2022; 12:838-842. [PMID: 36186268 DOI: 10.1016/j.jobcr.2022.09.005] [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: 04/26/2021] [Revised: 01/10/2022] [Accepted: 09/06/2022] [Indexed: 10/31/2022] Open
Abstract
Introduction Internet gaming disorder and cybervictimization has a disproportionate increase during the COVID 19 pandemic. Teenagers and adolescents have high risk for addiction to online gaming. Online gaming increases the stress level of the users affecting the general and the oral health. The study aims to determine the salivary cortisol and salivary pH among the gaming teenagers. Materials and methods The cross-sectional study recruited 45 gaming teenagers with the mean age of 15.5 years. The participants were stratified according to the type of online gaming played as mind, aggressive and betting gaming. Their salivary pH and salivary cortisol were assessed. The collected data was analyzed using SPSS version 23. Results The mean salivary pH was found to be less among females playing aggressive games (7.11 ± 0.30) and males playing mind games (7.24 ± 0.16). Salivary cortisol level was found to be statistically high in males (6.66 ± 1.33) and females (7.82 ± 0.66) playing aggressive games. The sub-group analysis of salivary pH and cortisol for gender showed a significant difference in the mean salivary cortisol level (p < 0.05) with no difference in the salivary pH (p > 0.05). Based on the number of hours spent in online gaming, there was a significant increase in the mean salivary cortisol level among the participants playing >3 h (p < 0.05). Conclusion Salivary cortisol is high among individuals playing aggressive games compared to mind and betting games. Also, salivary cortisol is high in females compared to males with no change in salivary pH.
Collapse
Affiliation(s)
- Sarika Balaganesh
- Department of Public Health Dentistry, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, No.162, Poonamallee High Road, Chennai, 600077, Tamil Nadu, India
| | - Arthi Balasubramaniam
- Department of Public Health Dentistry, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, No.162, Poonamallee High Road, Chennai, 600077, Tamil Nadu, India
| | - Meignana Arumugham Indiran
- Department of Public Health Dentistry, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, No.162, Poonamallee High Road, Chennai, 600077, Tamil Nadu, India
| | - Pradeep Kumar Rathinavelu
- Department of Public Health Dentistry, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, No.162, Poonamallee High Road, Chennai, 600077, Tamil Nadu, India
| | - M P Santhosh Kumar
- Department of Oral and Maxillofacial Surgery, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, No.162, Poonamallee High Road, Chennai, 600077, Tamil Nadu, India
| |
Collapse
|
30
|
Carda-Diéguez M, Moazzez R, Mira A. Functional changes in the oral microbiome after use of fluoride and arginine containing dentifrices: a metagenomic and metatranscriptomic study. MICROBIOME 2022; 10:159. [PMID: 36171634 PMCID: PMC9520947 DOI: 10.1186/s40168-022-01338-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 07/27/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND Tooth decay is one of the most prevalent diseases worldwide, and efficient tooth brushing with a fluoride-containing dentifrice is considered fundamental to caries prevention. Fluoride-containing dentifrices have been extensively studied in relation to enamel resistance to demineralization. Arginine (Arg) has also been proposed as a promising prebiotic to promote pH buffering through ammonia production. Here, we present the first metagenomic (DNA sequencing of the whole microbial community) and metatranscriptomic (RNAseq of the same community) analyses of human dental plaque to evaluate the effect of brushing with fluoride (Fl) and a Fl+Arg containing dentifrices on oral microbial composition and activity. Fifty-three patients were enrolled in a longitudinal clinical intervention study with two arms, including 26 caries-active and 27 caries-free adults. After a minimum 1-week washout period, dental plaque samples were collected at this post-washout baseline, 3 months after the use of a 1450-ppm fluoride dentifrice, and after 6 months of using a 1450-ppm fluoride with 1.5% arginine dentifrice. RESULTS There was a shift in both the composition and activity of the plaque microbiome after 3 months of brushing with the fluoride-containing toothpaste compared to the samples collected at the 1-week post-washout period, both for caries-active and caries-free sites. Although several caries-associated bacteria were reduced, there was also an increase in several health- and periodontitis-associated bacteria. Over 400 genes changed proportion in the metagenome, and between 180 and 300 genes changed their expression level depending on whether caries-free or caries-active sites were analyzed. The metagenome and metatranscriptome also changed after the subjects brushed with the Fl+Arg dentifrice. There was a further decrease of both caries- and periodontitis-associated organisms. In both caries-free and caries-active sites, a decrease of genes from the arginine biosynthesis pathway was also observed, in addition to an increase in the expression of genes associated with the arginine deiminase pathway, which catabolizes arginine into ammonia, thereby buffering acidic pH. Bacterial richness and diversity were not affected by either of the two treatments in the two arms of the study. CONCLUSIONS Our data demonstrate that long-term use of both assayed dentifrices changes the bacterial composition and functional profiles of human dental plaque towards a healthier microbial community, both in caries-free and caries-active sites. This observation was especially apparent for the Fl+Arg dentifrice. Thus, we conclude that the preventive benefits of tooth brushing go beyond the physical removal of dental plaque and that the active ingredients formulated within dentifrices have a positive effect not only on enamel chemistry but also on the metabolism of oral microbial populations. Video Abstract.
Collapse
Affiliation(s)
| | - Rebecca Moazzez
- Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London, UK
| | - Alex Mira
- Genomics and Health Department, FISABIO Institute, Valencia, Spain.
- Network of Epidemiology and Public Health, CIBERESP, Madrid, Spain.
| |
Collapse
|
31
|
Abstract
Streptococcus mutans is a primary cariogenic pathogen in humans. Arginine metabolism is required for bacterial growth. In S. mutans, however, the involvement of transcription factors in regulating arginine metabolism is unclear. The purpose of this study was to investigate the function and mechanism of ArgR family transcription factors in S. mutans. Here, we identified an ArgR (arginine repressor) family transcription factor named AhrC, which negatively regulates arginine biosynthesis and biofilm formation in S. mutans. The ahrC in-frame deletion strain exhibited slow growth and significantly increased intracellular arginine content. The strain overexpressing ahrC showed reduced intracellular arginine content, decreased biofilm biomass, reduced production of water-insoluble exopolysaccharides (EPS), and different biofilm structures. Furthermore, global gene expression profiles revealed differential expression levels of 233 genes in the ahrC-deficient strain, among which genes related to arginine biosynthesis (argJ, argB, argC, argD, argF, argG, argH) were significantly upregulated. In the ahrC overexpression strain, there are 89 differentially expressed genes, mostly related to arginine biosynthesis. The conserved DNA patterns bound by AhrC were identified by electrophoretic mobility shift assay (EMSA) and DNase I footprinting. In addition, the analysis of β-galactosidase activity showed that AhrC acted as a negative regulator. Taken together, our findings suggest that AhrC is an important transcription factor that regulates arginine biosynthesis gene expression and biofilm formation in S. mutans. These findings add new aspects to the complexity of regulating the expression of genes involved in arginine biosynthesis and biofilm formation in S. mutans. IMPORTANCE Arginine metabolism is essential for bacterial growth. The regulation of intracellular arginine metabolism in Streptococcus mutans, one of the major pathogens of dental caries, is unclear. In this study, we found that the transcription factor AhrC can directly and negatively regulate the expression of N-acetyl-gamma-glutamyl-phosphate reductase (argC), thus regulating arginine biosynthesis in S. mutans. In addition, the ahrC overexpression strain exhibited a significant decrease in biofilm and water-insoluble extracellular polysaccharides (EPS). This study adds new support to our understanding of the regulation of intracellular arginine metabolism in S. mutans.
Collapse
|
32
|
Abstract
Fusobacterium nucleatum is a common constituent of the oral microbiota in both periodontal health and disease. Previously, we discovered ornithine cross-feeding between F. nucleatum and Streptococcus gordonii, where S. gordonii secretes ornithine via an arginine-ornithine antiporter (ArcD), which in turn supports the growth and biofilm development of F. nucleatum; however, broader metabolic aspects of F. nucleatum within polymicrobial communities and their impact on periodontal pathogenesis have not been addressed. Here, we show that when cocultured with S. gordonii, F. nucleatum increased amino acid availability to enhance the production of butyrate and putrescine, a polyamine produced by ornithine decarboxylation. Coculture with Veillonella parvula, another common inhabitant of the oral microbiota, also increased lysine availability, promoting cadaverine production by F. nucleatum. We confirmed that ArcD-dependent S. gordonii-excreted ornithine induces synergistic putrescine production, and mass spectrometry imaging revealed that this metabolic capability creates a putrescine-rich microenvironment on the surface of F. nucleatum biofilms. We further demonstrated that polyamines caused significant changes in the biofilm phenotype of a periodontal pathogen, Porphyromonas gingivalis, with putrescine accelerating the biofilm life cycle of maturation and dispersal. This phenomenon was also observed with putrescine derived from S. gordonii-F. nucleatum coculture. Lastly, analysis of plaque samples revealed cooccurrence of P. gingivalis with genetic modules for putrescine production by S. gordonii and F. nucleatum. Overall, our results highlight the ability of F. nucleatum to induce synergistic polyamine production within multispecies consortia and provide insight into how the trophic web in oral biofilm ecosystems can eventually shape disease-associated communities. IMPORTANCE Periodontitis is caused by a pathogenic shift in subgingival biofilm ecosystems, which is accompanied by alterations in microbiome composition and function, including changes in the metabolic activity of the biofilm, which comprises multiple commensals and pathogens. While Fusobacterium nucleatum is a common constituent of the supra- and subgingival biofilms, its metabolic integration within polymicrobial communities and the impact on periodontal pathogenesis are poorly understood. Here, we report that amino acids supplied by other commensal bacteria induce polyamine production by F. nucleatum, creating polyamine-rich microenvironments. Polyamines reportedly have diverse functions in bacterial physiology and possible involvement in periodontal pathogenesis. We show that the F. nucleatum-integrated trophic network yielding putrescine from arginine through ornithine accelerates the biofilm life cycle of Porphyromonas gingivalis, a periodontal pathogen, from the planktonic state through biofilm formation to dispersal. This work provides insight into how cooperative metabolism within oral biofilms can tip the balance toward periodontitis.
Collapse
|
33
|
Zhu J, Chu W, Luo J, Yang J, He L, Li J. Dental Materials for Oral Microbiota Dysbiosis: An Update. Front Cell Infect Microbiol 2022; 12:900918. [PMID: 35846759 PMCID: PMC9280126 DOI: 10.3389/fcimb.2022.900918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 06/07/2022] [Indexed: 11/21/2022] Open
Abstract
The balance or dysbiosis of the microbial community is a major factor in maintaining human health or causing disease. The unique microenvironment of the oral cavity provides optimal conditions for colonization and proliferation of microbiota, regulated through complex biological signaling systems and interactions with the host. Once the oral microbiota is out of balance, microorganisms produce virulence factors and metabolites, which will cause dental caries, periodontal disease, etc. Microbial metabolism and host immune response change the local microenvironment in turn and further promote the excessive proliferation of dominant microbes in dysbiosis. As the product of interdisciplinary development of materials science, stomatology, and biomedical engineering, oral biomaterials are playing an increasingly important role in regulating the balance of the oral microbiome and treating oral diseases. In this perspective, we discuss the mechanisms underlying the pathogenesis of oral microbiota dysbiosis and introduce emerging materials focusing on oral microbiota dysbiosis in recent years, including inorganic materials, organic materials, and some biomolecules. In addition, the limitations of the current study and possible research trends are also summarized. It is hoped that this review can provide reference and enlightenment for subsequent research on effective treatment strategies for diseases related to oral microbiota dysbiosis.
Collapse
Affiliation(s)
- Jieyu Zhu
- State Key Laboratory of Oral Diseases, Department of Cariology and Endodontics, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Wenlin Chu
- State Key Laboratory of Oral Diseases, Department of Cariology and Endodontics, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, China
| | - Jun Luo
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, China
| | - Jiaojiao Yang
- State Key Laboratory of Oral Diseases, Department of Cariology and Endodontics, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- *Correspondence: Jiaojiao Yang, ; Libang He,
| | - Libang He
- State Key Laboratory of Oral Diseases, Department of Cariology and Endodontics, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- *Correspondence: Jiaojiao Yang, ; Libang He,
| | - Jiyao Li
- State Key Laboratory of Oral Diseases, Department of Cariology and Endodontics, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
34
|
Shi J, Wang Q, Ruan G, Chen Y, Zhao M, Shi D, Pan B, Xu Z, Zhang T, Wang F, Xu C. Efficacy of probiotics against dental caries in children: a systematic review and meta-analysis. Crit Rev Food Sci Nutr 2022; 63:9977-9994. [PMID: 35607893 DOI: 10.1080/10408398.2022.2077693] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Dental caries causes serious consequences and the financial burden of society especially in children with high morbidity rate. Here we carried out a meta-analysis to systematically evaluate the efficacy of probiotics against dental caries in children. Forty-three RCTs were eligible for this meta-analysis after searching the PubMed, Cochrane and Web of Science from the inception through October 2021. Pooled estimates demonstrated that treatment with probiotics significantly reduced noncavitated (dicdas2-6mft) (SMD = -0.18, 95% CI: -0.3 to -0.06, p = 0.002) and cavitated (dicdas5-6mft) carious lesions in children (SMD = -0.32, 95% CI: -0.5 to 0.14, p = 0.0004). Probiotics also reduced prevalence of noncavitated (dicdas2-6mft) carious lesions (RR = 0.8, 95% CI: 0.67 to-0.97, p = 0.02). Salivary Streptococcus mutans was declined after intervention (SMD = -1.17, 95% CI: -1.85 to -0.5, p = 0.0007), while Lactobacillus counts were upregulated (SMD = 1.19, 95% CI: 0.46-1.92, p = 0.001). However, no significant effects in total bacteria counts and salivary pH were observed. Our findings suggest that probiotics especially Lactobacillus could be a promising therapeutic strategy for clinical applications in children dental caries.
Collapse
Affiliation(s)
- Jiangmin Shi
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qinjian Wang
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Gaoyi Ruan
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yaoxuan Chen
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Min Zhao
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Dibang Shi
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Binhui Pan
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhihua Xu
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Tiaotiao Zhang
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Fangyan Wang
- Department of Pathophysiology, School of Basic Medicine Science, Wenzhou Medical University, Wenzhou, China
| | - Changlong Xu
- Department of Gastroenterology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
35
|
Culp DJ, Hull W, Schultz AC, Bryant AS, Lizarraga CA, Dupuis MR, Chakraborty B, Lee K, Burne RA. Testing of candidate probiotics to prevent dental caries induced by Streptococcus mutans in a mouse model. J Appl Microbiol 2022; 132:3853-3869. [PMID: 35262250 DOI: 10.1111/jam.15516] [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: 12/08/2021] [Revised: 02/22/2022] [Accepted: 03/01/2022] [Indexed: 11/29/2022]
Abstract
AIMS We evaluated two species of human oral commensal streptococci in protection against dental caries induced by Streptococcus mutans. METHODS AND RESULTS Candidate probiotics, Streptococcus sp. A12, Streptococcus sanguinis BCC23 and an arginine deiminase mutant of BCC23 (∆arcADS) were tested for their ability to reduce S. mutans-induced caries in an established mouse model. Mice were colonized with a probiotic, challenged with S. mutans, then intermittently reinoculated with a probiotic strain. Oral colonization of each strain and autochthonous bacteria was assessed by qPCR. Both BCC23 strains, but not A12, were associated with markedly reduced sulcal caries, persistently colonized mucosal and dental biofilms, and significantly lowered S. mutans counts. All three strains enhanced mucosal colonization of autochthonous bacteria. In a follow-up experiment, when S. mutans was established first, dental and mucosal colonization of S. mutans was unaltered by a subsequent challenge with either BCC23 strain. Results between BCC23 and BCC23 ∆arcADS were equivalent. CONCLUSIONS BCC23 is a potential probiotic to treat patients at high caries risk. Its effectiveness is independent of ADS activity, but initial dental cleaning to enhance establishment in dental biofilms may be required. SIGNIFICANCE AND IMPACT OF THE STUDY In vivo testing of candidate probiotics is highly informative, as effectiveness is not always reflected by genotype or in vitro behaviors.
Collapse
Affiliation(s)
- David J Culp
- University of Florida College of Dentistry, Department of Oral Biology, Gainesville, FL, USA
| | - William Hull
- University of Florida College of Dentistry, Department of Oral Biology, Gainesville, FL, USA
| | - Alexander C Schultz
- University of Florida College of Dentistry, Department of Oral Biology, Gainesville, FL, USA
| | - Ashley S Bryant
- University of Florida College of Dentistry, Department of Oral Biology, Gainesville, FL, USA
| | - Claudia A Lizarraga
- University of Florida College of Dentistry, Department of Oral Biology, Gainesville, FL, USA
| | - Madeline R Dupuis
- University of Florida College of Dentistry, Department of Oral Biology, Gainesville, FL, USA
| | - Brinta Chakraborty
- University of Florida College of Dentistry, Department of Oral Biology, Gainesville, FL, USA
| | - Kyulim Lee
- University of Florida College of Dentistry, Department of Oral Biology, Gainesville, FL, USA
| | - Robert A Burne
- University of Florida College of Dentistry, Department of Oral Biology, Gainesville, FL, USA
| |
Collapse
|
36
|
Zhang M, Liao Y, Tong X, Yan F. Novel urea derivative-loaded PLGA nanoparticles to inhibit caries-associated Streptococcus mutans. RSC Adv 2022; 12:4072-4080. [PMID: 35425421 PMCID: PMC8981093 DOI: 10.1039/d1ra09314b] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 01/25/2022] [Indexed: 12/19/2022] Open
Abstract
Caries is the most common chronic infectious disease in the human oral cavity and the existing anti-caries agents may lead to drug resistance and microecological imbalance. A novel urea derivative, 1,3-bis[3,5-bis(trifluoromethyl)phenyl]urea, has a potentially prominent antibacterial effect on cariogenic bacterial strain Streptococcus mutans UA159. In this study, we encapsulated the water-insoluble urea derivative in poly(lactic-co-glycolic acid) (PLGA) nanoparticles, performed physicochemical characterizations and explored its potential as a caries-preventive agent. The results showed that the drug-loaded PLGA nanoparticles exhibited satisfying surface morphology, particle size, size distribution and stability. With an optimized theoretical drug loading (10%), the drug-loaded PLGA nanoparticles exhibited negligible cytotoxicity against human oral squamous cell carcinoma cells. We noticed a biphasic drug release in vitro and the rate and cumulative release was higher in an acidic environment (pH 4.5) compared to a neutral environment (pH 7.4). The drug-loaded PLGA nanoparticles significantly inhibited the growth and lactic acid production of planktonic S. mutans as well as S. mutans biofilms. Our results indicate that the novel urea derivative-loaded PLGA nanoparticles serve as a promising anti-caries agent with remarkable pharmaceutical characteristics, low cytotoxicity, and satisfying antimicrobial effect.
Collapse
Affiliation(s)
- Mengyun Zhang
- Nanjing Stomatological Hospital, Medical School of Nanjing University Nanjing Jiangsu 210008 China
| | - Ying Liao
- Department of Pediatric Dentistry, Nanjing Stomatological Hospital, Medical School of Nanjing University Nanjing Jiangsu 210008 China
| | - Xin Tong
- Department of Oral Implantology, Nanjing Stomatological Hospital, Medical School of Nanjing University Nanjing Jiangsu 210008 China
| | - Fuhua Yan
- Department of Periodontology, Nanjing Stomatological Hospital, Medical School of Nanjing University Nanjing Jiangsu 210008 China
| |
Collapse
|
37
|
Ryvchin R, Dubinsky V, Rabinowitz K, Wasserberg N, Dotan I, Gophna U. Alteration in Urease-producing Bacteria in the Gut Microbiomes of Patients with Inflammatory Bowel Diseases. J Crohns Colitis 2021; 15:2066-2077. [PMID: 34111242 DOI: 10.1093/ecco-jcc/jjab101] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND AIMS Bacterial urease is a major virulence factor of human pathogens, and murine models have shown that it can contribute to the pathogenesis of inflammatory bowel diseases [IBD]. METHODS The distribution of urease-producing bacteria in IBD was assessed using public faecal metagenomic data from various cohorts, including non-IBD controls [n = 55], patients with Crohn's disease [n = 291] or ulcerative colitis [n = 214], and patients with a pouch [n = 53]. The ureA gene and the taxonomic markers gyrA, rpoB, and recA were used to estimate the percentage of urease producers in each sample. RESULTS Levels of urease producers in patients with IBD and non-IBD controls were comparable. In non-IBD controls and most IBD patients, urease producers were primarily acetate-producing genera such as Blautia and Ruminococcus. A shift in the type of the dominant urease producers towards Proteobacteria and Bacilli was observed in a subset of all IBD subtypes, which correlated with faecal calprotectin levels in one cohort. Some patients with IBD had no detectable urease producers. In patients with a pouch, the probiotic-associated species Streptococcus thermophilus was more common as a main urease producer than in other IBD phenotypes, and it generally did not co-occur with other Bacilli or with Proteobacteria. CONCLUSIONS Unlike all non-IBD controls, patients with IBD often showed a shift towards Bacilli or Proteobacteria or a complete loss of urease production. Probiotics containing the species S. thermophilus may have a protective effect against colonisation by undesirable urease-producing bacteria in a subset of patients with a pouch.
Collapse
Affiliation(s)
- Ron Ryvchin
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Vadim Dubinsky
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Keren Rabinowitz
- Division of Gastroenterology, Rabin Medical Center, Petah-Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Nir Wasserberg
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Division of Surgery, Rabin Medical Center, Petah-Tikva, Israel
| | - Iris Dotan
- Division of Gastroenterology, Rabin Medical Center, Petah-Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Uri Gophna
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
38
|
Development Strategy of Endodontic Filling Materials Based on Engineering and Medical Approaches. Processes (Basel) 2021. [DOI: 10.3390/pr9112014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
This article is a literature review aimed at presenting the general state of knowledge in manufacturing engineering and materials engineering to develop engineering materials applied for endodontic treatment as filling materials. Particular attention was paid to theoretical analyses concerning the selection of methods for developing and obturating root canals and discussing the results of experimental studies available in the literature. These activities aimed to compare the importance of the most commonly used endodontic filling materials based on gutta-percha or polymeric polyester materials, commonly known as resilon, respectively. The motivation to take up this complex, multi-faceted topic in this paper is the extent of caries, periodontal disease, and other oral diseases in 3 to 5 billion people, often affecting toothlessness and contributing to an increase in the index of disability-adjusted life years (number). Endodontics is an important element of the authors’ concept of Dentistry Sustainable Development (DSD) > 2020. The principles of qualifying patients for endodontic treatment are discussed. The introduction of rotary tools, especially manufactured from Nitinol alloy, to develop root canals and the latest thermohydraulic and condensation techniques for obturation guarantee progress in endodontics. The “Digital Twins” methodology was used, rooted in the idea of Industry 4.0 and the resulting idea of Dentistry 4.0, as well as knowledge management methods, to perform experimental research in virtual space, concerning methods of developing and obturating the root canal and assessing the tightness of fillings. Microscopic visualization methods were also used. Significant factors determining the effectiveness of endodontic obturation are the selection of the filling material and the appropriate obturation method. The generalized dendrological matrix of endodontic filling materials considers the criteria of mechanical strength influencing the potential root fracture and the quality of root canal filling. The results of the SWOT point analysis (strengths and weaknesses, opportunities, threats) were also compared. For both filling materials, the weaknesses are much less than the strengths, while the threats are slightly less than the opportunities for the gutta-percha-based material, while for resilon the opportunities are much smaller than the threats. It requires the application of an appropriate development strategy, i.e., MAXI-MAXI in the case of a filling material based on gutta-percha and MAXI-MINI in the case of resilon. Therefore, the analysis of these experimental data does not indicate the real competitiveness of resilon for the gutta-percha-based material. This material deservedly maintains its strong position as the “Gold Standard of Endodontics”.
Collapse
|
39
|
Spontaneous Mutants of Streptococcus sanguinis with Defects in the Glucose-Phosphotransferase System Show Enhanced Post-Exponential-Phase Fitness. J Bacteriol 2021; 203:e0037521. [PMID: 34460310 DOI: 10.1128/jb.00375-21] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Genetic truncations in a gene encoding a putative glucose-phosphotransferase system (PTS) protein (manL, EIIABMan) were identified in subpopulations of two separate laboratory stocks of Streptococcus sanguinis SK36; the mutants had reduced PTS activities on glucose and other monosaccharides. To understand the emergence of these mutants, we engineered deletion mutants of manL and showed that the ManL-deficient strain had improved bacterial viability in the stationary phase and was better able to inhibit the growth of the dental caries pathogen Streptococcus mutans. Transcriptional analysis and biochemical assays suggested that the manL mutant underwent reprograming of central carbon metabolism that directed pyruvate away from production of lactate, increasing production of hydrogen peroxide (H2O2) and excretion of pyruvate. Addition of pyruvate to the medium enhanced the survival of SK36 in overnight cultures. Meanwhile, elevated pyruvate levels were detected in the cultures of a small but significant percentage (∼10%) of clinical isolates of oral commensal bacteria. Furthermore, the manL mutant showed higher expression of the arginine deiminase system than the wild type, which enhanced the ability of the mutant to raise environmental pH when arginine was present. To our surprise, significant discrepancies in genome sequence were identified between strain SK36 obtained from ATCC and the sequence deposited in GenBank. As the conditions that are likely associated with the emergence of spontaneous manL mutations, i.e., excess carbohydrates and low pH, are those associated with caries development, we propose that glucose-PTS strongly influences commensal-pathogen interactions by altering the production of ammonia, pyruvate, and H2O2. IMPORTANCE A health-associated dental microbiome provides a potent defense against pathogens and diseases. Streptococcus sanguinis is an abundant member of a health-associated oral flora that antagonizes pathogens by producing hydrogen peroxide. There is a need for a better understanding of the mechanisms that allow bacteria to survive carbohydrate-rich and acidic environments associated with the development of dental caries. We report the isolation and characterization of spontaneous mutants of S. sanguinis with impairment in glucose transport. The resultant reprograming of the central metabolism in these mutants reduced the production of lactic acid and increased pyruvate accumulation; the latter enables these bacteria to better cope with hydrogen peroxide and low pH. The implications of these discoveries in the development of dental caries are discussed.
Collapse
|
40
|
Barbour A, Elebyary O, Fine N, Oveisi M, Glogauer M. Metabolites of the Oral Microbiome: Important Mediators of Multi-Kingdom Interactions. FEMS Microbiol Rev 2021; 46:6316110. [PMID: 34227664 DOI: 10.1093/femsre/fuab039] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 07/02/2021] [Indexed: 12/12/2022] Open
Abstract
The oral cavity hosts over 700 different microbial species that produce a rich reservoir of bioactive metabolites critical to oral health maintenance. Over the last two decades, new insights into the oral microbiome and its importance in health and disease have emerged mainly due to the discovery of new oral microbial species using next-generation sequencing (NGS). This advancement has revolutionized the documentation of unique microbial profiles associated with different niches and health/disease states within the oral cavity and the relation of the oral bacteria to systemic diseases. However, less work has been done to identify and characterize the unique oral microbial metabolites that play critical roles in maintaining equilibrium between the various oral microbial species and their human hosts. This article discusses the most significant microbial metabolites produced by these diverse communities of oral bacteria that can either foster health or contribute to disease. Finally, we shed light on how advances in genomics and genome mining can provide a high throughput platform for discovering novel bioactive metabolites derived from the human oral microbiome to tackle emerging human infections and systemic diseases.
Collapse
Affiliation(s)
- Abdelahhad Barbour
- Faculty of Dentistry, University of Toronto, Toronto, ON, Canada, M5G 1G6, Canada
| | - Omnia Elebyary
- Faculty of Dentistry, University of Toronto, Toronto, ON, Canada, M5G 1G6, Canada
| | - Noah Fine
- Faculty of Dentistry, University of Toronto, Toronto, ON, Canada, M5G 1G6, Canada
| | - Morvarid Oveisi
- Faculty of Dentistry, University of Toronto, Toronto, ON, Canada, M5G 1G6, Canada
| | - Michael Glogauer
- Faculty of Dentistry, University of Toronto, Toronto, ON, Canada, M5G 1G6, Canada.,Department of Dental Oncology, Maxillofacial and Ocular Prosthetics, Princess Margaret Cancer Centre, Toronto, ON, Canada, M5G 2M9, Canada
| |
Collapse
|
41
|
The Effects of Nonnutritive Sweeteners on the Cariogenic Potential of Oral Microbiome. BIOMED RESEARCH INTERNATIONAL 2021; 2021:9967035. [PMID: 34258285 PMCID: PMC8253641 DOI: 10.1155/2021/9967035] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 06/07/2021] [Accepted: 06/18/2021] [Indexed: 02/05/2023]
Abstract
Nonnutritive sweeteners (NNSs) are sugar substitutes widely used to reduce the negative health effects of excessive sugar consumption. Dental caries, one of the most prevalent chronic diseases globally, results from a pathogenic biofilm with microecological imbalance and frequent exposure to sugars. Some research has shown that certain NNSs possess less cariogenic potential than sucrose, indicating their putative effect on oral microbiome. To uncover the alterations of acidogenic pathogens and alkali-generating commensals, as well as the biofilm cariogenic potential under the influence of NNSs, we selected four common NNSs (acesulfame-K, aspartame, saccharin, and sucralose) and established single-, dual-, and multispecies in vitro culture model to assess their effects on Streptococcus mutans (S. mutans) and/or Streptococcus sanguinis (S. sanguinis) compared to sucrose with the same sweetness. The results showed that NNSs significantly suppressed the planktonic growth, acid production, and biofilm formation of S. mutans or S. sanguinis compared with sucrose in single-species cultures. Additionally, decreased S. mutans/S. sanguinis ratio, less EPS generation, and higher pH value were observed in dual-species and saliva-derived multispecies biofilms with supplementary NNSs. Collectively, this study demonstrates that NNSs inhibit the cariogenic potential of biofilms by maintaining microbial equilibrium, thus having a promising prospect as anticaries agents.
Collapse
|
42
|
Wu M, Huang S, Du J, Jiang S, Cai Z, Zhan L, Huang X. Role of D-alanylation of Streptococcus mutans lipoteichoic acid in interspecies competitiveness. Mol Oral Microbiol 2021; 36:233-242. [PMID: 33977670 DOI: 10.1111/omi.12344] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 04/24/2021] [Accepted: 04/26/2021] [Indexed: 11/28/2022]
Abstract
BACKGROUND The D-alanylation of lipoteichoic acid (LTA) is essential for the physiological metabolism of Streptococcus mutans (S. mutans). This study was designed to investigate the influence of D-alanylation of LTA on interspecies competitiveness of S. mutans. METHODS The process of D-alanylation was blocked by the inactivation of dltC. Agar competition assays, conditioned medium assays, and qRT-PCR were used to evaluate the production of antimicrobial compounds in S. mutans mutant. Dual-species biofilm was formed to investigate the competitiveness of S. mutans mutant cocultured with S. sanguinis or S. gordonii. RESULTS S. mutans mutant could not produce antimicrobial compounds efficiently when cocultured with commensal bacteria (*p < 0.05). The mutant showed compromised competitiveness in dual-species biofilms. The ratio of the mutant in dual-species biofilms decreased, and the terminal pH of the culture medium in mutant groups (mutant+S. sanguinis/S. gordonii) was higher than that in wild-type groups (*p < 0.05). Scanning electron microscope (SEM) showed weaker demineralization of enamel treated with dual-species biofilms consisting of mutant and commensal bacteria. CONCLUSION D-Alanylation is involved in interspecies competitiveness of S. mutans within oral biofilm by regulating mutacins and lactic acid production, which may modulate the profiles of dental biofilms. Results provide new insights into dental caries prevention and treatment.
Collapse
Affiliation(s)
- Minjing Wu
- Fujian Key Laboratory of Oral Diseases &, Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China.,Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Shan Huang
- Fujian Key Laboratory of Oral Diseases &, Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Jingyun Du
- Fujian Key Laboratory of Oral Diseases &, Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Shan Jiang
- School of Stomatology, Shenzhen University Health Science Center, Shenzhen, China
| | - Zhiyu Cai
- Department of Stomatology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Ling Zhan
- Division of Pediatric Dentistry, Department of Orofacial Sciences, Department of Preventive and Restorative Dental Sciences, University of California, San Francisco, CA, USA
| | - Xiaojing Huang
- Fujian Key Laboratory of Oral Diseases &, Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| |
Collapse
|
43
|
Jo JK, Seo SH, Park SE, Kim HW, Kim EJ, Na CS, Cho KM, Kwon SJ, Moon YH, Son HS. Identification of Salivary Microorganisms and Metabolites Associated with Halitosis. Metabolites 2021; 11:metabo11060362. [PMID: 34200451 PMCID: PMC8226648 DOI: 10.3390/metabo11060362] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 05/27/2021] [Accepted: 06/03/2021] [Indexed: 02/05/2023] Open
Abstract
Halitosis is mainly caused by the action of oral microbes. The purpose of this study was to investigate the differences in salivary microbes and metabolites between subjects with and without halitosis. Of the 52 participants, 22 were classified into the halitosis group by the volatile sulfur compound analysis on breath samples. The 16S rRNA gene amplicon sequencing and metabolomics approaches were used to investigate the difference in microbes and metabolites in saliva of the control and halitosis groups. The profiles of microbiota and metabolites were relatively different between the halitosis and control groups. The relative abundances of Prevotella, Alloprevotella, and Megasphaera were significantly higher in the halitosis group. In contrast, the relative abundances of Streptococcus, Rothia, and Haemophilus were considerably higher in the control group. The levels of 5-aminovaleric acid and n-acetylornithine were significantly higher in the halitosis group. The correlation between identified metabolites and microbiota reveals that Alloprevotella and Prevotella might be related to the cadaverine and putrescine pathways that cause halitosis. This study could provide insight into the mechanisms of halitosis.
Collapse
Affiliation(s)
- Jae-kwon Jo
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Korea; (J.-k.J.); (S.-E.P.); (H.-W.K.)
| | | | - Seong-Eun Park
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Korea; (J.-k.J.); (S.-E.P.); (H.-W.K.)
| | - Hyun-Woo Kim
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Korea; (J.-k.J.); (S.-E.P.); (H.-W.K.)
| | - Eun-Ju Kim
- Department of Korean Medicine, Dongshin University, Naju 58245, Korea; (E.-J.K.); (C.-S.N.)
| | - Chang-Su Na
- Department of Korean Medicine, Dongshin University, Naju 58245, Korea; (E.-J.K.); (C.-S.N.)
| | - Kwang-Moon Cho
- AccuGene Inc., Incheon 22006, Korea; (K.-M.C.); (S.-J.K.)
| | - Sun-Jae Kwon
- AccuGene Inc., Incheon 22006, Korea; (K.-M.C.); (S.-J.K.)
| | - Young-Ho Moon
- Naju Korean Medical Hospital, Dongshin University, Naju 58326, Korea
- Correspondence: (Y.-H.M.); (H.-S.S.); Tel.: +82-61-338-7812 (Y.-H.M.); +82-2-3290-3053 (H.-S.S.)
| | - Hong-Seok Son
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Korea; (J.-k.J.); (S.-E.P.); (H.-W.K.)
- Correspondence: (Y.-H.M.); (H.-S.S.); Tel.: +82-61-338-7812 (Y.-H.M.); +82-2-3290-3053 (H.-S.S.)
| |
Collapse
|
44
|
Rosier BT, Palazón C, García-Esteban S, Artacho A, Galiana A, Mira A. A Single Dose of Nitrate Increases Resilience Against Acidification Derived From Sugar Fermentation by the Oral Microbiome. Front Cell Infect Microbiol 2021; 11:692883. [PMID: 34195102 PMCID: PMC8238012 DOI: 10.3389/fcimb.2021.692883] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 05/17/2021] [Indexed: 12/23/2022] Open
Abstract
Tooth decay starts with enamel demineralization due to an acidic pH, which arises from sugar fermentation by acidogenic oral bacteria. Previous in vitro work has demonstrated that nitrate limits acidification when incubating complex oral communities with sugar for short periods (e.g., 1-5 h), driven by changes in the microbiota metabolism and/or composition. To test whether a single dose of nitrate can reduce acidification derived from sugar fermentation in vivo, 12 individuals received a nitrate-rich beetroot supplement, which was compared to a placebo in a blinded crossover setting. Sucrose-rinses were performed at baseline and 2 h after supplement or placebo intake, and the salivary pH, nitrate, nitrite, ammonium and lactate were measured. After nitrate supplement intake, the sucrose-induced salivary pH drop was attenuated when compared with the placebo (p < 0.05). Salivary nitrate negatively correlated with lactate production and positively with ΔpH after sucrose exposure (r= -0.508 and 0.436, respectively, both p < 0.05). Two additional pilot studies were performed to test the effect of sucrose rinses 1 h (n = 6) and 4 h (n = 6) after nitrate supplement intake. In the 4 h study, nitrate intake was compared with water intake and bacterial profiles were analysed using 16S rRNA gene Illumina sequencing and qPCR detection of Rothia. Sucrose rinses caused a significant pH drop (p < 0.05), except 1 h and 4 h after nitrate supplement intake. After 4 h of nitrate intake, there was less lactate produced compared to water intake (p < 0.05) and one genus; Rothia, increased in abundance. This small but significant increase was confirmed by qPCR (p < 0.05). The relative abundance of Rothia and Neisseria negatively correlated with lactate production (r = -0.601 and -0.669, respectively) and Neisseria positively correlated with pH following sucrose intake (r = 0.669, all p < 0.05). Together, these results show that nitrate can acutely limit acidification when sugars are fermented, which appears to result from lactate usage by nitrate-reducing bacteria. Future studies should assess the longitudinal impact of daily nitrate-rich vegetable or supplement intake on dental health.
Collapse
Affiliation(s)
- Bob T Rosier
- Department of Health and Genomics, Center for Advanced Research in Public Health, FISABIO Foundation, Valencia, Spain
| | - Carlos Palazón
- Department of Health and Genomics, Center for Advanced Research in Public Health, FISABIO Foundation, Valencia, Spain
| | - Sandra García-Esteban
- Department of Health and Genomics, Center for Advanced Research in Public Health, FISABIO Foundation, Valencia, Spain
| | - Alejandro Artacho
- Department of Health and Genomics, Center for Advanced Research in Public Health, FISABIO Foundation, Valencia, Spain
| | - Antonio Galiana
- Department of Microbiology, General University Hospital of Elche, FISABIO Foundation, Alicante, Spain
| | - Alex Mira
- Department of Health and Genomics, Center for Advanced Research in Public Health, FISABIO Foundation, Valencia, Spain
| |
Collapse
|
45
|
Zhang T, Kalimuthu S, Rajasekar V, Xu F, Yiu YC, Hui TKC, Neelakantan P, Chu Z. Biofilm inhibition in oral pathogens by nanodiamonds. Biomater Sci 2021; 9:5127-5135. [PMID: 33997876 DOI: 10.1039/d1bm00608h] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Complex microbial communities, e.g., biofilms residing in our oral cavity, have recognized clinical significance, as they are typically the main cause for infections. Particularly, they show high resistance to conventional antibiotics, and alternatives including nanotechnology are being intensively explored nowadays to provide more efficient therapeutics. Diamond nanoparticles, namely, nanodiamonds (NDs) with many promising physico-chemical properties, have been demonstrated to work as an effective antibacterial agent against planktonic cells (free-floating state). However, little is known about the behaviors of NDs against biofilms (sessile state). In this study, we uncovered their role in inhibiting biofilm formation and their disrupting effect on preformed biofilms in several selected orally and systemically important organisms. The current findings will advance the mechanistic understanding of NDs on oral pathogens and might accelerate corresponding clinical translation.
Collapse
Affiliation(s)
- Tongtong Zhang
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Khor B, Snow M, Herrman E, Ray N, Mansukhani K, Patel KA, Said-Al-Naief N, Maier T, Machida CA. Interconnections Between the Oral and Gut Microbiomes: Reversal of Microbial Dysbiosis and the Balance Between Systemic Health and Disease. Microorganisms 2021; 9:496. [PMID: 33652903 PMCID: PMC7996936 DOI: 10.3390/microorganisms9030496] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 02/22/2021] [Accepted: 02/23/2021] [Indexed: 12/13/2022] Open
Abstract
The human microbiota represents a complex array of microbial species that influence the balance between the health and pathology of their surrounding environment. These microorganisms impart important biological benefits to their host, such as immune regulation and resistance to pathogen colonization. Dysbiosis of microbial communities in the gut and mouth precede many oral and systemic diseases such as cancer, autoimmune-related conditions, and inflammatory states, and can involve the breakdown of innate barriers, immune dysregulation, pro-inflammatory signaling, and molecular mimicry. Emerging evidence suggests that periodontitis-associated pathogens can translocate to distant sites to elicit severe local and systemic pathologies, which necessitates research into future therapies. Fecal microbiota transplantation, probiotics, prebiotics, and synbiotics represent current modes of treatment to reverse microbial dysbiosis through the introduction of health-related bacterial species and substrates. Furthermore, the emerging field of precision medicine has been shown to be an effective method in modulating host immune response through targeting molecular biomarkers and inflammatory mediators. Although connections between the human microbiome, immune system, and systemic disease are becoming more apparent, the complex interplay and future innovations in treatment modalities will become elucidated through continued research and cross-disciplinary collaboration.
Collapse
Affiliation(s)
- Brandon Khor
- Academic DMD Program, Oregon Health & Science University, 2730 SW Moody Avenue, Portland, OR 97201, USA; (B.K.); (M.S.); (E.H.); (N.R.); (K.M.); (K.A.P.)
| | - Michael Snow
- Academic DMD Program, Oregon Health & Science University, 2730 SW Moody Avenue, Portland, OR 97201, USA; (B.K.); (M.S.); (E.H.); (N.R.); (K.M.); (K.A.P.)
| | - Elisa Herrman
- Academic DMD Program, Oregon Health & Science University, 2730 SW Moody Avenue, Portland, OR 97201, USA; (B.K.); (M.S.); (E.H.); (N.R.); (K.M.); (K.A.P.)
| | - Nicholas Ray
- Academic DMD Program, Oregon Health & Science University, 2730 SW Moody Avenue, Portland, OR 97201, USA; (B.K.); (M.S.); (E.H.); (N.R.); (K.M.); (K.A.P.)
| | - Kunal Mansukhani
- Academic DMD Program, Oregon Health & Science University, 2730 SW Moody Avenue, Portland, OR 97201, USA; (B.K.); (M.S.); (E.H.); (N.R.); (K.M.); (K.A.P.)
| | - Karan A. Patel
- Academic DMD Program, Oregon Health & Science University, 2730 SW Moody Avenue, Portland, OR 97201, USA; (B.K.); (M.S.); (E.H.); (N.R.); (K.M.); (K.A.P.)
| | - Nasser Said-Al-Naief
- Department of Integrative Biomedical and Diagnostic Sciences, School of Dentistry, Oregon Health & Science University 2730 SW Moody Avenue, Portland, OR 97201, USA; (N.S.-A.-N.); (T.M.)
| | - Tom Maier
- Department of Integrative Biomedical and Diagnostic Sciences, School of Dentistry, Oregon Health & Science University 2730 SW Moody Avenue, Portland, OR 97201, USA; (N.S.-A.-N.); (T.M.)
| | - Curtis A. Machida
- Department of Integrative Biomedical and Diagnostic Sciences, School of Dentistry, Oregon Health & Science University 2730 SW Moody Avenue, Portland, OR 97201, USA; (N.S.-A.-N.); (T.M.)
| |
Collapse
|
47
|
In Vivo Colonization with Candidate Oral Probiotics Attenuates Colonization and Virulence of Streptococcus mutans. Appl Environ Microbiol 2021; 87:AEM.02490-20. [PMID: 33277269 PMCID: PMC7851695 DOI: 10.1128/aem.02490-20] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
A collection of 113 Streptococcus strains from supragingival dental plaque of caries-free individuals were recently tested in vitro for direct antagonism of the dental caries pathogen Streptococcus mutans, and for their capacity for arginine catabolism via the arginine deiminase system (ADS). To advance their evaluation as potential probiotics, twelve strains of commensal oral streptococci with various antagonistic and ADS potentials were assessed in a mouse model for oral (i.e., oral mucosal pellicles and saliva) and dental colonization under four diets (healthy or high-sucrose, with or without prebiotic arginine). Colonization by autochthonous bacteria was also monitored. One strain failed to colonize, whereas oral colonization by the other eleven strains varied by 3 log units. Dental colonization was high for five strains regardless of diet, six strains increased colonization with at least one high-sucrose diet, and added dietary arginine decreased dental colonization of two strains. Streptococcus sp. A12 (high in vitro ADS activity and antagonism) and two engineered mutants lacking the ADS (ΔarcADS) or pyruvate oxidase-mediated H2O2 production (ΔspxB) were tested for competition against S. mutans UA159. A12 wild type and ΔarcADS colonized only transiently, whereas ΔspxB persisted, but without altering oral or dental colonization by S. mutans In testing four additional candidates, S. sanguinis BCC23 markedly attenuated S. mutans' oral and dental colonization, enhanced colonization of autochthonous bacteria, and decreased severity of smooth surface caries under highly cariogenic conditions. Results demonstrate the utility of the mouse model to evaluate potential probiotics, revealing little correlation between in vitro antagonism and competitiveness against S. mutans in vivo IMPORTANCE Our results demonstrate in vivo testing of potential oral probiotics can be accomplished and can yield information to facilitate the ultimate design and optimization of novel anti-caries probiotics. We show human oral commensals associated with dental health are an important source of potential probiotics that may be used to colonize patients under dietary conditions of highly varying cariogenicity. Assessment of competitiveness against dental caries pathogen Streptococcus mutans and impact on caries identified strains or genetic elements for further study. Results also uncovered strains that enhanced oral and dental colonization by autochthonous bacteria when challenged with S. mutans, suggesting cooperative interactions for future elucidation. Distinguishing a rare strain that effectively compete with S. mutans under conditions that promote caries further validates our systematic approach to more critically evaluate probiotics for use in humans.
Collapse
|
48
|
Abstract
This paper concerns the assessment of the current state of dentistry in the world and the prospects of its sustainable development. A traditional Chinese censer was adopted as the pattern, with a strong and stable support on three legs. The dominant diseases of the oral cavity are caries and periodontal diseases, with the inevitable consequence of toothlessness. From the caries 3.5–5 billion people suffer. Moreover, each of these diseases has a wide influence on the development of systemic complications. The territorial range of these diseases and their significant differentiation in severity in different countries and their impact on disability-adjusted life years index are presented (DALY). Edentulousness has a significant impact on the oral health-related quality of life (OHRQoL). The etiology of these diseases is presented, as well as the preventive and therapeutic strategies undertaken as a result of modifying the Deming circle through the fives’ rules idea. The state of development of Dentistry 4.0 is an element of the current stage of the industrial revolution Industry 4.0 and the great achievements of modern dental engineering. Dental treatment examples from the authors’ own clinical practice are given. The systemic safety of a huge number of dentists in the world is discussed, in place of the passive strategy of using more and more advanced personal protective equipment (PPE), introducing our own strategy for the active prevention of the spread of pathogenic microorganisms, including SARS-CoV-2. The ethical aspects of dentists’ activity towards their own patients and the ethical obligations of the dentist community towards society are discussed in detail. This paper is a polemic arguing against the view presented by a group of eminent specialists in the middle of last year in The Lancet. It is impossible to disagree with these views when it comes to waiting for egalitarianism in dental care, increasing the scope of prevention and eliminating discrimination in this area on the basis of scarcity and poverty. The views on the discrimination of dentistry in relation to other branches of medicine are far more debatable. Therefore, relevant world statistics for other branches of medicine are presented. The authors of this paper do not agree with the thesis that interventional dental treatment can be replaced with properly implemented prophylaxis. The final remarks, therefore, present a discussion of the prospects for the development of dentistry based on three pillars, analogous to the traditional Chinese censer obtaining a stable balance thanks to its three legs. The Dentistry Sustainable Development (DSD) > 2020 model, consisting of Global Dental Prevention (GDP), Advanced Interventionist Dentistry 4.0 (AID 4.0), and Dentistry Safety System (DSS), is presented.
Collapse
|
49
|
Baker JL, Morton JT, Dinis M, Alvarez R, Tran NC, Knight R, Edlund A. Deep metagenomics examines the oral microbiome during dental caries, revealing novel taxa and co-occurrences with host molecules. Genome Res 2020; 31:64-74. [PMID: 33239396 PMCID: PMC7849383 DOI: 10.1101/gr.265645.120] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 11/23/2020] [Indexed: 12/25/2022]
Abstract
Dental caries, the most common chronic infectious disease worldwide, has a complex etiology involving the interplay of microbial and host factors that are not completely understood. In this study, the oral microbiome and 38 host cytokines and chemokines were analyzed across 23 children with caries and 24 children with healthy dentition. De novo assembly of metagenomic sequencing obtained 527 metagenome-assembled genomes (MAGs), representing 150 bacterial species. Forty-two of these species had no genomes in public repositories, thereby representing novel taxa. These new genomes greatly expanded the known pangenomes of many oral clades, including the enigmatic Saccharibacteria clades G3 and G6, which had distinct functional repertoires compared to other oral Saccharibacteria. Saccharibacteria are understood to be obligate epibionts, which are dependent on host bacteria. These data suggest that the various Saccharibacteria clades may rely on their hosts for highly distinct metabolic requirements, which would have significant evolutionary and ecological implications. Across the study group, Rothia, Neisseria, and Haemophilus spp. were associated with good dental health, whereas Prevotella spp., Streptococcus mutans, and Human herpesvirus 4 (Epstein-Barr virus [EBV]) were more prevalent in children with caries. Finally, 10 of the host immunological markers were significantly elevated in the caries group, and co-occurrence analysis provided an atlas of potential relationships between microbes and host immunological molecules. Overall, this study illustrated the oral microbiome at an unprecedented resolution and contributed several leads for further study that will increase the understanding of caries pathogenesis and guide therapeutic development.
Collapse
Affiliation(s)
- Jonathon L Baker
- Genomic Medicine Group, J. Craig Venter Institute, La Jolla, California 92037, USA
| | - James T Morton
- Systems Biology Group, Flatiron Institute, New York, New York 10010, USA
| | - Márcia Dinis
- Section of Pediatric Dentistry, UCLA School of Dentistry, Los Angeles, California 90095-1668, USA
| | - Ruth Alvarez
- Section of Pediatric Dentistry, UCLA School of Dentistry, Los Angeles, California 90095-1668, USA
| | - Nini C Tran
- Section of Pediatric Dentistry, UCLA School of Dentistry, Los Angeles, California 90095-1668, USA
| | - Rob Knight
- Center for Microbiome Innovation, University of California at San Diego, La Jolla, California 92161, USA.,Department of Pediatrics, University of California at San Diego, La Jolla, California 92161, USA.,Department of Computer Science and Engineering, University of California at San Diego, La Jolla, California 92093, USA.,Department of Bioengineering, University of California at San Diego, La Jolla, California 92093, USA
| | - Anna Edlund
- Genomic Medicine Group, J. Craig Venter Institute, La Jolla, California 92037, USA.,Department of Pediatrics, University of California at San Diego, La Jolla, California 92161, USA
| |
Collapse
|
50
|
Rosier BT, Moya-Gonzalvez EM, Corell-Escuin P, Mira A. Isolation and Characterization of Nitrate-Reducing Bacteria as Potential Probiotics for Oral and Systemic Health. Front Microbiol 2020; 11:555465. [PMID: 33042063 PMCID: PMC7522554 DOI: 10.3389/fmicb.2020.555465] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 08/24/2020] [Indexed: 12/29/2022] Open
Abstract
Recent evidence indicates that the reduction of salivary nitrate by oral bacteria can contribute to prevent oral diseases, as well as increase systemic nitric oxide levels that can improve conditions such as hypertension and diabetes. The objective of the current manuscript was to isolate nitrate-reducing bacteria from the oral cavity of healthy donors and test their in vitro probiotic potential to increase the nitrate-reduction capacity (NRC) of oral communities. Sixty-two isolates were obtained from five different donors of which 53 were confirmed to be nitrate-reducers. Ten isolates were selected based on high NRC as well as high growth rates and low acidogenicity, all being Rothia species. The genomes of these ten isolates confirmed the presence of nitrate- and nitrite reductase genes, as well as lactate utilization genes, and the absence of antimicrobial resistance, mobile genetic elements and virulence genes. The pH at which most nitrate was reduced differed between strains. However, acidic pH 6 always stimulated the reduction of nitrite compared to neutral pH 7 or slightly alkaline pH 7.5 (p < 0.01). We tested the effect of six out of 10 isolates on in vitro oral biofilm development in the presence or absence of 6.5 mM nitrate. The integration of the isolates into in vitro communities was confirmed by Illumina sequencing. The NRC of the bacterial communities increased when adding the isolates compared to controls without isolates (p < 0.05). When adding nitrate (prebiotic treatment) or isolates in combination with nitrate (symbiotic treatment), a smaller decrease in pH derived from sugar metabolism was observed (p < 0.05), which for some symbiotic combinations appeared to be due to lactate consumption. Interestingly, there was a strong correlation between the NRC of oral communities and ammonia production even in the absence of nitrate (R = 0.814, p < 0.01), which indicates that bacteria involved in these processes are related. As observed in our study, individuals differ in their NRC. Thus, some may have direct benefits from nitrate as a prebiotic as their microbiota naturally reduces significant amounts, while others may benefit more from a symbiotic combination (nitrate + nitrate-reducing probiotic). Future clinical studies should test the effects of these treatments on oral and systemic health.
Collapse
Affiliation(s)
| | | | | | - Alex Mira
- Department of Health and Genomics, Center for Advanced Research in Public Health, FISABIO Foundation, Valencia, Spain
| |
Collapse
|