1
|
Zhuo H, Zhang S, Wang H, Deng J, Zhang X. Gelatin methacryloyl @MP196/exos hydrogel induced neutrophil apoptosis and macrophage M2 polarization to inhibit periodontal bone loss. Colloids Surf B Biointerfaces 2024; 248:114466. [PMID: 39729702 DOI: 10.1016/j.colsurfb.2024.114466] [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: 11/24/2024] [Revised: 12/13/2024] [Accepted: 12/17/2024] [Indexed: 12/29/2024]
Abstract
OBJECTIVES Periodontitis is an inflammatory and destructive disease caused by dental plaque, which can result in the immune microenvironment disorders and loss of periodontal support tissue. In order to promote the restoration of local microenvironment stability, a functional biomaterial Gelatin methacryloyl @MP196/exos based on characteristics of disease occurrence is designed. METHODS Transmission electron microscopy, nanosight particle tracking analysis and western blot analysis were applied to prove the presence of exos in GelMA@MP196/exos. The swelling and degradation rates of GelMA@MP196/exos were evaluated. Cell proliferation, antibacterial ability and cellular uptake and intracellular internalization of exos were assessed in the study. Efferocytosis and M2 polarization of macrophages was estimated and the effects of GelMA@MP196/exos were proved in vivo. RESULTS GelMA@MP196/exos upregulated the expression of genes and proteins related to neutrophil apoptosis and promoted neutrophil apoptosis, macrophage M2 polarization, and efferocytosis. Furthermore, GelMA@MP196/exos exhibited significant antibacterial activity against Streptococcus gordonii, Fusobacterium nucleatum, and Porphyromonas gingivalis. GelMA@MP196/exos alleviated periodontitis and reduced alveolar bone loss in vivo in rat models. CONCLUSIONS GelMA@MP196/exos can serve as a potential strategy for the treatment of periodontitis. CLINICAL SIGNIFICANCE The main aim of periodontal therapy is to remove dental plaque and eliminate inflammation. However, some patients with low plaque scores and insufficient neutrophil clearance, resulting in poor responsiveness to periodontal therapy. Under the circumstances, local Application of drug that regulate the immune microenvironment had significance in controlling the progression of inflammation.
Collapse
Affiliation(s)
- Haiwei Zhuo
- Department of Periodontology, Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, No.12 Qixiangtai Road, Heping District, Tianjin 300070, PR China; Tianjin Medical University Institute of Stomatology, No.12 Qixiangtai Road, Heping District, Tianjin 300070, PR China
| | - Shuting Zhang
- Department of Periodontology, Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, No.12 Qixiangtai Road, Heping District, Tianjin 300070, PR China; Tianjin Medical University Institute of Stomatology, No.12 Qixiangtai Road, Heping District, Tianjin 300070, PR China
| | - Hongbo Wang
- Department of Periodontology, Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, No.12 Qixiangtai Road, Heping District, Tianjin 300070, PR China; Tianjin Medical University Institute of Stomatology, No.12 Qixiangtai Road, Heping District, Tianjin 300070, PR China
| | - Jiayin Deng
- Department of Periodontology, Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, No.12 Qixiangtai Road, Heping District, Tianjin 300070, PR China; Tianjin Medical University Institute of Stomatology, No.12 Qixiangtai Road, Heping District, Tianjin 300070, PR China.
| | - Xi Zhang
- Department of Periodontology, Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, No.12 Qixiangtai Road, Heping District, Tianjin 300070, PR China; Tianjin Medical University Institute of Stomatology, No.12 Qixiangtai Road, Heping District, Tianjin 300070, PR China.
| |
Collapse
|
2
|
Afonso AC, Saavedra MJ, Simões M, Simões LC. The role of the proteosurfaceome and exoproteome in bacterial coaggregation. Biotechnol Adv 2024; 79:108505. [PMID: 39694122 DOI: 10.1016/j.biotechadv.2024.108505] [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: 04/17/2024] [Revised: 12/06/2024] [Accepted: 12/12/2024] [Indexed: 12/20/2024]
Abstract
Bacterial coaggregation is a critical process in multispecies biofilm formation, driven by specific molecular interactions that facilitate the adhesion and aggregation of bacterial cells. These interactions are essential for the development and persistence of complex microbial communities. This review provides a comprehensive analysis of the roles of the proteosurfaceome and exoproteome in bacterial coaggregation. The proteosurfaceome, comprising surface-bound molecules such as adhesins, drives species-specific interactions crucial for partner recognition and adhesion. In parallel, the exoproteome, particularly extracellular polymeric substances (EPS), enhances aggregate stability by reinforcing structural integrity and facilitating intercellular communication, although its direct role in coaggregation remains to be fully clarified. By integrating these perspectives, this review aims to elucidate how the proteosurfaceome and exoproteome influence bacterial coaggregation, offering insights into their combined impact on microbial community structure and function. Furthermore, we highlight existing knowledge gaps and propose directions for future research.
Collapse
Affiliation(s)
- Ana C Afonso
- LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr Roberto Frias, 4200-465 Porto, Portugal; ALiCE - Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr Roberto Frias, 4200-465 Porto, Portugal; CITAB, Department of Veterinary Sciences, University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal; CEB-LABBELS, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
| | - Maria J Saavedra
- CITAB, Department of Veterinary Sciences, University of Trás-os-Montes e Alto Douro, 5000-801 Vila Real, Portugal
| | - Manuel Simões
- LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr Roberto Frias, 4200-465 Porto, Portugal; ALiCE - Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr Roberto Frias, 4200-465 Porto, Portugal.
| | - Lúcia C Simões
- CEB-LABBELS, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal.
| |
Collapse
|
3
|
Ahirwar P, Kozlovskaya V, Pukkanasut P, Nikishau P, Nealy S, Harber G, Michalek SM, Antony L, Wu H, Kharlampieva E, Velu SE. Polymer vesicles for the delivery of inhibitors of cariogenic biofilm. Dent Mater 2024; 40:1937-1953. [PMID: 39317560 PMCID: PMC11580801 DOI: 10.1016/j.dental.2024.09.006] [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: 06/14/2024] [Revised: 09/06/2024] [Accepted: 09/10/2024] [Indexed: 09/26/2024]
Abstract
OBJECTIVES The goal of this study is to develop a novel drug delivery platform for the pH-responsive delivery of biofilm inhibitors as a potential avenue to prevent and treat dental caries. METHODS Biofilm and growth inhibition assays were performed in polystyrene microtiter 96-well plates. Docking analysis was performed using the reported GtfB + HA5 co-crystal structure (PDB code: 8fg8) in SeeSAR 13.0.1 software. Polymersome vesicles were assembled from poly(N-vinylpyrrolidone)8-block-poly(dimethylsiloxane)64-block-poly(N-vinylpyrrolidone)8 (PVPON8-PDMS64-PVPON8) triblock copolymer using a nanoprecipitation method. Microbiome analysis of biofilm inhibitors and the in vivo drug release and antivirulence activities of polymersome encapsulated inhibitors have been carried out in a S. mutans induced rat caries model. RESULTS Biofilm inhibitors for HA5 and HA6 have shown species-specific selectivity towards S. mutans and the ability to preserve the oral microbiome in a S. mutans induced dental caries model. The inhibitors were encapsulated into pH-responsive block copolymer vesicles to generate polymersome-encapsulated biofilm inhibitors, and their biofilm and growth inhibitory activities against S. mutans and representative strains of oral commensal streptococci have been assessed. A 4-week treatment of S. mutans UA159 infected gnotobiotic rats with 100 µM of polymersome-encapsulated biofilm inhibitor, PEHA5 showed significant reductions in buccal, sulcal, and proximal caries scores compared to an untreated control group. SIGNIFICANCE Taken together, our data suggests that the biofilm-selective therapy using the polymersome-encapsulated biofilm inhibitors is a viable approach for the prevention and treatment of dental caries while preserving the oral microbiome.
Collapse
Affiliation(s)
- Parmanand Ahirwar
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Veronika Kozlovskaya
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Piyasuda Pukkanasut
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Pavel Nikishau
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Sarah Nealy
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Gregory Harber
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Suzanne M Michalek
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Linto Antony
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Hui Wu
- Department of Integrative Biomedical and Diagnostic Sciences, Oregon Health and Science University, Portland, OR 97239, USA
| | - Eugenia Kharlampieva
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, AL 35294, USA; Center of Nanoscale Materials and Biointegration, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | - Sadanandan E Velu
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, AL 35294, USA; Microbiome Center, Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA; Global Center for Craniofacial Oral and Dental Disorders, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| |
Collapse
|
4
|
De Angelis F, D’Arcangelo C, Di Lodovico S, Sorrentino E, Buonvivere M, D’Ercole S. Influence of Polymerization Protocol on Adhesion and Proliferation of Streptococcus mutans on Three Dental Composite Resins. Biomedicines 2024; 12:2235. [PMID: 39457548 PMCID: PMC11505629 DOI: 10.3390/biomedicines12102235] [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: 08/05/2024] [Revised: 09/23/2024] [Accepted: 09/27/2024] [Indexed: 10/28/2024] Open
Abstract
Background/Objectives: The aim of this in vitro study was to analyze and compare the Streptococcus mutans ability to adhere and form biofilm on the surface of light-cured VS heat-cured dental composite resins; Methods: Three composite resins with different chemical formulations were selected: GrandioSO (GR), Venus Diamond (VD) and Enamel Plus Hri Biofunction (BF). Disk-shaped specimens were manufactured by light-curing the composite resins (light-cured subgroups) and subjecting them to a further heat-curing cycle at 80° for 10 min (heat-cured subgroups). Specimens were analyzed for planktonic CFU count (CFU/mL), sessile CFU count (CFU/mL) and for biomass quantification (OD570nm); Results: The planktonic CFU count was higher in all the light-cured subgroups than in the heat-cured subgroups (light-cured: GR = 7.23 × 106, VD = 2.14 × 107, BF = 4.40 × 107; heat-cured: GR = 4.89 × 106, VD = 4.95 × 106, BF = 2.80 × 107), with a statistically significant increase for BF and VD. Focusing on the sessile CFUs, both GR (light-cured = 7.49 × 106; heat-cured = 3.97 × 106) and VD (light-cured = 2.93 × 107; heat-cured = 6.07 × 106) showed a significantly increased number of colonies in the light-cured subgroups. The OD570nm values recorded for the light-cured BF subgroup (0.4280) were significantly increased compared to the heat-cured BF subgroup (0.1931); Conclusions: A more complete polymerization protocol seems to lead to a potential reduction in the risk of secondary caries.
Collapse
Affiliation(s)
- Francesco De Angelis
- Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University of Chieti–Pescara, Via dei Vestini 31, 66100 Chieti, Italy; (C.D.); (E.S.); (M.B.); (S.D.)
| | - Camillo D’Arcangelo
- Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University of Chieti–Pescara, Via dei Vestini 31, 66100 Chieti, Italy; (C.D.); (E.S.); (M.B.); (S.D.)
| | - Silvia Di Lodovico
- Department of Pharmacy, “G. d’Annunzio” University of Chieti–Pescara, Via dei Vestini 31, 66100 Chieti, Italy;
| | - Edoardo Sorrentino
- Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University of Chieti–Pescara, Via dei Vestini 31, 66100 Chieti, Italy; (C.D.); (E.S.); (M.B.); (S.D.)
| | - Matteo Buonvivere
- Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University of Chieti–Pescara, Via dei Vestini 31, 66100 Chieti, Italy; (C.D.); (E.S.); (M.B.); (S.D.)
| | - Simonetta D’Ercole
- Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University of Chieti–Pescara, Via dei Vestini 31, 66100 Chieti, Italy; (C.D.); (E.S.); (M.B.); (S.D.)
| |
Collapse
|
5
|
Al-Asfour A, Bhardwaj RG, Karched M. Growth Suppression of Oral Squamous Cell Carcinoma Cells by Lactobacillus Acidophilus. Int Dent J 2024; 74:1151-1160. [PMID: 38679518 PMCID: PMC11561490 DOI: 10.1016/j.identj.2024.03.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 03/18/2024] [Accepted: 03/28/2024] [Indexed: 05/01/2024] Open
Abstract
OBJECTIVES Oral squamous cell carcinoma (OSCC) is a highly aggressive form of oral cancer. Probiotic lactobacilli have demonstrated anticancer effects, whilst their interaction with Streptococcus mutans in this context remains unexplored. The objective of this study was to investigate the antiproliferative effect of Lactobacillus acidophilus on OSCC and to understand the effect of S mutans on OSCCs and whether it affects the antiproliferative potential of L acidophilus when co-exposed to OSCC. METHODS The human head and neck squamous cell carcinoma cells of the oral cavity (HNO97 cell line) were exposed to cultures of L acidophilus and S mutans separately and in combination. Further, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay was performed to assess the viability of HNO97 cells. Bacterial adhesion to HNO97 cells was examined by confocal microscopy and apoptosis by Nexin staining. To understand the underlying mechanism of apoptosis, expression of the tumour necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) gene and protein were determined by real-time polymerase chain reaction and quantitative enzyme-linked immunosorbent assay, respectively. RESULTS A significant decrease (53%-56%) in the viability of HNO97 cells on exposure to L acidophilus, S mutans, and the 2 species together demonstrated the antiproliferative activity of L acidophilus and S mutans. Both bacteria showed adhesion to HNO97 cells. The expression of the TRAIL gene increased 5-fold in HNO97 cells on treatment with L acidophilus and S mutans, which further increased to ∼17-fold with both species present. Expression levels of the TRAIL protein were significantly (P < .05) increased in bacteria-treated cell lysates. Further, bacteria-treated HNO97 cells exhibited lower live and intact cell percentages with higher proportions of cells in early and late apoptotic stages. CONCLUSIONS L acidophilus exhibits the antiproliferative activity against OSCC cells possibly partially via a TRAIL-induced mechanism of apoptosis, which is not affected by the presence of S mutans. These findings may encourage further investigation into the possible therapeutic application of probiotic L acidophilus in OSCC.
Collapse
Affiliation(s)
- Adel Al-Asfour
- Department of Surgical Sciences, College of Dentistry, Kuwait University, Kuwait City, Kuwait
| | - Radhika G Bhardwaj
- Oral Microbiology Research Laboratory, Department of Bioclinical Sciences, College of Dentistry, Kuwait University, Kuwait City, Kuwait; Department of Biotechnology, School of Arts and Science, American International University, Kuwait
| | - Maribasappa Karched
- Oral Microbiology Research Laboratory, Department of Bioclinical Sciences, College of Dentistry, Kuwait University, Kuwait City, Kuwait.
| |
Collapse
|
6
|
Kang J, Liang Y, Liu J, Hu M, Lin S, Zhong J, Wang C, Zeng Q, Zhang C. Dual roles of photosynthetic hydrogel with sustained oxygen generation in promoting cell survival and eradicating anaerobic infection. Mater Today Bio 2024; 28:101197. [PMID: 39221211 PMCID: PMC11364899 DOI: 10.1016/j.mtbio.2024.101197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 07/27/2024] [Accepted: 08/08/2024] [Indexed: 09/04/2024] Open
Abstract
Tissue engineering offers a promising alternative for oral and maxillofacial tissue defect rehabilitation; however, cells within a sizeable engineered tissue construct after transplantation inevitably face prolonged and severe hypoxic conditions, which may compromise the survivability of the transplanted cells and arouse the concern of anaerobic infection. Microalgae, which can convert carbon dioxide and water into oxygen and glucose through photosynthesis, have been studied as a source of oxygen supply for several biomedical applications, but their promise in orofacial tissue regeneration remains unexplored. Here, we demonstrated that through photosynthetic oxygenation, Chlamydomonas reinhardtii (C. reinhardtii) supported dental pulp stem cell (DPSC) energy production and survival under hypoxia. We developed a multifunctional photosynthetic hydrogel by embedding DPSCs and C. reinhardtii encapsulated alginate microspheres (CAMs) within gelatin methacryloyl hydrogel (GelMA) (CAMs@GelMA). This CAMs@GelMA hydrogel can generate a sustainable and sufficient oxygen supply, reverse intracellular hypoxic status, and enhance the metabolic activity and viability of DPSCs. Furthermore, the CAMs@GelMA hydrogel exhibited selective antibacterial activity against oral anaerobes and remarkable antibiofilm effects on multispecies biofilms by disrupting the hypoxic microenvironment and increasing reactive oxygen species generation. Our work presents an innovative photosynthetic strategy for oral tissue engineering and opens new avenues for addressing other hypoxia-related challenges.
Collapse
Affiliation(s)
- Jun Kang
- Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Ye Liang
- Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Junqing Liu
- Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Mingxin Hu
- Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Shulan Lin
- Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Jialin Zhong
- Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Chaogang Wang
- Guangdong Technology Research Center for Marine Algal Bioengineering, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
| | - Qinglu Zeng
- Department of Ocean Science, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Chengfei Zhang
- Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
7
|
Fakeeha G, AlHarbi S, Auda S, Balto H. The Impact of Silver Nanoparticles' Size on Biofilm Eradication. Int Dent J 2024:S0020-6539(24)01418-7. [PMID: 39266402 DOI: 10.1016/j.identj.2024.08.007] [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: 06/25/2024] [Revised: 07/29/2024] [Accepted: 08/06/2024] [Indexed: 09/14/2024] Open
Abstract
INTRODUCTION Efficient intracanal disinfection is required for a successful regenerative endodontic treatment. Thus, this study aimed to identify the silver nanoparticles' (NPs) size (AgNPs) with the highest antibiofilm efficacy when mixed with calcium hydroxide [Ca(OH)2] to eradicate an in vitro endodontic biofilm. METHODS The various sizes of AgNPs and mixtures were characterized by scanning electron microscopy, transmission electron microscopy, and ultraviolet-visible spectroscopy. A total of 168 dentin root segments were prepared, sterilized, and inoculated for 3 weeks with Actinomyces naeslundii and Fusobacterium nucleatum. Samples were randomly allocated to 4 experimental groups (n = 28/group): 2 nm AgNPs + 35% Ca(OH)2, 5 nm AgNPs + 35% Ca(OH)2, 10 nm AgNPs + 35% Ca(OH)2, and 35% Ca(OH)2 alone. Samples exposed to saline and triple antibiotic paste (TAP) acted as negative and positive control groups, respectively. After 1 and 2 weeks, samples were stained with LIVE/DEAD BacLight dye and examined under a confocal laser scanning microscope to determine the proportion of dead bacteria. RESULTS The characterization procedure revealed a spherical NP's structure with minor aggregations. Except for Ca(OH)2 group, all groups had significantly higher antibiofilm efficacy at 2 weeks. Both the 10 nm mixture (99.5%) and TAP (99.2%) exhibited the highest antibiofilm efficacy at 2 weeks and were not significantly different from one another (P > .05). No significant difference was noted between the 2 and 5 nm mixtures at 1 week (81% and 84%) and 2 weeks (89% and 91%). CONCLUSION The 10 nm AgNPs (0.02%) + 35% Ca(OH)2 mixture exhibited the highest antibiofilm efficacy at 2 weeks compared to all other mixtures at both observation periods. Interestingly, the 10 nm mixture performed similarly to TAP at 2 weeks. Excluding Ca(OH)2 group, longer application significantly improved the antibiofilm efficacy of all tested medicaments. CLINICAL RELEVANCE The 10 nm AgNPs + 35% Ca(OH)2 mixture revealed promising results as an intracanal medicament in the regenerative endodontic treatment protocol.
Collapse
Affiliation(s)
- Ghazal Fakeeha
- Department of Restorative Dental Sciences, College of Dentistry, King Saud University, Riyadh, Saudi Arabia
| | - Sarah AlHarbi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Sayed Auda
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Hanan Balto
- Department of Restorative Dental Sciences, College of Dentistry, King Saud University, Riyadh, Saudi Arabia.
| |
Collapse
|
8
|
Zhang S, Zhao Y, Lalsiamthara J, Peng Y, Qi L, Deng S, Wang Q. Current research progress on Prevotella intermedia and associated diseases. Crit Rev Microbiol 2024:1-18. [PMID: 39140115 DOI: 10.1080/1040841x.2024.2390594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 05/29/2024] [Accepted: 08/05/2024] [Indexed: 08/15/2024]
Abstract
Prevotella intermedia is a Gram-negative anaerobic bacterium that is a common pathogen of periodontitis. Recent studies have revealed that P. intermedia is closely associated with a variety of diseases involving multiple systems. Under the action of its virulence factors such as cysteine protease and adhesins, P. intermedia has the ability to bind and invade various host cells including gingival fibroblasts. It can also copolymerize a variety of pathogenic bacteria, leading to interference with the host's immune inflammatory response and causing various diseases. In this article, we review the progress of research on P. intermedia virulence factors and bacterial pathogenesis, and the correlation between P. intermedia and various diseases.
Collapse
Affiliation(s)
- Shuyang Zhang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Key Laboratory of Pollution Exposure and Health Intervention of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, China
| | - Yuheng Zhao
- College of Biology and Environmental Engineering, Zhejiang Shuren University, Hangzhou, China
| | - Jonathan Lalsiamthara
- Molecular Microbiology & Immunology, School of Medicine, Oregon Health & Science University, Portland, Oregon, USA
| | - Yan Peng
- Key Laboratory of Green Cleaning Technology& Detergent of Zhejiang Province, Hangzhou, China
| | - Linlong Qi
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Key Laboratory of Pollution Exposure and Health Intervention of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, China
| | - Shuli Deng
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Qingjing Wang
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Key Laboratory of Pollution Exposure and Health Intervention of Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, China
| |
Collapse
|
9
|
Tanwar H, Gnanasekaran JM, Allison D, Chuang LS, He X, Aimetti M, Baima G, Costalonga M, Cross RK, Sears C, Mehandru S, Cho J, Colombel JF, Raufman JP, Thumbigere-Math V. Unravelling the Oral-Gut Axis: Interconnection Between Periodontitis and Inflammatory Bowel Disease, Current Challenges, and Future Perspective. J Crohns Colitis 2024; 18:1319-1341. [PMID: 38417137 PMCID: PMC11324343 DOI: 10.1093/ecco-jcc/jjae028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 12/04/2023] [Accepted: 02/27/2024] [Indexed: 03/01/2024]
Abstract
As the opposite ends of the orodigestive tract, the oral cavity and the intestine share anatomical, microbial, and immunological ties that have bidirectional health implications. A growing body of evidence suggests an interconnection between oral pathologies and inflammatory bowel disease [IBD], implying a shift from the traditional concept of independent diseases to a complex, reciprocal cycle. This review outlines the evidence supporting an 'oral-gut' axis, marked by a higher prevalence of periodontitis and other oral conditions in IBD patients and vice versa. We present an in-depth examination of the interconnection between oral pathologies and IBD, highlighting the shared microbiological and immunological pathways, and proposing a 'multi-hit' hypothesis in the pathogenesis of periodontitis-mediated intestinal inflammation. Furthermore, the review underscores the critical need for a collaborative approach between dentists and gastroenterologists to provide holistic oral-systemic healthcare.
Collapse
Affiliation(s)
- Himanshi Tanwar
- Division of Periodontology, University of Maryland School of Dentistry, Baltimore, MD, USA
| | | | - Devon Allison
- Division of Periodontology, University of Maryland School of Dentistry, Baltimore, MD, USA
| | - Ling-shiang Chuang
- Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Xuesong He
- Department of Microbiology, The Forsyth Institute, Cambridge, MA, USA
| | - Mario Aimetti
- Department of Surgical Sciences, C.I.R. Dental School, University of Turin, Turin, Italy
| | - Giacomo Baima
- Department of Surgical Sciences, C.I.R. Dental School, University of Turin, Turin, Italy
| | - Massimo Costalonga
- Department of Diagnostic and Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN, USA
| | - Raymond K Cross
- Division of Gastroenterology & Hepatology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Cynthia Sears
- Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Saurabh Mehandru
- Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Judy Cho
- Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jean-Frederic Colombel
- Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jean-Pierre Raufman
- Division of Gastroenterology & Hepatology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Vivek Thumbigere-Math
- Division of Periodontology, University of Maryland School of Dentistry, Baltimore, MD, USA
- National Institute of Dental and Craniofacial Research, NIH, Bethesda, MD, USA
| |
Collapse
|
10
|
Christian N, Burden D, Emam A, Brenk A, Sperber S, Kalu M, Cuadra G, Palazzolo D. Effects of E-Liquids and Their Aerosols on Biofilm Formation and Growth of Oral Commensal Streptococcal Communities: Effect of Cinnamon and Menthol Flavors. Dent J (Basel) 2024; 12:232. [PMID: 39195076 DOI: 10.3390/dj12080232] [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: 06/04/2024] [Revised: 07/11/2024] [Accepted: 07/19/2024] [Indexed: 08/29/2024] Open
Abstract
(1) Background: The rise in electronic cigarette (E-cigarette) popularity, especially among adolescents, has prompted research to investigate potential effects on health. Although much research has been carried out on the effect on lung health, the first site exposed to vaping-the oral cavity-has received relatively little attention. The aims of this study were twofold: to examine the effects of E-liquids on the viability and hydrophobicity of oral commensal streptococci, and the effects of E-cigarette-generated aerosols on the biomass and viability of oral commensal streptococci. (2) Methods: Quantitative and confocal biofilm analysis, live-dead staining, and hydrophobicity assays were used to determine the effect on oral commensal streptococci after exposure to E-liquids and/or E-cigarette-generated aerosols. (3) Results: E-liquids and flavors have a bactericidal effect on multispecies oral commensal biofilms and increase the hydrophobicity of oral commensal streptococci. Flavorless and some flavored E-liquid aerosols have a bactericidal effect on oral commensal biofilms while having no effect on overall biomass. (4) Conclusions: These results indicate that E-liquids/E-cigarette-generated aerosols alter the chemical interactions and viability of oral commensal streptococci. Consequently, the use of E-cigarettes has the potential to alter the status of disease and health in the oral cavity and, by extension, affect systemic health.
Collapse
Affiliation(s)
- Nicole Christian
- Biology Department, Muhlenberg College, Allentown, PA 18104, USA
- School of Dentistry, University of Michigan, Ann Arbor, MI 48109, USA
| | - Daniel Burden
- Biology Department, Muhlenberg College, Allentown, PA 18104, USA
- School of Dental Medicine, University of Colorado, Aurora, CO 80045, USA
| | - Alexander Emam
- Biology Department, Muhlenberg College, Allentown, PA 18104, USA
| | - Alvin Brenk
- Debusk College of Osteopathic Medicine, Lincoln Memorial University, Harrogate, TN 37752, USA
- Yale New Haven Hospital, New Haven, CT 06510, USA
| | - Sarah Sperber
- Debusk College of Osteopathic Medicine, Lincoln Memorial University, Harrogate, TN 37752, USA
| | - Michael Kalu
- Debusk College of Osteopathic Medicine, Lincoln Memorial University, Harrogate, TN 37752, USA
| | - Giancarlo Cuadra
- Biology Department, Muhlenberg College, Allentown, PA 18104, USA
| | - Dominic Palazzolo
- Debusk College of Osteopathic Medicine, Lincoln Memorial University, Harrogate, TN 37752, USA
| |
Collapse
|
11
|
Han JM, Yun I, Yang KM, Kim HS, Kim YY, Jeong W, Hong SS, Hwang I. Ethanol extract from Astilbe chinensis inflorescence suppresses inflammation in macrophages and growth of oral pathogenic bacteria. PLoS One 2024; 19:e0306543. [PMID: 38959234 PMCID: PMC11221678 DOI: 10.1371/journal.pone.0306543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 06/18/2024] [Indexed: 07/05/2024] Open
Abstract
Chronic oral inflammation and biofilm-mediated infections drive diseases such as dental caries and periodontitis. This study investigated the anti-inflammatory and antibacterial potential of an ethanol extract from Astilbe chinensis inflorescence (GA-13-6) as a prominent candidate for natural complex substances (NCS) with therapeutic potential. In LPS-stimulated RAW 264.7 macrophages, GA-13-6 significantly suppressed proinflammatory mediators, including interleukin-6 (IL-6), tumor necrosis factor (TNF), and nitric oxide (NO), surpassing purified astilbin, a known bioactive compound found in A. chinensis. Furthermore, GA-13-6 downregulated the expression of cyclooxygenase-2 (COX2) and inducible nitric oxide synthase (iNOS), indicating an inhibitory effect on the inflammatory cascade. Remarkably, GA-13-6 exhibited selective antibacterial activity against Streptococcus mutans, Streptococcus sanguinis, and Porphyromonas gingivalis, key players in dental caries and periodontitis, respectively. These findings suggest that complex GA-13-6 holds the potential for the treatment or prevention of periodontal and dental diseases, as well as various other inflammation-related conditions, while averting the induction of antibiotic resistance.
Collapse
Affiliation(s)
- Jong Min Han
- DOCSmedi OralBiome Co. Ltd., Goyang-si, Republic of Korea
| | - Ina Yun
- Apple Tree Institute of Biomedical Science, Apple Tree Medical Foundation, Goyang-si, Gyeonggi-do, Republic of Korea
| | - Kyung Mi Yang
- Apple Tree Institute of Biomedical Science, Apple Tree Medical Foundation, Goyang-si, Gyeonggi-do, Republic of Korea
| | - Hye-Sung Kim
- Apple Tree Institute of Biomedical Science, Apple Tree Medical Foundation, Goyang-si, Gyeonggi-do, Republic of Korea
| | - Young-Youn Kim
- Apple Tree Institute of Biomedical Science, Apple Tree Medical Foundation, Goyang-si, Gyeonggi-do, Republic of Korea
| | - Wonsik Jeong
- Bio Industry Department, Gyeonggido Business & Science Accelerator (GBSA), Suwon-si, Gyeonggi-do, Republic of Korea
| | - Seong Su Hong
- Bio Industry Department, Gyeonggido Business & Science Accelerator (GBSA), Suwon-si, Gyeonggi-do, Republic of Korea
| | - Inseong Hwang
- Apple Tree Institute of Biomedical Science, Apple Tree Medical Foundation, Goyang-si, Gyeonggi-do, Republic of Korea
| |
Collapse
|
12
|
Fang L, Zhang Y, Cheng L, Zheng H, Wang Y, Qin L, Cai Y, Cheng L, Zhou W, Liu F, Wang S. Silica nanoparticles containing nano-silver and chlorhexidine to suppress Porphyromonas gingivalis biofilm and modulate multispecies biofilms toward healthy tendency. J Oral Microbiol 2024; 16:2361403. [PMID: 38847000 PMCID: PMC11155433 DOI: 10.1080/20002297.2024.2361403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 05/23/2024] [Indexed: 06/09/2024] Open
Abstract
Objectives This research first investigated the effect of mesoporous silica nanoparticles (nMS) carrying chlorhexidine and silver (nMS-nAg-Chx) on periodontitis-related biofilms. This study aimed to investigate (1) the antibacterial activity on Porphyromonas gingivalis (P. gingivalis) biofilm; (2) the suppressing effect on virulence of P. gingivalis biofilm; (3) the regulating effect on periodontitis-related multispecies biofilm. Methods Silver nanoparticles (nAg) and chlorhexidine (Chx) were co-loaded into nMS to form nMS-nAg-Chx. Inhibitory zone test and minimum inhibitory concentration (MIC) against P. gingivalis were tested. Growth curves, crystal violet (CV) staining, live/dead staining and scanning electron microscopy (SEM) observation were performed. Biofilm virulence was assessed. The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay and Quantitative Real Time-PCR (qPCR) were performed to validate the activity and composition changes of multispecies biofilm (P. gingivalis, Streptococcus gordonii and Streptococcus sanguinis). Results nMS-nAg-Chx inhibited P. gingivalis biofilm dose-dependently (p<0.05), with MIC of 18.75 µg/mL. There were fewer live bacteria, less biomass and less virulence in nMS-nAg-Chx groups (p<0.05). nMS-nAg-Chx inhibited and modified periodontitis-related biofilms. The proportion of pathogenic bacteria decreased from 16.08 to 1.07% and that of helpful bacteria increased from 82.65 to 94.31% in 25 μg/mL nMS-nAg-Chx group for 72 h. Conclusions nMS-nAg-Chx inhibited P. gingivalis growth, decreased biofilm virulence and modulated periodontitis-related multispecies biofilms toward healthy tendency. pH-sensitive nMS-nAg-Chx inhibit the pathogens and regulate oral microecology, showing great potential in periodontitis adjunctive therapy.
Collapse
Affiliation(s)
- Lixin Fang
- Stomatology Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- The Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yishuang Zhang
- Stomatology Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- The Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Long Cheng
- Stomatology Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- The Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Hao Zheng
- Stomatology Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- The Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yiyi Wang
- Stomatology Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- The Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Lu Qin
- Stomatology Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- The Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yingchun Cai
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lei Cheng
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, National Clinical Research Center for Oral Diseases, Sichuan University, Chengdu, China
- Department of Operative Dentistry and Endodontics, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Wen Zhou
- School and Hospital of Stomatology, Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key lab of Fujian College and University, Fuzhou, China
| | - Fei Liu
- Stomatology Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Suping Wang
- Stomatology Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
13
|
Xiao S, Sun G, Huang S, Lin C, Li Y. Nanoarchitectonics-Based Materials as a Promising Strategy in the Treatment of Endodontic Infections. Pharmaceutics 2024; 16:759. [PMID: 38931881 PMCID: PMC11207628 DOI: 10.3390/pharmaceutics16060759] [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: 04/20/2024] [Revised: 05/19/2024] [Accepted: 05/29/2024] [Indexed: 06/28/2024] Open
Abstract
Endodontic infections arise from the interactive activities of microbial communities colonizing in the intricate root canal system. The present study aims to update the latest knowledge of nanomaterials, their antimicrobial mechanisms, and their applications in endodontics. A detailed literature review of the current knowledge of nanomaterials used in endodontic applications was performed using the PubMed database. Antimicrobial nanomaterials with a small size, large specific surface area, and high chemical activity are introduced to act as irrigants, photosensitizer delivery systems, and medicaments, or to modify sealers. The application of nanomaterials in the endodontic field could enhance antimicrobial efficiency, increase dentin tubule penetration, and improve treatment outcomes. This study supports the potential of nanomaterials as a promising strategy in treating endodontic infections.
Collapse
Affiliation(s)
- Suli Xiao
- Department of Endodontics, Stomatological Hospital of Xiamen Medical College, Xiamen 361003, China;
- Xiamen Key Laboratory of Stomatological Disease Diagnosis and Treatment, Xiamen 361003, China
| | - Guanwen Sun
- Department of Stomatology, Fujian Medical University Xiamen Humanity Hospital, Xiamen 361018, China;
| | - Shan Huang
- Department of Stomatology, Zhongshan Hospital Affiliated to Xiamen University, Xiamen 361005, China;
| | - Chen Lin
- Department of Endodontics, Stomatological Hospital of Xiamen Medical College, Xiamen 361003, China;
- Xiamen Key Laboratory of Stomatological Disease Diagnosis and Treatment, Xiamen 361003, China
| | - Yijun Li
- Department of Endodontics, Stomatological Hospital of Xiamen Medical College, Xiamen 361003, China;
- Xiamen Key Laboratory of Stomatological Disease Diagnosis and Treatment, Xiamen 361003, China
| |
Collapse
|
14
|
Peřina V, Šmucler R, Němec P, Barták V. Update on Focal Infection Management: A Czech Interdisciplinary Consensus. Int Dent J 2024; 74:510-518. [PMID: 38044216 PMCID: PMC11123548 DOI: 10.1016/j.identj.2023.11.001] [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: 07/03/2023] [Revised: 10/17/2023] [Accepted: 11/01/2023] [Indexed: 12/05/2023] Open
Abstract
BACKGROUND The focal infection theory has been used to explain several chronic systemic diseases in the past. Systemic diseases were thought to be caused by focal infections, such as caries and periodontal diseases, and dentists were held responsible for these diseases due to the spread of oral infections. As knowledge of the interrelationship between oral microorganisms and the host immune response has evolved over the last few decades, the focal infection theory has been modified in various ways. The relationship between oral and systemic health appears to be more complex than that suggested by the classical theory of focal infections. Indeed, the contribution of the oral microbiota to some systemic diseases is gaining acceptance, as there are strong associations between periodontal disease and atherosclerotic vascular disease, diabetes, and hospital-associated pneumonia, amongst others. As many jurisdictions have various protocols for managing this oral-systemic axis of disease, we sought to provide a consensus on this notion with the help of a multidisciplinary team from the Czech Republic. METHODS A multidisciplinary team comprising physicians/surgeons in the specialities of dentistry, ear-nose and throat (ENT), cardiology, orthopaedics, oncology, and diabetology were quetioned with regard to their conceptual understanding of the focal infection theory particularly in relation to the oral-systemic axis. The team also established a protocol to determine the strength of these associations and to plan the therapeutic steps needed to treat focal odontogenic infections whenever possible. RESULTS Scoring algorithms were devised for odontogenic inflammatory diseases and systemic risks, and standardised procedures were developed for general use. CONCLUSIONS The designed algorithm of the oral-systemic axis will be helpful for all health care workers in guiding their patient management protocol.
Collapse
Affiliation(s)
- Vojtěch Peřina
- Department of Oral and Maxillofacial Surgery, Masaryk University, Faculty of Medicine and University Hospital Brno, Brno, Czech Republic; Czech Dental Chamber, Prague, Czech Republic.
| | - Roman Šmucler
- Czech Dental Chamber, Prague, Czech Republic; Department of Stomatology, 1st Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic; Department of Stomatology, Faculty of Medicine, Slovak Medical University, Bratislava, Slovak Republic
| | - Petr Němec
- Department of Cardiovascular Surgery and Transplantations, Masaryk University, Faculty of Medicine and St. Anne´s University Hospital, Brno, Czech Republic
| | - Vladislav Barták
- 1. Orthopedic Clinic of the 1st Medical Faculty of Charles University and University Hospital Motol, Prague, Czech Republic
| |
Collapse
|
15
|
Ravichandran A, Sivapackiam J, Periasamy S. Oral bacterial insights from a comparative study between healthy and comorbid diseased human individuals. Microb Pathog 2024; 191:106643. [PMID: 38631413 DOI: 10.1016/j.micpath.2024.106643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 03/04/2024] [Accepted: 04/10/2024] [Indexed: 04/19/2024]
Abstract
The human oral cavity is colonized by a diverse microbial community, which includes both native and transient colonizers. The microbial composition is crucial for maintaining oral homeostasis, but due to overgrowth or imbalances of these microbial communities, dysbiosis can occur. There is a lack of understanding of the research of native and transient colonizers in the oral cavity of the Indian subpopulation Therefore, in our present study, we explored the role and prevalence of transient and native colonizers between healthy and comorbid oral diseased human individuals. Culture-dependent techniques and culture-independent 16S r DNA metagenomic analyses were employed to isolate and study the interactions of native and transient colonizers from human oral samples. Among the 66 human individuals of both healthy and comorbid individuals, the most abundant isolate was found to be Bacillus amyloliquefaciens MCC 4424. In addition, the more prevalent culturable isolate from the healthy samples was Streptococcus salivarius MTCC 13009, whereas in comorbid samples Staphylococcus pasteuri MTCC 13076, Rothia dentocariosa MTCC 13010 and Pseudomonas aeruginosa MTCC 13077 were prevalent to a greater extent. 16S rDNA metagenomic analyses revealed the prevalence and abundance of genera such as Bacteroidetes and Proteobacteria in healthy individuals; consequently, Fusobacteria and Firmicutes were observed mostly in comorbid individuals. The significant differences in bacterial population density were observed in terms of the Shannon index (p = 0.5145) and Simpson index (p = 0.9061) between the healthy and comorbid groups. B. amyloliquefaciens MCC 4424 exhibits antagonistic behavior when grown as a dual-species with native and transient colonizers. This result is very consistent with the findings of antibiofilm studies using confocal laser scanning microscopy, which revealed a significant reduction in biofilm biovolume (73 %) and maximum thickness (80 %) and an increase in the rough coefficient of biofilms (30 %). Our data suggested that B. amyloliquefaciens MCC 4424 can be a native colonizer of Indian sub-populations. It may act as a novel candidate for oral healthcare applications and greatly aids in the regulation of transient species in the oral cavity.
Collapse
Affiliation(s)
- Anand Ravichandran
- Centre of Excellence in Biofilms, Department of Biotechnology, Rajalakshmi Engineering College (Autonomous), Thandalam, Chennai, 602105, Tamilnadu, India
| | - Jothilingam Sivapackiam
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Saravanan Periasamy
- Centre of Excellence in Biofilms, Department of Biotechnology, Rajalakshmi Engineering College (Autonomous), Thandalam, Chennai, 602105, Tamilnadu, India.
| |
Collapse
|
16
|
Ma Z, Jiang Z, Dong H, Xu W, Yan S, Chen J, Li A, Wang X. Microbial Communities and Functional Genes in Periodontitis and Healthy Controls. Int Dent J 2024; 74:638-646. [PMID: 38448300 PMCID: PMC11123521 DOI: 10.1016/j.identj.2024.01.012] [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: 08/29/2023] [Revised: 01/05/2024] [Accepted: 01/17/2024] [Indexed: 03/08/2024] Open
Abstract
BACKGROUND Periodontitis is a chronic progressive disease and the leading cause of tooth loss in adults. Recent studies have shown the impact of oral microbial communities on systemic health and diseases such as cancer, atherosclerosis, rheumatoid arthritis, inflammatory bowel disease, diabetes, hypertension, and Alzheimer's disease. In previous case control studies investigatin the relationship between periodontal disease and the oral microbiota, little attention has been paid to the intersections of these domains. METHODS Here, we used high-throughput 16S rRNA sequencing to analyse the differences in the microbial composition in saliva between a group of patients with chronic periodontitis (C; n = 51) and a healthy control group (H; n = 61) and predicted the functional gene composition by Phylogenetic Investigation of Communities by Reconstruction of Unobserved States. RESULTS We found significant alterations in oral microbial diversity between C and H (P = 0.002). Sixteen genera were significantly different between C and H, and 15 of them were enriched in C linear discriminant analysis (LDA > 2). Fifty functional genes were significantly different between C and H, and 34 of them were enriched in C (P < .025). CONCLUSIONS Periodontitis is associated with significant changes in the oral microbial community.
Collapse
Affiliation(s)
- Zhonghui Ma
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Gene Hospital of Henan Province, Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ze Jiang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Haoxin Dong
- Department of Stomatology, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Wenhua Xu
- Department of Stomatology, Zhengzhou People's Hospital, Zhengzhou, China
| | - Su Yan
- Health Management Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jingfeng Chen
- Health Management Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ang Li
- Gene Hospital of Henan Province, Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Xi Wang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
17
|
Min K, Bosma ML, John G, McGuire JA, DelSasso A, Milleman J, Milleman KR. Quantitative analysis of the effects of brushing, flossing, and mouthrinsing on supragingival and subgingival plaque microbiota: 12-week clinical trial. BMC Oral Health 2024; 24:575. [PMID: 38760758 PMCID: PMC11102210 DOI: 10.1186/s12903-024-04362-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 05/10/2024] [Indexed: 05/19/2024] Open
Abstract
BACKGROUND Translational microbiome research using next-generation DNA sequencing is challenging due to the semi-qualitative nature of relative abundance data. A novel method for quantitative analysis was applied in this 12-week clinical trial to understand the mechanical vs. chemotherapeutic actions of brushing, flossing, and mouthrinsing against the supragingival dental plaque microbiome. Enumeration of viable bacteria using vPCR was also applied on supragingival plaque for validation and on subgingival plaque to evaluate interventional effects below the gingival margin. METHODS Subjects with gingivitis were enrolled in a single center, examiner-blind, virtually supervised, parallel group controlled clinical trial. Subjects with gingivitis were randomized into brushing only (B); brushing and flossing (BF); brushing and rinsing with Listerine® Cool Mint® Antiseptic (BA); brushing and rinsing with Listerine® Cool Mint® Zero (BZ); or brushing, flossing, and rinsing with Listerine® Cool Mint® Zero (BFZ). All subjects brushed twice daily for 1 min with a sodium monofluorophosphate toothpaste and a soft-bristled toothbrush. Subjects who flossed used unflavored waxed dental floss once daily. Subjects assigned to mouthrinses rinsed twice daily. Plaque specimens were collected at the baseline visit and after 4 and 12 weeks of intervention. Bacterial cell number quantification was achieved by adding reference amounts of DNA controls to plaque samples prior to DNA extraction, followed by shallow shotgun metagenome sequencing. RESULTS 286 subjects completed the trial. The metagenomic data for supragingival plaque showed significant reductions in Shannon-Weaver diversity, species richness, and total and categorical bacterial abundances (commensal, gingivitis, and malodor) after 4 and 12 weeks for the BA, BZ, and BFZ groups compared to the B group, while no significant differences were observed between the B and BF groups. Supragingival plaque vPCR further validated these results, and subgingival plaque vPCR demonstrated significant efficacy for the BFZ intervention only. CONCLUSIONS This publication reports on a successful application of a quantitative method of microbiome analysis in a clinical trial demonstrating the sustained and superior efficacy of essential oil mouthrinses at controlling dental plaque compared to mechanical methods. The quantitative microbiological data in this trial also reinforce the safety and mechanism of action of EO mouthrinses against plaque microbial ecology and highlights the importance of elevating EO mouthrinsing as an integral part of an oral hygiene regimen. TRIAL REGISTRATION The trial was registered on ClinicalTrials.gov on 31/10/2022. The registration number is NCT05600231.
Collapse
Affiliation(s)
- Kyungrok Min
- Johnson & Johnson Consumer Inc, 199 Grandview Rd, Skillman, NJ, USA.
| | - Mary Lynn Bosma
- Johnson & Johnson Consumer Inc, 199 Grandview Rd, Skillman, NJ, USA
| | - Gabriella John
- Johnson & Johnson Consumer Inc, 199 Grandview Rd, Skillman, NJ, USA
| | - James A McGuire
- Johnson & Johnson Consumer Inc, 199 Grandview Rd, Skillman, NJ, USA
| | - Alicia DelSasso
- Johnson & Johnson Consumer Inc, 199 Grandview Rd, Skillman, NJ, USA
| | - Jeffery Milleman
- Salus Research, Inc, 1220 Medical Park Drive, Building 4, Fort Wayne, IN, USA
| | - Kimberly R Milleman
- Salus Research, Inc, 1220 Medical Park Drive, Building 4, Fort Wayne, IN, USA
| |
Collapse
|
18
|
Lamont RJ, Kuboniwa M. The polymicrobial pathogenicity of Porphyromonas gingivalis. FRONTIERS IN ORAL HEALTH 2024; 5:1404917. [PMID: 38736461 PMCID: PMC11082793 DOI: 10.3389/froh.2024.1404917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 04/12/2024] [Indexed: 05/14/2024] Open
Abstract
Accumulating microbiome data and mechanistic studies in vitro and in vivo have refined our understanding of the oral microbiota as a functionally integrated polymicrobial community. Emergent properties of these communities are driven to a large extent by interspecies communication which can be based on physical association, secreted small molecules or nutritional exchange. Porphyromonas gingivalis is a consensus periodontal pathogen; however, virulence is only expressed in the context of a polymicrobial community. Multivalent fimbriae mediate attachment to other oral species which can initiate a distinct transcriptional program in both constituents of the binding pair. P. gingivalis also responds to small molecules and nutritional cues produced by partner organisms. Physiological interdependence forms the basis of complex networks of cooperating organisms which begin to resemble an organismal entity exhibiting a spectrum of pathogenic potential.
Collapse
Affiliation(s)
- Richard J. Lamont
- Department of Oral Immunology and Infectious Diseases, School of Dentistry, University of Louisville, Louisville, KY, United States
| | - Masae Kuboniwa
- Department of Preventive Dentistry, Osaka University Graduate School of Dentistry, Suita, Osaka, Japan
| |
Collapse
|
19
|
Huang Y, Han Q, Zhou J, Meng X, Huo L, Lei Y. The effect of bovine trypsin on dental biofilm dispersion: an in vitro study. Odontology 2024; 112:501-511. [PMID: 37955766 DOI: 10.1007/s10266-023-00869-y] [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: 11/27/2022] [Accepted: 10/24/2023] [Indexed: 11/14/2023]
Abstract
To investigate the degradation effect of bovine trypsin on multispecies biofilm of caries-related bacteria and provide an experimental foundation for the prevention of dental caries. Standard strains of S. mutans, S. sanguis, S. gordonii, and L. acidophilus were co-cultured to form 24 h, 48 h, and 72 h biofilms. The experimental groups were treated with bovine trypsin for 30 s, 1 min, and 3 min. Morphological observation and quantitative analysis of extracellular polymeric substances (EPS), live bacteria, and dead bacteria were conducted using the confocal laser scanning microscope (CLSM). The morphological changes of EPS and bacteria were also observed using a scanning electron microscope (SEM). When biofilm was treated for 1 min, the minimal inhibitory concentration (MIC) of bovine trypsin to reduce EPS was 0.5 mg/mL in 24 h and 48 h biofilms, and the MIC of bovine trypsin was 2.5 mg/mL in 72 h biofilms (P < 0.05). When biofilm was treated for 3 min, the MIC of bovine trypsin to reduce EPS was 0.25 mg/mL in 24 h and 48 h biofilms, the MIC of bovine trypsin was 1 mg/mL in 72 h biofilm (P < 0.05). The ratio of live-to-dead bacteria in the treatment group was significantly lower than blank group in 24 h, 48 h, and 72 h multispecies biofilms (P < 0.05). Bovine trypsin can destroy multispecies biofilm structure, disperse biofilm and bacteria flora, and reduce the EPS and bacterial biomass in vitro, which are positively correlated with the application time and concentration.
Collapse
Affiliation(s)
- Yinxue Huang
- Department of Operative Dentistry, Preventive Dentistry and Endodontics, Kunming Medical University and Hospital of Stomatology, Kunming, 650106, China
- Yunnan Key Laboratory of Stomatology, Kunming, 650106, China
| | - Qunchao Han
- Department of Operative Dentistry, Preventive Dentistry and Endodontics, Kunming Medical University and Hospital of Stomatology, Kunming, 650106, China
| | - Jing Zhou
- Department of Operative Dentistry, Preventive Dentistry and Endodontics, Kunming Medical University and Hospital of Stomatology, Kunming, 650106, China
- Yunnan Key Laboratory of Stomatology, Kunming, 650106, China
| | - Xinhui Meng
- Department of Operative Dentistry, Preventive Dentistry and Endodontics, Kunming Medical University and Hospital of Stomatology, Kunming, 650106, China
- Yunnan Key Laboratory of Stomatology, Kunming, 650106, China
| | - Lijun Huo
- Department of Operative Dentistry, Preventive Dentistry and Endodontics, Kunming Medical University and Hospital of Stomatology, Kunming, 650106, China.
| | - Yayan Lei
- Department of Operative Dentistry, Preventive Dentistry and Endodontics, Kunming Medical University and Hospital of Stomatology, Kunming, 650106, China
| |
Collapse
|
20
|
Zou T, Liang Y, Kang J, Liu J, Kang W, Jiang S, Zhang C. Oxygen enrichment mediated by calcium peroxide loaded gelatin methacrylate hydrogel eradicates periodontal biofilms. Int J Biol Macromol 2024; 265:130868. [PMID: 38492687 DOI: 10.1016/j.ijbiomac.2024.130868] [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: 10/09/2023] [Revised: 03/11/2024] [Accepted: 03/12/2024] [Indexed: 03/18/2024]
Abstract
The low oxygen environment of the periodontal pocket favors pathogenic anaerobes' growth, biofilm formation, and quick recurrence after periodontal treatment. In contrast, oxygen is detrimental to anaerobes, such as Porphyromonas gingivalis (P. gingivalis), since they lack a complete anti-oxidation mechanism to detoxify the oxygen challenge. Therefore, consistently feeding pathogenic anaerobes with abundant oxygen would be an effective strategy to combat them. Here, we reported injectable oxygen-generating hydrogels as oxygen mediators to alleviate the local anaerobic environment and eliminate periodontal pathogens. Gelatin methacrylate (GelMA) hydrogels loaded with calcium peroxide (CPO) possessed excellent injectability and exhibited burst releases of oxygen within 24 h with a 40 % oxygen tension peak. CPO-GelMA hydrogels with CPO concentrations of 5, 10, and 15 % reduced 60, 99, and 89.9 % viable P. gingivalis, respectively. Five percentage CPO-GelMA hydrogel downregulated gingipain and fimA gene expression in P. gingivalis without resistance development. Moreover, the CPO-GelMA hydrogels remarkably prevented biofilm formation and eradicated both monospecies and multispecies bacterial biofilms. In conclusion, CPO-GelMA hydrogels exert remarkable antimicrobial and antibiofilm effects on subgingival biofilms, providing a promising strategy for periodontal treatment.
Collapse
Affiliation(s)
- Ting Zou
- Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong; Shenzhen Stomatology Hospital (Pingshan), Southern Medical University, Shenzhen, China
| | - Ye Liang
- Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong
| | - Jun Kang
- Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong
| | - Junqing Liu
- Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong
| | - Wenyan Kang
- Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong
| | - Shan Jiang
- Shenzhen Stomatology Hospital (Pingshan), Southern Medical University, Shenzhen, China
| | - Chengfei Zhang
- Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong.
| |
Collapse
|
21
|
Olczak T, Śmiga M, Antonyuk SV, Smalley JW. Hemophore-like proteins of the HmuY family in the oral and gut microbiome: unraveling the mystery of their evolution. Microbiol Mol Biol Rev 2024; 88:e0013123. [PMID: 38305743 PMCID: PMC10966948 DOI: 10.1128/mmbr.00131-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2024] Open
Abstract
SUMMARY Heme (iron protoporphyrin IX, FePPIX) is the main source of iron and PPIX for host-associated pathogenic bacteria, including members of the Bacteroidota (formerly Bacteroidetes) phylum. Porphyromonas gingivalis, a keystone oral pathogen, uses a unique heme uptake (Hmu) system, comprising a hemophore-like protein, designated as the first member of the novel HmuY family. Compared to classical, secreted hemophores utilized by Gram-negative bacteria or near-iron transporter domain-based hemophores utilized by Gram-positive bacteria, the HmuY family comprises structurally similar proteins that have undergone diversification during evolution. The best characterized are P. gingivalis HmuY and its homologs from Tannerella forsythia (Tfo), Prevotella intermedia (PinO and PinA), Bacteroides vulgatus (Bvu), and Bacteroides fragilis (BfrA, BfrB, and BfrC). In contrast to the two histidine residues coordinating heme iron in P. gingivalis HmuY, Tfo, PinO, PinA, Bvu, and BfrA preferentially use two methionine residues. Interestingly, BfrB, despite conserved methionine residue, binds the PPIX ring without iron coordination. BfrC binds neither heme nor PPIX in keeping with the lack of conserved histidine or methionine residues used by other members of the HmuY family. HmuY competes for heme binding and heme sequestration from host hemoproteins with other members of the HmuY family to increase P. gingivalis competitiveness. The participation of HmuY in the host immune response confirms its relevance in relation to the survival of P. gingivalis and its ability to induce dysbiosis not only in the oral microbiome but also in the gut microbiome or other host niches, leading to local injuries and involvement in comorbidities.
Collapse
Affiliation(s)
- Teresa Olczak
- Laboratory of Medical Biology, Faculty of Biotechnology, University of Wrocław, Wrocław, Poland
| | - Michał Śmiga
- Laboratory of Medical Biology, Faculty of Biotechnology, University of Wrocław, Wrocław, Poland
| | - Svetlana V. Antonyuk
- Molecular Biophysics Group, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, the University of Liverpool, Liverpool, United Kingdom
| | - John W. Smalley
- Institute of Life Course and Medical Sciences, School of Dentistry, the University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
22
|
Kadirvelu L, Sivaramalingam SS, Jothivel D, Chithiraiselvan DD, Karaiyagowder Govindarajan D, Kandaswamy K. A review on antimicrobial strategies in mitigating biofilm-associated infections on medical implants. CURRENT RESEARCH IN MICROBIAL SCIENCES 2024; 6:100231. [PMID: 38510214 PMCID: PMC10951465 DOI: 10.1016/j.crmicr.2024.100231] [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: 03/22/2024] Open
Abstract
Biomedical implants are crucial in providing support and functionality to patients with missing or defective body parts. However, implants carry an inherent risk of bacterial infections that are biofilm-associated and lead to significant complications. These infections often result in implant failure, requiring replacement by surgical restoration. Given these complications, it is crucial to study the biofilm formation mechanism on various biomedical implants that will help prevent implant failures. Therefore, this comprehensive review explores various types of implants (e.g., dental implant, orthopedic implant, tracheal stent, breast implant, central venous catheter, cochlear implant, urinary catheter, intraocular lens, and heart valve) and medical devices (hemodialyzer and pacemaker) in use. In addition, the mechanism of biofilm formation on those implants, and their pathogenesis were discussed. Furthermore, this article critically reviews various approaches in combating implant-associated infections, with a special emphasis on novel non-antibiotic alternatives to mitigate biofilm infections.
Collapse
Affiliation(s)
- Lohita Kadirvelu
- Research Center for Excellence in Microscopy, Department of Biotechnology, Kumaraguru College of Technology, Coimbatore, 641049, Tamil Nadu, India
| | - Sowmiya Sri Sivaramalingam
- Research Center for Excellence in Microscopy, Department of Biotechnology, Kumaraguru College of Technology, Coimbatore, 641049, Tamil Nadu, India
| | - Deepsikha Jothivel
- Research Center for Excellence in Microscopy, Department of Biotechnology, Kumaraguru College of Technology, Coimbatore, 641049, Tamil Nadu, India
| | - Dhivia Dharshika Chithiraiselvan
- Research Center for Excellence in Microscopy, Department of Biotechnology, Kumaraguru College of Technology, Coimbatore, 641049, Tamil Nadu, India
| | | | - Kumaravel Kandaswamy
- Research Center for Excellence in Microscopy, Department of Biotechnology, Kumaraguru College of Technology, Coimbatore, 641049, Tamil Nadu, India
| |
Collapse
|
23
|
Zhang T, Di Carlo D, Lim CT, Zhou T, Tian G, Tang T, Shen AQ, Li W, Li M, Yang Y, Goda K, Yan R, Lei C, Hosokawa Y, Yalikun Y. Passive microfluidic devices for cell separation. Biotechnol Adv 2024; 71:108317. [PMID: 38220118 DOI: 10.1016/j.biotechadv.2024.108317] [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: 11/01/2023] [Revised: 12/27/2023] [Accepted: 01/06/2024] [Indexed: 01/16/2024]
Abstract
The separation of specific cell populations is instrumental in gaining insights into cellular processes, elucidating disease mechanisms, and advancing applications in tissue engineering, regenerative medicine, diagnostics, and cell therapies. Microfluidic methods for cell separation have propelled the field forward, benefitting from miniaturization, advanced fabrication technologies, a profound understanding of fluid dynamics governing particle separation mechanisms, and a surge in interdisciplinary investigations focused on diverse applications. Cell separation methodologies can be categorized according to their underlying separation mechanisms. Passive microfluidic separation systems rely on channel structures and fluidic rheology, obviating the necessity for external force fields to facilitate label-free cell separation. These passive approaches offer a compelling combination of cost-effectiveness and scalability when compared to active methods that depend on external fields to manipulate cells. This review delves into the extensive utilization of passive microfluidic techniques for cell separation, encompassing various strategies such as filtration, sedimentation, adhesion-based techniques, pinched flow fractionation (PFF), deterministic lateral displacement (DLD), inertial microfluidics, hydrophoresis, viscoelastic microfluidics, and hybrid microfluidics. Besides, the review provides an in-depth discussion concerning cell types, separation markers, and the commercialization of these technologies. Subsequently, it outlines the current challenges faced in the field and presents a forward-looking perspective on potential future developments. This work hopes to aid in facilitating the dissemination of knowledge in cell separation, guiding future research, and informing practical applications across diverse scientific disciplines.
Collapse
Affiliation(s)
- Tianlong Zhang
- College of Mechanical Engineering, Jiangsu University of Science and Technology, Zhenjiang 212100, China
| | - Dino Di Carlo
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
| | - Chwee Teck Lim
- Department of Biomedical Engineering, National University of Singapore, Singapore 117583, Singapore
| | - Tianyuan Zhou
- College of Mechanical Engineering, Jiangsu University of Science and Technology, Zhenjiang 212100, China
| | - Guizhong Tian
- College of Mechanical Engineering, Jiangsu University of Science and Technology, Zhenjiang 212100, China.
| | - Tao Tang
- Department of Biomedical Engineering, National University of Singapore, Singapore 117583, Singapore
| | - Amy Q Shen
- Micro/Bio/Nanofluidics Unit, Okinawa Institute of Science and Technology Graduate University, Onna-son, Okinawa 904-0495, Japan
| | - Weihua Li
- School of Mechanical, Materials, Mechatronic and Biomedical Engineering, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Ming Li
- School of Mechanical and Manufacturing Engineering, University of New South Wales, Sydney, NSW 2052, Australia
| | - Yang Yang
- Institute of Deep-Sea Science and Engineering, Chinese Academy of Sciences, Sanya, Hainan 572000, China
| | - Keisuke Goda
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA; Department of Chemistry, University of Tokyo, Tokyo 113-0033, Japan; The Institute of Technological Sciences, Wuhan University, Wuhan 430072, China
| | - Ruopeng Yan
- The Institute of Technological Sciences, Wuhan University, Wuhan 430072, China
| | - Cheng Lei
- The Institute of Technological Sciences, Wuhan University, Wuhan 430072, China
| | - Yoichiroh Hosokawa
- Division of Materials Science, Nara Institute of Science and Technology, Nara 630-0192, Japan
| | - Yaxiaer Yalikun
- Division of Materials Science, Nara Institute of Science and Technology, Nara 630-0192, Japan.
| |
Collapse
|
24
|
Tankova HI, Lazarova ZB, Mitova NG. Dynamics of Gingival Indices and Microbiological Findings During Treatment of Plaque-Induced Gingivitis in Children Aged 10-14 Years. Niger J Clin Pract 2024; 27:361-367. [PMID: 38528357 DOI: 10.4103/njcp.njcp_757_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 02/07/2024] [Indexed: 03/27/2024]
Abstract
BACKGROUND The study analyzed the dynamics of the clinical periodontal status during the treatment of adolescents with generalized plaque-induced gingivitis. AIM Assessment of the predominant subgingival microflora in the case of a diagnosed inflammatory process in the gingiva in childhood. METHODS Full-mouth periodontal assessment of plaque accumulation and bleeding on probing with an electronic periodontal probe was performed during the treatment of 34 adolescents with generalized plaque-induced gingivitis. The treatment protocol includes five visits (1, 3, 7, 14, and 30 days). Subgingival biofilm sampling was performed by real-time PCR testing to identify, follow-up in dynamics, and determine the quantities of main subgingival periodontopathogens during treatment. Three samples per child were taken from five teeth with the most severe inflammation. RESULTS For children aged 10-14 years with generalized plaque-induced gingivitis, two weeks after the start of treatment, the index values for bleeding on probing decreased twice from 53 to 27%. C. gingivalis was isolated before the start of treatment in all children, followed by P. intermedia, P. micros (70,4%) and T. denticola, T. forsythia (52,9%). Representatives of the red complex according to Socransky showing greater resistance to the therapy performed in terms of frequency and amount. CONCLUSION The predominant subgingival microflora in adolescents with generalized plaque-induced gingivitis is representative of the orange and red Socransky complex, with index values decreasing smoothly at each subsequent visit during treatment.
Collapse
Affiliation(s)
- H I Tankova
- Department of Pediatric Dental Medicine, Faculty of Dental Medicine, Medical University, Sofia, Bulgaria
| | | | | |
Collapse
|
25
|
Choi A, Dong K, Williams E, Pia L, Batagower J, Bending P, Shin I, Peters DI, Kaspar JR. Human saliva modifies growth, biofilm architecture, and competitive behaviors of oral streptococci. mSphere 2024; 9:e0077123. [PMID: 38319113 PMCID: PMC10900908 DOI: 10.1128/msphere.00771-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 01/08/2024] [Indexed: 02/07/2024] Open
Abstract
The bacteria within supragingival biofilms participate in complex exchanges with other microbes inhabiting the same niche. One example is the mutans group streptococci (Streptococcus mutans), implicated in the development of tooth decay, and other health-associated commensal streptococci species. Previously, our group transcriptomically characterized intermicrobial interactions between S. mutans and several species of oral bacteria. However, these experiments were carried out in a medium without human saliva. To better mimic their natural environment, we first evaluated how inclusion of saliva affected growth and biofilm formation of eight Streptococcus species individually and found saliva to positively benefit growth rates while negatively influencing biofilm biomass accumulation and altering spatial arrangement. These results carried over during evaluation of 29 saliva-derived isolates of various species. Surprisingly, we also found that addition of saliva increased the competitive behaviors of S. mutans in coculture competitions against commensal streptococci that led to increases in biofilm microcolony volumes. Through transcriptomically characterizing mono- and cocultures of S. mutans and Streptococcus oralis with and without saliva, we determined that each species developed a nutritional niche under mixed-species growth, with S. mutans upregulating carbohydrate uptake and utilization pathways while S. oralis upregulated genome features related to peptide uptake and glycan foraging. S. mutans also upregulated genes involved in oxidative stress tolerance, particularly manganese uptake, which we could artificially manipulate by supplementing in manganese leading to an advantage over its opponent. Our report highlights observable changes in microbial behaviors through leveraging environmental- and host-supplied resources over their competitors. IMPORTANCE Dental caries (tooth decay) is the most prevalent disease for both children and adults nationwide. Caries are initiated from demineralization of the enamel due to organic acid production through the metabolic activity of oral bacteria growing in biofilm communities attached to the tooth's surface. Mutans group streptococci are closely associated with caries development and initiation of the cariogenic cycle, which decreases the amount of acid-sensitive, health-associated commensal bacteria while selecting for aciduric and acidogenic species that then further drives the disease process. Defining the exchanges that occur between mutans group streptococci and oral commensals in a condition that closely mimics their natural environment is of critical need toward identifying factors that can influence odontopathogen establishment, persistence, and outgrowth. The goal of our research is to develop strategies, potentially through manipulation of microbial interactions characterized here, that prevent the emergence of mutans group streptococci while keeping the protective flora intact.
Collapse
Affiliation(s)
- Allen Choi
- Division of Biosciences, The Ohio State University College of Dentistry, Columbus, Ohio, USA
| | - Kevin Dong
- Division of Biosciences, The Ohio State University College of Dentistry, Columbus, Ohio, USA
| | - Emily Williams
- Division of Biosciences, The Ohio State University College of Dentistry, Columbus, Ohio, USA
| | - Lindsey Pia
- Division of Biosciences, The Ohio State University College of Dentistry, Columbus, Ohio, USA
| | - Jordan Batagower
- Division of Biosciences, The Ohio State University College of Dentistry, Columbus, Ohio, USA
| | - Paige Bending
- Division of Biosciences, The Ohio State University College of Dentistry, Columbus, Ohio, USA
| | - Iris Shin
- Division of Biosciences, The Ohio State University College of Dentistry, Columbus, Ohio, USA
| | - Daniel I. Peters
- Division of Biosciences, The Ohio State University College of Dentistry, Columbus, Ohio, USA
| | - Justin R. Kaspar
- Division of Biosciences, The Ohio State University College of Dentistry, Columbus, Ohio, USA
| |
Collapse
|
26
|
Loewe MF, Doll-Nikutta K, Stiesch M, Schwestka-Polly R. Biofilm volume and acidification within initial biofilms formed in situ on buccally and palatally exposed bracket material. J Orofac Orthop 2024:10.1007/s00056-024-00515-4. [PMID: 38409443 DOI: 10.1007/s00056-024-00515-4] [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: 03/17/2023] [Accepted: 12/22/2023] [Indexed: 02/28/2024]
Abstract
PURPOSE Acidification by bacterial biofilms at the bracket/tooth interface is one of the most common problems in fixed orthodontic treatments, which can lead to white spot lesions (WSL) and caries. As lingual brackets were shown to exhibit reduced WSL formation clinically, the aim of this in situ study was to compare initial intraoral biofilm formation and acidification on bracket-like specimens placed buccally and palatally in the upper jaw as a possible cause for this observation. METHODS Intraoral biofilm was collected from splints equipped with buccally and palatally exposed test specimens, which were worn by 12 volunteers for a total of 48 h. The test specimens consisted of standard bracket material cylinders on top of a hydroxyapatite disc to represent the bracket/tooth interface. They were analyzed for three-dimensional biofilm volume and live/dead distribution by fluorescence staining and confocal laser scanning microscopy as well as for acidification by fluorescence-based pH ratiometry. RESULTS Similar general biofilm morphology with regard to volume and viability could be detected for buccally and palatally exposed specimens. For pH values, biofilms from both positions showed increased acidification at the bottom layer. Interestingly, the pH value at the top layers of the biofilms was slightly lower on palatally than on buccally exposed specimens, which may likely be due to anatomic conditions. CONCLUSION Based on the results of this study, initial intraoral biofilm formation and acidification is almost similar on the bracket material/biomimetic tooth interface when placed buccally or palatally in the upper jaw. As lingual brackets were shown to exhibit reduced WSL formation clinically, future studies should investigate further factors like bracket geometry.
Collapse
Affiliation(s)
- Micha Frederic Loewe
- Department of Prosthetic Dentistry and Biomedical Materials Science, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), Stadtfelddamm 34, 30625, Hannover, Germany.
| | - Katharina Doll-Nikutta
- Department of Prosthetic Dentistry and Biomedical Materials Science, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), Stadtfelddamm 34, 30625, Hannover, Germany
| | - Meike Stiesch
- Department of Prosthetic Dentistry and Biomedical Materials Science, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
- Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), Stadtfelddamm 34, 30625, Hannover, Germany
| | - Rainer Schwestka-Polly
- Department of Orthodontics, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| |
Collapse
|
27
|
Smirnov A, Yanushevich O, Krikheli N, Solis Pinargote NW, Peretyagin P, Grigoriev S, Alou L, Sevillano D, López-Piriz R, Guitian F, Bartolomé JF. 3Y-TZP/Ta Biocermet as a Dental Material: An Analysis of the In Vitro Adherence of Streptococcus Oralis Biofilm and an In Vivo Pilot Study in Dogs. Antibiotics (Basel) 2024; 13:175. [PMID: 38391561 PMCID: PMC10886202 DOI: 10.3390/antibiotics13020175] [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: 11/24/2023] [Revised: 01/31/2024] [Accepted: 02/05/2024] [Indexed: 02/24/2024] Open
Abstract
The surface adhesion of bacterial cells and the in vivo biocompatibility of a new ceramic-metal composite made of zirconium dioxide and tantalum were evaluated. Within the framework of an in vitro study using the crystal violet staining and colony counting methods, a relatively similar adhesion of Streptococcus oralis to the 3Y-TZP/Ta biocermet (roughness Ra = 0.12 ± 0.04 µm) and Ti-Al6-V4 titanium alloy (Ra = 0.04 ± 0.01 µm) was found. In addition, in an in vivo preliminary study focused on the histological analysis of a series of rods implanted in the jaws of beagle dogs for a six-month period, the absence of any fibrous tissue or inflammatory reaction at the interface between the implanted 3Y-TZP/Ta biocermets and the new bone was found. Thus, it can be concluded that the developed ceramic-metal biocomposite may be a promising new material for use in dentistry.
Collapse
Affiliation(s)
- Anton Smirnov
- Spark Plasma Sintering Research Laboratory, Moscow State University of Technology "STANKIN", Vadkovsky per. 1, Moscow 127055, Russia
| | - Oleg Yanushevich
- Scientific Department, A.I. Evdokimov Moscow State University of Medicine and Dentistry, Delegatskaya St., 20, p. 1, Moscow 127473, Russia
| | - Natella Krikheli
- Scientific Department, A.I. Evdokimov Moscow State University of Medicine and Dentistry, Delegatskaya St., 20, p. 1, Moscow 127473, Russia
| | - Nestor Washington Solis Pinargote
- Spark Plasma Sintering Research Laboratory, Moscow State University of Technology "STANKIN", Vadkovsky per. 1, Moscow 127055, Russia
| | - Pavel Peretyagin
- Spark Plasma Sintering Research Laboratory, Moscow State University of Technology "STANKIN", Vadkovsky per. 1, Moscow 127055, Russia
- Scientific Department, A.I. Evdokimov Moscow State University of Medicine and Dentistry, Delegatskaya St., 20, p. 1, Moscow 127473, Russia
| | - Sergey Grigoriev
- Spark Plasma Sintering Research Laboratory, Moscow State University of Technology "STANKIN", Vadkovsky per. 1, Moscow 127055, Russia
| | - Luis Alou
- Microbiology Department, School of Medicine, Universidad Complutense, Avda. Complutense s/n, 28040 Madrid, Spain
| | - David Sevillano
- Microbiology Department, School of Medicine, Universidad Complutense, Avda. Complutense s/n, 28040 Madrid, Spain
| | - Roberto López-Piriz
- Instituto de Cirugía Oral Avanzada-ICOA, Calle de Fray Luis de León, 14, 28012 Madrid, Spain
| | - Francisco Guitian
- Instituto de Materiales, iMATUS-USC, Santiago de Compostela, Avenida do Mestre Mateo 25, 15782 La Coruña, Spain
| | - José Florindo Bartolomé
- Instituto de Ciencia de Materiales de Madrid (ICMM), Consejo Superior de Investigaciones Científicas (CSIC), Campus de Cantoblanco, Calle Sor Juana Inés de la Cruz 3, 28049 Madrid, Spain
| |
Collapse
|
28
|
Laugisch O, Ruppert-Jungck MC, Auschill TM, Eick S, Sculean A, Heumann C, Timmermann L, Pedrosa DJ, Eggers C, Arweiler NB. Glucose-6-Phosphatase-Dehydrogenase activity as modulative association between Parkinson's disease and periodontitis. Front Cell Infect Microbiol 2024; 14:1298546. [PMID: 38404290 PMCID: PMC10885135 DOI: 10.3389/fcimb.2024.1298546] [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: 09/21/2023] [Accepted: 01/12/2024] [Indexed: 02/27/2024] Open
Abstract
The association between periodontitis (PD) and Parkinson's disease (PK) is discussed due to the inflammatory component of neurodegenerative processes. PK severity and affected areas were determined using the following neuropsychological tests: Unified Parkinson's Disease Rating Score (UPDRS) and Hoehn and Yahr; non-motoric symptoms by Non-Motor Symptoms Scale (NMSS), and cognitive involvement by Mini-Mental State Examination (MMSE). Neuroinflammation and the resulting Glucose-6-Phosphatase-Dehydrogenase (G6PD) dysfunction are part of the pathophysiology of PK. This study aimed to evaluate these associations in periodontal inflammation. Clinical data and saliva-, serum-, and RNA-biobank samples of 50 well-characterized diametric patients with PK and five age- and sex-matched neurologically healthy participants were analyzed for G6PD function, periodontal pathogens (Aggregatibacter actinomycetemcomitans, Porphyromonas gingivalis, Tannerella forsythia, Treponema denticola, Prevotella intermedia, Campylobacter rectus, Fusobacterium nucleatum, and Filifactor alocis), monocyte chemoattractant protein (MCP) 1, and interleukin (IL) 1-beta. Regression analysis was used to identify associations between clinical and behavioral data, and t-tests were used to compare health and disease. Compared with PK, no pathogens and lower inflammatory markers (p < 0.001) were detectible in healthy saliva and serum, PK-severity/UPDRS interrelated with the occurrence of Prevotella intermedia in serum as well as IL1-beta levels in serum and saliva (p = 0.006, 0.019, 0.034), Hoehn and Yahr correlated with Porphyromonas gingivalis, Prevotella intermedia, RNA IL1-beta regulation, serum, and saliva IL1-beta levels, with p-values of 0.038, 0.011, 0.008, <0.001, and 0.010, while MMSE was associated with Aggregatibacter actinomycetemcomitans, Fusobacterium nucleatum, serum MCP 1 levels, RNA IL1-beta regulation and G6PD serum activity (p = 0.036, 0.003, 0.045, <0.001, and 0.021). Cognitive and motor skills seem to be important as representative tests are associated with periodontal pathogens and oral/general inflammation, wherein G6PD-saliva dysfunction might be involved. Clinical trial registration https://www.bfarm.de/DE/Das-BfArM/Aufgaben/Deutsches-Register-Klinischer-Studien/_node.html, identifier DRKS00005388.
Collapse
Affiliation(s)
- Oliver Laugisch
- Department of Periodontology and Peri-Implant Diseases, Universitätsklinikum Giessen und Marburg (UKGM), Philipps University, Marburg, Germany
| | - Marina C. Ruppert-Jungck
- Department of Neurology, University Hospital Giessen and Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), Universities of Giessen and Marburg, Marburg, Germany
| | - Thorsten M. Auschill
- Department of Periodontology and Peri-Implant Diseases, Universitätsklinikum Giessen und Marburg (UKGM), Philipps University, Marburg, Germany
| | - Sigrun Eick
- Department of Periodontology, School of Dental Medicine, University of Bern, Bern, Switzerland
| | - Anton Sculean
- Department of Periodontology, School of Dental Medicine, University of Bern, Bern, Switzerland
| | - Christian Heumann
- Department of Statistics, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Lars Timmermann
- Department of Neurology, University Hospital Giessen and Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), Universities of Giessen and Marburg, Marburg, Germany
| | - David J. Pedrosa
- Department of Neurology, University Hospital Giessen and Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), Universities of Giessen and Marburg, Marburg, Germany
| | - Carsten Eggers
- Department of Neurology, University Hospital Giessen and Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), Universities of Giessen and Marburg, Marburg, Germany
- Department of Neurology, Knappschaftskrankenhaus Bottrop, Bottrop, Germany
| | - Nicole B. Arweiler
- Department of Periodontology and Peri-Implant Diseases, Universitätsklinikum Giessen und Marburg (UKGM), Philipps University, Marburg, Germany
| |
Collapse
|
29
|
Blochberger BL, Symmank J, Nitzsche Á, Nietzsche S, Steiniger F, Guellmar A, Reise M, Sigusch B, Jacobs C, Hennig CL. Influence of the orthodontic bonding procedure on biofilm formation. Orthod Craniofac Res 2024; 27:95-101. [PMID: 37470303 DOI: 10.1111/ocr.12692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 07/06/2023] [Accepted: 07/07/2023] [Indexed: 07/21/2023]
Abstract
INTRODUCTION In orthodontics, white spot lesions are a persistent and widespread problem caused by the demineralization of buccal tooth surfaces around bonded brackets. The remaining adhesive around the brackets leads to surface roughness, which might contribute to demineralization. The present in vitro study aimed to compare a conventional and a modern adhesive system (APC Flash-Free technology) for orthodontic brackets with regard to the adhesion of Streptococcus sobrinus, a leading caries pathogen. METHODS This in vitro study included 20 premolar teeth and compared 10 APC Flash-Free adhesive-coated ceramic brackets (FF)with 10 conventionally bonded (CB) ceramic clarity brackets. Specimens were incubated in an S. sobrinus suspension for 3 h. To evaluate the bacterial formation, samples were analysed with a scanning electron microscope (SEM). Imaging software was used to quantify and statistically compare percentage values of colonization (PVC) in both groups' adhesion and transition areas. RESULTS We found a significant difference in biofilm formation between the groups for the adhesive and transition areas. PVC in the adhesive area was approximately 10.3-fold greater for the CB group compared with the FF group (median: 3.2 vs 0.31; P < 0.0001). For the transition area, median PVC was approximately 2.4-fold greater for the CB group compared with the FF group (median: 53.17 vs 22.11; P < 0.01). CONCLUSIONS There was a significantly lower level of S. sobrinus formation around the FF bracket system than there was surrounding the conventionally bonded group. This study suggests that the FF adhesive bracket system can help reduce the occurrence of bacterial growth around orthodontic brackets.
Collapse
Affiliation(s)
| | - Judit Symmank
- Department of Orthodontics, Center of Dental Medicine, University Hospital Jena, Jena, Germany
| | - Ánn Nitzsche
- Department of Orthodontics, Center of Dental Medicine, University Hospital Jena, Jena, Germany
| | - Sandor Nietzsche
- Electron Microscopy Center, University Hospital Jena, Jena, Germany
| | - Frank Steiniger
- Electron Microscopy Center, University Hospital Jena, Jena, Germany
| | - André Guellmar
- Department of Conservative Dentistry and Periodontology, Center of Dental Medicine, University Hospital Jena, Jena, Germany
| | - Markus Reise
- Department of Conservative Dentistry and Periodontology, Center of Dental Medicine, University Hospital Jena, Jena, Germany
| | - Bernd Sigusch
- Department of Conservative Dentistry and Periodontology, Center of Dental Medicine, University Hospital Jena, Jena, Germany
| | - Collin Jacobs
- Department of Orthodontics, Center of Dental Medicine, University Hospital Jena, Jena, Germany
| | - Christoph-Ludwig Hennig
- Department of Orthodontics, Center of Dental Medicine, University Hospital Jena, Jena, Germany
| |
Collapse
|
30
|
Spatafora G, Li Y, He X, Cowan A, Tanner ACR. The Evolving Microbiome of Dental Caries. Microorganisms 2024; 12:121. [PMID: 38257948 PMCID: PMC10819217 DOI: 10.3390/microorganisms12010121] [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: 12/03/2023] [Revised: 12/28/2023] [Accepted: 01/04/2024] [Indexed: 01/24/2024] Open
Abstract
Dental caries is a significant oral and public health problem worldwide, especially in low-income populations. The risk of dental caries increases with frequent intake of dietary carbohydrates, including sugars, leading to increased acidity and disruption of the symbiotic diverse and complex microbial community of health. Excess acid production leads to a dysbiotic shift in the bacterial biofilm composition, demineralization of tooth structure, and cavities. Highly acidic and acid-tolerant species associated with caries include Streptococcus mutans, Lactobacillus, Actinomyces, Bifidobacterium, and Scardovia species. The differences in microbiotas depend on tooth site, extent of carious lesions, and rate of disease progression. Metagenomics and metatranscriptomics not only reveal the structure and genetic potential of the caries-associated microbiome, but, more importantly, capture the genetic makeup of the metabolically active microbiome in lesion sites. Due to its multifactorial nature, caries has been difficult to prevent. The use of topical fluoride has had a significant impact on reducing caries in clinical settings, but the approach is costly; the results are less sustainable for high-caries-risk individuals, especially children. Developing treatment regimens that specifically target S. mutans and other acidogenic bacteria, such as using nanoparticles, show promise in altering the cariogenic microbiome, thereby combatting the disease.
Collapse
Affiliation(s)
- Grace Spatafora
- Biology and Program in Molecular Biology and Biochemistry, Middlebury College, Middlebury, VT 05753, USA
| | - Yihong Li
- Department of Public and Ecosystem Health, Cornell University, Ithaca, NY 14853, USA;
| | - Xuesong He
- ADA-Forsyth Institute, Cambridge, MA 02142, USA;
| | - Annie Cowan
- The Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | | |
Collapse
|
31
|
El Zawawy NA, El-Safty S, Kenawy ER, Ibrahim Salem S, Ali SS, Mahmoud YAG. Exploring the biomedical potential of a novel modified glass ionomer cement against the pandrug-resistant oral pathogen Candida albicans SYN-01. J Oral Microbiol 2023; 15:2195741. [PMID: 37008537 PMCID: PMC10064826 DOI: 10.1080/20002297.2023.2195741] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2023] Open
Abstract
Dental caries is an infectious disease that is a major concern for dentists. Streptococci and Lactobacilli were long thought to be the primary etiology responsible for caries. Candida albicans with acidogenic and aciduric characteristics has recently been implicated in the onset and progression of cariogenic lesions. Moreover, due to the increased resistance to common antimicrobials, the discovery of innovative candidates is in high demand. Therefore, our study might be the first report that explores the efficacy of glass ionomer cement (GIC) incorporated with a newly modified carboxylated chitosan derivative (CS-MC) against multidrug-resistant (MDR) and/or pandrug resistant (PDR) C. albicans isolated from the oral cavity. In this work, four CS-MC-GIC groups with different concentrations were formulated. Group four (CS-MC-GIC-4) gave a significant performance as an anticandidal agent against selected PDR Candida strain, with an obvious decrease in its cell viability and high antibiofilm activity. It also, enhanced all the mechanical properties and supports cell viability of Vero cells as a nontoxic compound. Moreover, CS-MC-GIC-4 inhibited neuraminidases completely, which might provide a novel mechanism to prevent dental/oral infections. Thus, findings in this study open up new prospect of the utilization of CS-MC-GIC as a novel dental filling material against oral drug-resistant Candida.
![]() ![]()
Collapse
Affiliation(s)
- Nessma A. El Zawawy
- Botany Department, Faculty of Science Tanta University, Tanta, Egypt
- CONTACT Nessma A. El Zawawy Botany Department, Faculty of Science, Tanta University, Tanta31527, Egypt
| | - Samy El-Safty
- Biomaterials Department, Faculty of Dentistry, Tanta University, Tanta, Egypt
| | - El-Refaie Kenawy
- Polymer Research Group, Department of Chemistry, Faculty of Science Tanta University, Tanta, Egypt
| | - Sara Ibrahim Salem
- Polymer Research Group, Department of Chemistry, Faculty of Science Tanta University, Tanta, Egypt
| | - Sameh S. Ali
- Botany Department, Faculty of Science Tanta University, Tanta, Egypt
| | | |
Collapse
|
32
|
Li S, Li H, Kong H, Wu SY, Cheng CK, Xu J. Endogenous and microbial biomarkers for periodontitis and type 2 diabetes mellitus. Front Endocrinol (Lausanne) 2023; 14:1292596. [PMID: 38149100 PMCID: PMC10750125 DOI: 10.3389/fendo.2023.1292596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 11/20/2023] [Indexed: 12/28/2023] Open
Abstract
It has been well documented that there is a two-way relationship between diabetes mellitus and periodontitis. Diabetes mellitus represents an established risk factor for chronic periodontitis. Conversely, chronic periodontitis adversely modulates serum glucose levels in diabetic patients. Activated immune and inflammatory responses are noted during diabetes and periodontitis, under the modulation of similar biological mediators. These activated responses result in increased activity of certain immune-inflammatory mediators including adipokines and microRNAs in diabetic patients with periodontal disease. Notably, certain microbes in the oral cavity were identified to be involved in the occurrence of diabetes and periodontitis. In other words, these immune-inflammatory mediators and microbes may potentially serve as biomarkers for risk assessment and therapy selection in diabetes and periodontitis. In this review, we briefly provide an updated overview on different potential biomarkers, providing novel diagnostic and therapeutic insights on periodontal complications and diabetes mellitus.
Collapse
Affiliation(s)
- Songjun Li
- Longgang Ear-Nose-Throat (ENT) Hospital, Institute of Ear-Nose-Throat (ENT) and Shenzhen Key Laboratory of Ear-Nose-Throat (ENT), Shenzhen, China
| | - Hongwen Li
- Longgang Ear-Nose-Throat (ENT) Hospital, Institute of Ear-Nose-Throat (ENT) and Shenzhen Key Laboratory of Ear-Nose-Throat (ENT), Shenzhen, China
- Shenzhen Longgang Institute of Stomatology, Longgang Ear-Nose-Throat (ENT) Hospital, Shenzhen, China
| | - Haiying Kong
- Longgang Ear-Nose-Throat (ENT) Hospital, Institute of Ear-Nose-Throat (ENT) and Shenzhen Key Laboratory of Ear-Nose-Throat (ENT), Shenzhen, China
| | - Shang Ying Wu
- Department of Laboratory Medicine, Shenzhen Hospital, Peking University, Shenzhen, China
| | - Chak Kwong Cheng
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, China
| | - Jian Xu
- Longgang Ear-Nose-Throat (ENT) Hospital, Institute of Ear-Nose-Throat (ENT) and Shenzhen Key Laboratory of Ear-Nose-Throat (ENT), Shenzhen, China
- Shenzhen Longgang Institute of Stomatology, Longgang Ear-Nose-Throat (ENT) Hospital, Shenzhen, China
| |
Collapse
|
33
|
Etebarian A, Sheshpari T, Kabir K, Sadeghi H, Moradi A, Hafedi A. Oral Lactobacillus species and their probiotic capabilities in patients with periodontitis and periodontally healthy individuals. Clin Exp Dent Res 2023; 9:746-756. [PMID: 37078410 PMCID: PMC10582226 DOI: 10.1002/cre2.740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 03/16/2023] [Accepted: 03/31/2023] [Indexed: 04/21/2023] Open
Abstract
OBJECTIVES This study aimed to identify oral Lactobacillus species and characterize their adhesion properties and antibacterial activity in patients with periodontitis compared with periodontally healthy individuals. MATERIALS AND METHODS Three hundred and fifty-four isolates from the saliva, subgingival, and tongue plaque of 59 periodontitis patients and 59 healthy individuals were analyzed. Oral Lactobacillus species were identified through the culture method in the modified MRS medium and confirmed by molecular testing. Moreover, the radial diffusion assay and cell culture methods were used to determine the antibacterial activities of oral strains against oral pathogens and their adhesion activity in vitro. RESULTS 67.7% of the cases and 75.7% of the control samples were positive for the Lactobacillus species. Lacticaseibacillus paracasei and Limosilactobacillus fermentum were the dominant species in the case group, whereas Lacticaseibacillus casei and Lactiplantibacillus plantarum were dominant in the control group. Lactobacillus crispatus and Lactobacillus gasseri had higher antibacterial effects against oral pathogens. Moreover, Ligilactobacillus salivarius and L. fermentum demonstrated the highest ability to adhere to oral mucosal cells and salivary-coated hydroxyapatite. CONCLUSION L. crispatus, L. gasseri, L. fermentum, and L. salivarius can be introduced as probiotic candidates since they demonstrated appropriate adherence to oral mucosal cells and salivary-coated hydroxyapatite and also antibacterial activities. However, further studies should be conducted to assess the safety of probiotic interventions using these strains in patients with periodontal disease.
Collapse
Affiliation(s)
- Arghavan Etebarian
- Oral and Maxillofacial Pathology Department, School of DentistryAlborz University of Medical SciencesKarajIran
| | - Tahere Sheshpari
- Microbiology Department, School of MedicineAlborz University of Medical SciencesKarajIran
| | - Kourosh Kabir
- Community Medicine Department, Dietary Supplements and Probiotic Research CenterAlborz University of Medical SciencesKarajIran
| | - Hanieh Sadeghi
- Student Research CommitteeAlborz University of Medical SciencesKarajIran
| | - Abouzar Moradi
- Periodontology Department, School of DentistryAlborz University of Medical SciencesKarajIran
| | - Avin Hafedi
- Student Research CommitteeAlborz University of Medical SciencesKarajIran
| |
Collapse
|
34
|
Lamont RJ, Hajishengallis G, Koo H. Social networking at the microbiome-host interface. Infect Immun 2023; 91:e0012423. [PMID: 37594277 PMCID: PMC10501221 DOI: 10.1128/iai.00124-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/19/2023] Open
Abstract
Microbial species colonizing host ecosystems in health or disease rarely do so alone. Organisms conglomerate into dynamic heterotypic communities or biofilms in which interspecies and interkingdom interactions drive functional specialization of constituent species and shape community properties, including nososymbiocity or pathogenic potential. Cell-to-cell binding, exchange of signaling molecules, and nutritional codependencies can all contribute to the emergent properties of these communities. Spatial constraints defined by community architecture also determine overall community function. Multilayered interactions thus occur between individual pairs of organisms, and the relative impact can be determined by contextual cues. Host responses to heterotypic communities and impact on host surfaces are also driven by the collective action of the community. Additionally, the range of interspecies interactions can be extended by bacteria utilizing host cells or host diet to indirectly or directly influence the properties of other organisms and the community microenvironment. In contexts where communities transition to a dysbiotic state, their quasi-organismal nature imparts adaptability to nutritional availability and facilitates resistance to immune effectors and, moreover, exploits inflammatory and acidic microenvironments for their persistence.
Collapse
Affiliation(s)
- Richard J. Lamont
- Department of Oral Immunology and Infectious Diseases, School of Dentistry, University of Louisville, Louisville, Kentucky, USA
| | - George Hajishengallis
- Department of Basic and Translational Sciences, Laboratory of Innate Immunity and Inflammation, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Hyun Koo
- Department of Orthodontics and Divisions of Pediatric Dentistry and Community Oral Health, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Biofilm Research Laboratories, Center for Innovation & Precision Dentistry, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
35
|
Johnston W, Rosier BT, Carda-Diéguez M, Paterson M, Watson P, Piela K, Goulding M, Ramage G, Baranyia D, Chen T, Al-Hebshi NN, Mira A, Culshaw S. Longitudinal changes in subgingival biofilm composition following periodontal treatment. J Periodontol 2023; 94:1065-1077. [PMID: 36960491 DOI: 10.1002/jper.22-0749] [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: 12/22/2022] [Revised: 02/22/2023] [Accepted: 03/10/2023] [Indexed: 03/25/2023]
Abstract
BACKGROUND Current periodontal treatment involves instrumentation using hand and/or ultrasonic instruments, which are used either alone or in combination based on patient and clinician preference, with comparable clinical outcomes. This study sought to investigate early and later changes in the subgingival biofilm following periodontal treatment, to identify whether these changes were associated with treatment outcomes, and to investigate whether the biofilm responded differently to hand compared with ultrasonic instruments. METHODS This was a secondary-outcome analysis of a randomized-controlled trial. Thirty-eight periodontitis patients received full-mouth subgingival instrumentation using hand (n = 20) or ultrasonic instrumentation (n = 18). Subgingival plaque was sampled at baseline and 1, 7, and 90 days following treatment. Bacterial DNA was analyzed using 16S rRNA sequencing. Periodontal clinical parameters were evaluated before and after treatment. RESULTS Biofilm composition was comparable in both (hand and ultrasonics) treatment groups at all time points (all genera and species; p[adjusted] > 0.05). Large-scale changes were observed within groups across time points. At days 1 and 7, taxonomic diversity and dysbiosis were reduced, with an increase in health-associated genera including Streptococcus and Rothia equating to 30% to 40% of the relative abundance. When reassessed at day 90 a subset of samples reformed a microbiome more comparable with baseline, which was independent of instrumentation choice and residual disease. CONCLUSIONS Hand and ultrasonic instruments induced comparable impacts on the subgingival plaque microbiome. There were marked early changes in the subgingival biofilm composition, although there was limited evidence that community shifts associated with treatment outcomes.
Collapse
Affiliation(s)
- William Johnston
- Oral Sciences, Dental School, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Bob T Rosier
- Department of Genomics and Health, The Foundation for the Promotion of Health and Biomedical Research (FISABIO), Valencia, Spain
| | - Miguel Carda-Diéguez
- Department of Genomics and Health, The Foundation for the Promotion of Health and Biomedical Research (FISABIO), Valencia, Spain
| | - Michael Paterson
- Oral Sciences, Dental School, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Paddy Watson
- Oral Sciences, Dental School, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Krystyna Piela
- Oral Sciences, Dental School, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- Division of Dentistry, Medical University of Lodz, Lodz, Poland
| | - Marilyn Goulding
- Global Clinical Affairs, Dentsply Sirona, York, Pennsylvania, USA
| | - Gordon Ramage
- Oral Sciences, Dental School, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Divyashri Baranyia
- Department of Oral Health Sciences, Temple University, Philadelphia, Pennsylvania, USA
| | - Tsute Chen
- Department of Microbiology, Forsyth Institute, Cambridge, Massachusetts, USA
| | - Nezar N Al-Hebshi
- Department of Oral Health Sciences, Temple University, Philadelphia, Pennsylvania, USA
| | - Alex Mira
- Department of Genomics and Health, The Foundation for the Promotion of Health and Biomedical Research (FISABIO), Valencia, Spain
- CIBER Center for Epidemiology and Public Health, Madrid, Spain
| | - Shauna Culshaw
- Oral Sciences, Dental School, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- Department of Periodontology, University Center for Dental Medicine, University of Basel, Basel, Switzerland
| |
Collapse
|
36
|
De Angelis F, D’Ercole S, Di Giulio M, Vadini M, Biferi V, Buonvivere M, Vanini L, Cellini L, Di Lodovico S, D’Arcangelo C. In Vitro Evaluation of Candida albicans Adhesion on Heat-Cured Resin-Based Dental Composites. MATERIALS (BASEL, SWITZERLAND) 2023; 16:5818. [PMID: 37687511 PMCID: PMC10488390 DOI: 10.3390/ma16175818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 08/18/2023] [Accepted: 08/22/2023] [Indexed: 09/10/2023]
Abstract
Microbial adhesion on dental restorative materials may jeopardize the restorative treatment long-term outcome. The goal of this in vitro study was to assess Candida albicans capability to adhere and form a biofilm on the surface of heat-cured dental composites having different formulations but subjected to identical surface treatments and polymerization protocols. Three commercially available composites were evaluated: GrandioSO (GR), Venus Diamond (VD) and Enamel Plus HRi Biofunction (BF). Cylindrical specimens were prepared for quantitative determination of C. albicans S5 planktonic CFU count, sessile cells CFU count and biomass optical density (OD570 nm). Qualitative Concanavalin-A assays (for extracellular polymeric substances of a biofilm matrix) and Scanning Electron Microscope (SEM) analyses (for the morphology of sessile colonies) were also performed. Focusing on planktonic CFU count, a slight but not significant reduction was observed with VD as compared to GR. Regarding sessile cells CFU count and biomass OD570 nm, a significant increase was observed for VD compared to GR and BF. Concanavalin-A assays and SEM analyses confirmed the quantitative results. Different formulations of commercially available resin composites may differently interact with C. albicans. The present results showed a relatively more pronounced antiadhesive effect for BF and GR, with a reduction in sessile cells CFU count and biomass quantification.
Collapse
Affiliation(s)
- Francesco De Angelis
- Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University of Chieti–Pescara, 66100 Chieti, Italy; (F.D.A.); (M.V.); (V.B.); (M.B.); (C.D.)
| | - Simonetta D’Ercole
- Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University of Chieti–Pescara, 66100 Chieti, Italy; (F.D.A.); (M.V.); (V.B.); (M.B.); (C.D.)
| | - Mara Di Giulio
- Department of Pharmacy, “G. d’Annunzio” University of Chieti–Pescara, 66100 Chieti, Italy; (M.D.G.); (L.C.); (S.D.L.)
| | - Mirco Vadini
- Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University of Chieti–Pescara, 66100 Chieti, Italy; (F.D.A.); (M.V.); (V.B.); (M.B.); (C.D.)
| | - Virginia Biferi
- Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University of Chieti–Pescara, 66100 Chieti, Italy; (F.D.A.); (M.V.); (V.B.); (M.B.); (C.D.)
| | - Matteo Buonvivere
- Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University of Chieti–Pescara, 66100 Chieti, Italy; (F.D.A.); (M.V.); (V.B.); (M.B.); (C.D.)
| | | | - Luigina Cellini
- Department of Pharmacy, “G. d’Annunzio” University of Chieti–Pescara, 66100 Chieti, Italy; (M.D.G.); (L.C.); (S.D.L.)
| | - Silvia Di Lodovico
- Department of Pharmacy, “G. d’Annunzio” University of Chieti–Pescara, 66100 Chieti, Italy; (M.D.G.); (L.C.); (S.D.L.)
| | - Camillo D’Arcangelo
- Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University of Chieti–Pescara, 66100 Chieti, Italy; (F.D.A.); (M.V.); (V.B.); (M.B.); (C.D.)
| |
Collapse
|
37
|
Tanwar H, Gnanasekaran JM, Allison D, Chuang LS, He X, Aimetti M, Baima G, Costalonga M, Cross RK, Sears C, Mehandru S, Cho J, Colombel JF, Raufman JP, Thumbigere-Math V. Unraveling the Link between Periodontitis and Inflammatory Bowel Disease: Challenges and Outlook. ARXIV 2023:arXiv:2308.10907v1. [PMID: 37645044 PMCID: PMC10462160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Periodontitis and Inflammatory Bowel Disease (IBD) are chronic inflammatory conditions, characterized by microbial dysbiosis and hyper-immunoinflammatory responses. Growing evidence suggest an interconnection between periodontitis and IBD, implying a shift from the traditional concept of independent diseases to a complex, reciprocal cycle. This review outlines the evidence supporting an "Oral-Gut" axis, marked by a higher prevalence of periodontitis in IBD patients and vice versa. The specific mechanisms linking periodontitis and IBD remain to be fully elucidated, but emerging evidence points to the ectopic colonization of the gut by oral bacteria, which promote intestinal inflammation by activating host immune responses. This review presents an in-depth examination of the interconnection between periodontitis and IBD, highlighting the shared microbiological and immunological pathways, and proposing a "multi-hit" hypothesis in the pathogenesis of periodontitis-mediated intestinal inflammation. Furthermore, the review underscores the critical need for a collaborative approach between dentists and gastroenterologists to provide holistic oral-systemic healthcare.
Collapse
Affiliation(s)
- Himanshi Tanwar
- Division of Periodontology, University of Maryland School of Dentistry, Baltimore, MD, USA
| | | | - Devon Allison
- Division of Periodontology, University of Maryland School of Dentistry, Baltimore, MD, USA
| | - Ling-shiang Chuang
- Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Xuesong He
- Department of Microbiology, The Forsyth Institute, Cambridge, MA, USA
| | - Mario Aimetti
- Department of Surgical Sciences, C.I.R. Dental School, University of Turin, Turin, Italy
| | - Giacomo Baima
- Department of Surgical Sciences, C.I.R. Dental School, University of Turin, Turin, Italy
| | - Massimo Costalonga
- Department of Diagnostic and Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, USA
| | - Raymond K. Cross
- Division of Gastroenterology & Hepatology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Cynthia Sears
- Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Saurabh Mehandru
- Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Judy Cho
- Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jean-Frederic Colombel
- Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jean-Pierre Raufman
- Division of Gastroenterology & Hepatology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Vivek Thumbigere-Math
- Division of Periodontology, University of Maryland School of Dentistry, Baltimore, MD, USA
- National Institute of Dental and Craniofacial Research, NIH, Bethesda, MD, USA
| |
Collapse
|
38
|
Irie K, Azuma T, Tomofuji T, Yamamoto T. Exploring the Role of IL-17A in Oral Dysbiosis-Associated Periodontitis and Its Correlation with Systemic Inflammatory Disease. Dent J (Basel) 2023; 11:194. [PMID: 37623290 PMCID: PMC10453731 DOI: 10.3390/dj11080194] [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: 05/16/2023] [Revised: 08/01/2023] [Accepted: 08/09/2023] [Indexed: 08/26/2023] Open
Abstract
Oral microbiota play a pivotal role in maintaining homeostasis, safeguarding the oral cavity, and preventing the onset of disease. Oral dysbiosis has the potential to trigger pro-inflammatory effects and immune dysregulation, which can have a negative impact on systemic health. It is regarded as a key etiological factor for periodontitis. The emergence and persistence of oral dysbiosis have been demonstrated to mediate inflammatory pathology locally and at distant sites. The heightened inflammation observed in oral dysbiosis is dependent upon the secretion of interleukin-17A (IL-17A) by various innate and adaptive immune cells. IL-17A has been found to play a significant role in host defense mechanisms by inducing antibacterial peptides, recruiting neutrophils, and promoting local inflammation via cytokines and chemokines. This review seeks to present the current knowledge on oral dysbiosis and its prevention, as well as the underlying role of IL-17A in periodontitis induced by oral dysbiosis and its impact on systemic inflammatory disease.
Collapse
Affiliation(s)
- Koichiro Irie
- Department of Preventive Dentistry and Dental Public Health, Kanagawa Dental University, Yokosuka 238-8580, Japan;
| | - Tetsuji Azuma
- Department of Community Oral Health, School of Dentistry, Asahi University, Mizuho 501-0296, Japan; (T.A.); (T.T.)
| | - Takaaki Tomofuji
- Department of Community Oral Health, School of Dentistry, Asahi University, Mizuho 501-0296, Japan; (T.A.); (T.T.)
| | - Tatsuo Yamamoto
- Department of Preventive Dentistry and Dental Public Health, Kanagawa Dental University, Yokosuka 238-8580, Japan;
| |
Collapse
|
39
|
Makkar H, Lim CT, Tan KS, Sriram G. Modeling periodontal host-microbe interactions using vascularized gingival connective tissue equivalents. Biofabrication 2023; 15:045008. [PMID: 37473752 DOI: 10.1088/1758-5090/ace935] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 07/20/2023] [Indexed: 07/22/2023]
Abstract
Gingival connective tissue and its vasculature play a crucial role in the host's immune response against the periodontal microbiome and serve as a bridge between the oral and systemic environments. However, there is a lack of representative models that mimic the complex features of vascularized gingival connective tissue and its interaction with the periodontal microbiome, hindering our understanding of periodontal health and disease. Towards this pursuit, we present the characterization of vascularized gingival connective tissue equivalents (CTEs) as a model to study the interactions between oral biofilm colonizers and gingival tissues in healthy and diseased states. Whole-mount immunolabeling and label-free confocal reflectance microscopy of human fibrin-based matrix embedded with gingival fibroblasts and microvascular endothelial cells demonstrated the generation of bi-cellular vascularized gingival CTEs. Next, we investigated the response of the vascularized gingival CTEs to early, intermediate, and late oral biofilm colonizers. Despite colonization, the early colonizers did not elicit any significant change in the production of the cytokines and chemokines by the CTEs representative of the commensal and homeostatic state. In contrast, intermediate and late colonizers representing a transition to a diseased state exhibited connective tissue and vascular invasion, and elicited a differential immune response accompanied by increased monocyte migration. The culture supernatants produced by the vascularized gingival CTEs in response to early and intermediate colonizers polarized macrophages towards an immunomodulatory M2-like phenotype which activates and protects the host, while the late colonizers polarized towards a pro-inflammatory M1-like phenotype. Lastly,in silicoanalysis showed a high strength of associations between the proteins and transcripts investigated with periodontitis and vascular diseases. In conclusion, the vascularized gingival CTEs provide a biomimeticin vitroplatform to study host-microbiome interactions and innate immune response in periodontal health and diseased states, which potentially paves the way toward the development and assessment of novel periodontal therapeutics.
Collapse
Affiliation(s)
- Hardik Makkar
- Faculty of Dentistry, National University of Singapore, Singapore 119085, Singapore
| | - Chwee Teck Lim
- Institute for Health Innovation and Technology (iHealthtech), National University of Singapore, Singapore 117599, Singapore
- Department of Biomedical Engineering, National University of Singapore, Singapore 117583, Singapore
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore
| | - Kai Soo Tan
- Faculty of Dentistry, National University of Singapore, Singapore 119085, Singapore
- ORCHIDS: Oral Care Health Innovations and Designs Singapore, National University of Singapore, Singapore 119085, Singapore
| | - Gopu Sriram
- Faculty of Dentistry, National University of Singapore, Singapore 119085, Singapore
- ORCHIDS: Oral Care Health Innovations and Designs Singapore, National University of Singapore, Singapore 119085, Singapore
| |
Collapse
|
40
|
Iniesta M, Chamorro C, Ambrosio N, Marín MJ, Sanz M, Herrera D. Subgingival microbiome in periodontal health, gingivitis and different stages of periodontitis. J Clin Periodontol 2023; 50:905-920. [PMID: 36792073 DOI: 10.1111/jcpe.13793] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 02/04/2023] [Accepted: 02/10/2023] [Indexed: 02/17/2023]
Abstract
AIM To characterize the subgingival microbiome in subjects with different periodontal health statuses. MATERIALS AND METHODS In this cross-sectional observational study, subgingival samples were harvested from Spanish subjects with different periodontal health statuses, based on the 2018 Classification of Periodontal and Peri-Implant Diseases and Conditions. Samples were processed using high-throughput sequencing technologies (Illumina MiSeq). Taxa differentially abundant were identified using Analysis of Compositions of Microbiomes with Bias Correction (ANCOM-BC). α- and β-diversity metrics were calculated using q2-diversity in QIIME2. The analyses were adjusted for age, gender and smoking status. RESULTS The identified subgingival microbiome showed statistically significant differences among subjects, categorized into periodontal health, gingivitis and stages I-II and III-IV periodontitis (p < .05). In patients with severe (stages III-IV) periodontitis, the genera Filifactor and Fretibacterium were detected 24 times more frequently than in periodontally healthy subjects. Similarly, the genera Porphyromonas, Prevotella and Tannerella were detected four times more frequently (p < .05). The genera Granulicatella, Streptococcus, Paracoccus, Pseudomonas, Haemophilus, Actinobacteria, Bergeyella and Capnocytophaga were significantly associated with healthier periodontal status (p < .05). CONCLUSIONS Significant differences were detected in the subgingival microbiome among periodontal health, gingivitis and stages I-II or III-IV periodontitis, suggesting overlapping, yet distinguishable microbial profiles.
Collapse
Affiliation(s)
- Margarita Iniesta
- ETEP (Etiology and Therapy of Periodontal and Peri-Implant Diseases) Research Group, School of Dentistry, Complutense University of Madrid, Madrid, Spain
| | - Cristina Chamorro
- ETEP (Etiology and Therapy of Periodontal and Peri-Implant Diseases) Research Group, School of Dentistry, Complutense University of Madrid, Madrid, Spain
| | - Nagore Ambrosio
- ETEP (Etiology and Therapy of Periodontal and Peri-Implant Diseases) Research Group, School of Dentistry, Complutense University of Madrid, Madrid, Spain
| | - María José Marín
- ETEP (Etiology and Therapy of Periodontal and Peri-Implant Diseases) Research Group, School of Dentistry, Complutense University of Madrid, Madrid, Spain
| | - Mariano Sanz
- ETEP (Etiology and Therapy of Periodontal and Peri-Implant Diseases) Research Group, School of Dentistry, Complutense University of Madrid, Madrid, Spain
| | - David Herrera
- ETEP (Etiology and Therapy of Periodontal and Peri-Implant Diseases) Research Group, School of Dentistry, Complutense University of Madrid, Madrid, Spain
| |
Collapse
|
41
|
Ahirwar P, Kozlovskaya V, Nijampatnam B, Rojas EM, Pukkanasut P, Inman D, Dolmat M, Law AC, Schormann N, Deivanayagam C, Harber GJ, Michalek SM, Wu H, Kharlampieva E, Velu SE. Hydrogel-Encapsulated Biofilm Inhibitors Abrogate the Cariogenic Activity of Streptococcus mutans. J Med Chem 2023; 66:7909-7925. [PMID: 37285134 PMCID: PMC11188996 DOI: 10.1021/acs.jmedchem.3c00272] [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] [Indexed: 06/08/2023]
Abstract
We designed and synthesized analogues of a previously identified biofilm inhibitor IIIC5 to improve solubility, retain inhibitory activities, and to facilitate encapsulation into pH-responsive hydrogel microparticles. The optimized lead compound HA5 showed improved solubility of 120.09 μg/mL, inhibited Streptococcus mutans biofilm with an IC50 value of 6.42 μM, and did not affect the growth of oral commensal species up to a 15-fold higher concentration. The cocrystal structure of HA5 with GtfB catalytic domain determined at 2.35 Å resolution revealed its active site interactions. The ability of HA5 to inhibit S. mutans Gtfs and to reduce glucan production has been demonstrated. The hydrogel-encapsulated biofilm inhibitor (HEBI), generated by encapsulating HA5 in hydrogel, selectively inhibited S. mutans biofilms like HA5. Treatment of S. mutans-infected rats with HA5 or HEBI resulted in a significant reduction in buccal, sulcal, and proximal dental caries compared to untreated, infected rats.
Collapse
Affiliation(s)
- Parmanand Ahirwar
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Veronika Kozlovskaya
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | - Edwin M. Rojas
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- School of Dentistry, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Piyasuda Pukkanasut
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Daniel Inman
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Maksim Dolmat
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Anna C. Law
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Norbert Schormann
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Champion Deivanayagam
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Gregory J. Harber
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Suzanne M. Michalek
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Hui Wu
- Department of Integrative Biomedical and Diagnostic Sciences, Oregon Health and Science University, Portland, OR 97239, USA
| | - Eugenia Kharlampieva
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Center of Nanoscale Materials and Biointegration, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Sadanandan E. Velu
- Department of Chemistry, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Microbiome Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
42
|
Abdulkareem AA, Al-Taweel FB, Al-Sharqi AJ, Gul SS, Sha A, Chapple IL. Current concepts in the pathogenesis of periodontitis: from symbiosis to dysbiosis. J Oral Microbiol 2023; 15:2197779. [PMID: 37025387 PMCID: PMC10071981 DOI: 10.1080/20002297.2023.2197779] [Citation(s) in RCA: 72] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 03/28/2023] [Indexed: 04/05/2023] Open
Abstract
The primary etiological agent for the initiation and progression of periodontal disease is the dental plaque biofilm which is an organized aggregation of microorganisms residing within a complex intercellular matrix. The non-specific plaque hypothesis was the first attempt to explain the role of the dental biofilm in the pathogenesis of periodontal diseases. However, the introduction of sophisticated diagnostic and laboratory assays has led to the realisation that the development of periodontitis requires more than a mere increase in the biomass of dental plaque. Indeed, multispecies biofilms exhibit complex interactions between the bacteria and the host. In addition, not all resident microorganisms within the biofilm are pathogenic, since beneficial bacteria exist that serve to maintain a symbiotic relationship between the plaque microbiome and the host's immune-inflammatory response, preventing the emergence of pathogenic microorganisms and the development of dysbiosis. This review aims to highlight the development and structure of the dental plaque biofilm and to explore current literature on the transition from a healthy (symbiotic) to a diseased (dysbiotic) biofilm in periodontitis and the associated immune-inflammatory responses that drive periodontal tissue destruction and form mechanistic pathways that impact other systemic non-communicable diseases.
Collapse
Affiliation(s)
- Ali A. Abdulkareem
- Department of Periodontics, College of Dentistry, University of Baghdad, Baghdad, Iraq
| | - Firas B. Al-Taweel
- Department of Periodontics, College of Dentistry, University of Baghdad, Baghdad, Iraq
| | - Ali J.B. Al-Sharqi
- Department of Periodontics, College of Dentistry, University of Baghdad, Baghdad, Iraq
| | - Sarhang S. Gul
- College of Dentistry, University of Sulaimani, Sulaimani, Iraq
| | - Aram Sha
- College of Dentistry, University of Sulaimani, Sulaimani, Iraq
| | - Iain L.C. Chapple
- Periodontal Research Group, Institute of Clinical Sciences, College of Medical & Dental Sciences, University of Birmingham, Birmingham, UK
| |
Collapse
|
43
|
Tagg JR, Harold LK, Jain R, Hale JDF. Beneficial modulation of human health in the oral cavity and beyond using bacteriocin-like inhibitory substance-producing streptococcal probiotics. Front Microbiol 2023; 14:1161155. [PMID: 37056747 PMCID: PMC10086258 DOI: 10.3389/fmicb.2023.1161155] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 03/03/2023] [Indexed: 03/30/2023] Open
Abstract
The human oral cavity contains a diversity of microbial habitats that have been adopted and adapted to as homeland by an amazingly heterogeneous population of microorganisms collectively referred to as the oral microbiota. These microbes generally co-habit in harmonious homeostasis. However, under conditions of imposed stress, as with changes to the host’s physiology or nutritional status, or as a response to foreign microbial or antimicrobial incursions, some components of the oral “microbiome” (viz. the in situ microbiota) may enter a dysbiotic state. This microbiome dysbiosis can manifest in a variety of guises including streptococcal sore throats, dental caries, oral thrush, halitosis and periodontal disease. Most of the strategies currently available for the management or treatment of microbial diseases of the oral cavity focus on the repetitive “broad sweep” and short-term culling of oral microbe populations, hopefully including the perceived principal pathogens. Both physical and chemical techniques are used. However, the application of more focused approaches to the harnessing or elimination of key oral cavity pathogens is now feasible through the use of probiotic strains that are naturally adapted for oral cavity colonization and also are equipped to produce anti-competitor molecules such as the bacteriocins and bacteriocin-like inhibitory substances (viz BLIS). Some of these probiotics are capable of suppressing the proliferation of a variety of recognized microbial pathogens of the human mouth, thereby assisting with the restoration of oral microbiome homeostasis. BLIS K12 and BLIS M18, the progenitors of the BLIS-producing oral probiotics, are members of the human oral cavity commensal species Streptococcus salivarius. More recently however, a number of other streptococcal and some non-streptococcal candidate oral probiotics have also been promoted. What is becoming increasingly apparent is that the future for oral probiotic applications will probably extend well beyond the attempted limitation of the direct pathological consequences of oral microbiome dysbiosis to also encompass a plethora of systemic diseases and disorders of the human host. The background to and the evolving prospects for the beneficial modulation of the oral microbiome via the application of BLIS-producing S. salivarius probiotics comprises the principal focus of the present review.
Collapse
|
44
|
Hu Z, Tang Y, Jiang B, Xu Y, Liu S, Huang C. Functional liposome loaded curcumin for the treatment of Streptococcus mutans biofilm. Front Chem 2023; 11:1160521. [PMID: 37007057 PMCID: PMC10065455 DOI: 10.3389/fchem.2023.1160521] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 03/06/2023] [Indexed: 03/19/2023] Open
Abstract
Introduction: Plaque biofilms, mainly formed by Streptococcus mutans (S. mutans), play an important role in the occurrence and development of dental caries. Antibiotic treatment is the traditional way to control plaque. However, problems such as poor drug penetration and antibiotic resistance have encouraged the search for alternative strategies. In this paper, we hope to avoid antibiotic resistance through the antibacterial effect of curcumin, a natural plant extract with photodynamic effects, on S. mutans. However, the clinical application of curcumin is limited due to its low water solubility, poor stability, high metabolic rate, fast clearance rate, and limited bioavailability. In recent years, liposomes have become a widely used drug carrier due to their numerous advantages, such as high drug loading efficiency, high stability in the biological environment, controlled release, biocompatibility, non-toxic, and biodegradability. So, we constructed a curcumin-loaded liposome (Cur@LP) to avoid the defect of curcumin.Methods: Cur@LP functioned with NHS can adhere to the surface of the S. mutans biofilm by condensation reaction. Liposome (LP) and Cur@LP was characterized by transmission electron microscopy (TEM) and dynamic light scattering (DLS). The cytotoxicity of Cur@LP was evaluated by CCK-8 assay and LDH assay. The adhesion of Cur@LP to S. mutans biofilm was observed by confocal laser scanning microscope (CLSM). The antibiofilm efficiency of Cur@LP were evaluated by crystal violet staining, CLSM, and scanning electron microscope (SEM).Results: The mean diameter of LP and Cur@LP were 206.67 ± 8.38 nm and 312 ± 18.78 nm respectively. The ζ-potential of LP and Cur@LP were ∼−19.3 mV and ∼−20.8 mV respectively. The encapsulation efficiency of Cur@LP was (42.61 ± 2.19) %, and curcumin was rapidly released up to ±21% at 2 h. Cur@LP has negligible cytotoxicity, and can effectively adhered to the S. mutans biofilm and inhibited its growth.Discussion: Curcumin has been widely studied in many fields such as cancer, which can be attributed to its antioxidant and anti-inflammatory effects. At present, there are few studies on the delivery of curcumin to S. mutans biofilm. In this study, we verified the adhesion and antibiofilm of Cur@LP to S. mutans biofilm. This biofilm removal strategy has the potential to be translated into the clinic.
Collapse
|
45
|
Butyrate as a Potential Driver of a Dysbiotic Shift of the Tongue Microbiota. mSphere 2023; 8:e0049022. [PMID: 36507724 PMCID: PMC9942584 DOI: 10.1128/msphere.00490-22] [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] [Indexed: 12/15/2022] Open
Abstract
The tongue dorsum is colonized by a stable microbiota, mostly comprising common commensal taxa. However, the predominance of each taxon varies among individuals. We hypothesized that equilibrium in the tongue microbiota is affected by exposure to butyrate in the oral fluid, which is reported to affect the growth of specific microorganisms. In this study, the bacterial composition of the tongue microbiotas of 69 male adults was determined via 16S rRNA gene sequencing to investigate its relationship to n-butyric acid concentration in oral rinse samples. The tongue microbiotas of individuals with a higher n-butyric acid level had higher relative abundances of Prevotella histicola, Veillonella atypica, and Streptococcus parasanguinis and lower relative abundances of Neisseria subflava and Porphyromonas pasteri. Subsequently, tongue microbiota samples collected from 12 adults were cultivated for 13 h in basal medium containing mucin and different concentrations of sodium butyrate (0, 0.8, 1.6, and 3.2 mM) to assess its effect on the growth of tongue microbiota organisms. The bacterial composition of the cultivated tongue microbiotas also demonstrated a significant gradual shift with an increase in sodium butyrate levels in beta-diversity analysis. N. subflava was significantly less predominant in the microbiota after cultivation with an increased addition of sodium butyrate, although no statistical difference was observed in the other aforementioned taxa. These results suggest that butyrate in the oral fluid is partially involved in the dysbiotic shift of the tongue microbiota. IMPORTANCE Oral microbial populations that are always ingested with saliva have attracted increasing attention because more oral microorganisms than previously known reach distal organs, such as the lungs and intestinal tract, thereby affecting our health. However, although such organisms are predominately derived from the tongue dorsum, the dynamics and determinants of the tongue microbiota composition remain unclear. This study demonstrated that exposure to butyrate could lead to a dysbiotic shift in the tongue microbiota using an observational epidemiological and microbiota cultivation approach. This result adds a new dimension to tongue microbiota ecology.
Collapse
|
46
|
Chen WA, Dou Y, Fletcher HM, Boskovic DS. Local and Systemic Effects of Porphyromonas gingivalis Infection. Microorganisms 2023; 11:470. [PMID: 36838435 PMCID: PMC9963840 DOI: 10.3390/microorganisms11020470] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/31/2023] [Accepted: 02/02/2023] [Indexed: 02/16/2023] Open
Abstract
Porphyromonas gingivalis, a gram-negative anaerobe, is a leading etiological agent in periodontitis. This infectious pathogen can induce a dysbiotic, proinflammatory state within the oral cavity by disrupting commensal interactions between the host and oral microbiota. It is advantageous for P. gingivalis to avoid complete host immunosuppression, as inflammation-induced tissue damage provides essential nutrients necessary for robust bacterial proliferation. In this context, P. gingivalis can gain access to the systemic circulation, where it can promote a prothrombotic state. P. gingivalis expresses a number of virulence factors, which aid this pathogen toward infection of a variety of host cells, evasion of detection by the host immune system, subversion of the host immune responses, and activation of several humoral and cellular hemostatic factors.
Collapse
Affiliation(s)
- William A. Chen
- Division of Biochemistry, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
| | - Yuetan Dou
- Division of Microbiology and Molecular Genetics, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
| | - Hansel M. Fletcher
- Division of Microbiology and Molecular Genetics, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
| | - Danilo S. Boskovic
- Division of Biochemistry, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
| |
Collapse
|
47
|
Kreth J, Merritt J. Illuminating the oral microbiome and its host interactions: tools and approaches for molecular ecological studies. FEMS Microbiol Rev 2023; 47:fuac052. [PMID: 36564013 PMCID: PMC9936263 DOI: 10.1093/femsre/fuac052] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 12/20/2022] [Accepted: 12/22/2022] [Indexed: 12/25/2022] Open
Abstract
A more comprehensive understanding of oral diseases like caries and periodontitis is dependent on an intimate understanding of the microbial ecological processes that are responsible for disease development. With this review, we provide a comprehensive overview of relevant molecular ecology techniques that have played critical roles in the current understanding of human oral biofilm development, interspecies interactions, and microbiome biogeography. The primary focus is on relevant technologies and examples available in the oral microbiology literature. However, most, if not all, of the described technologies should be readily adaptable for studies of microbiomes from other mucosal sites in the body. Therefore, this review is intended to serve as a reference guide used by microbiome researchers as they inevitably transition into molecular mechanistic studies of the many significant phenotypes observed clinically.
Collapse
Affiliation(s)
- Jens Kreth
- Department of Restorative Dentistry, School of Dentistry, Oregon Health and Science University, MRB433, 3181 SW Sam Jackson Park Rd., #L595, Portland, OR 97239, United States
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR, United States
| | - Justin Merritt
- Department of Restorative Dentistry, School of Dentistry, Oregon Health and Science University, MRB433, 3181 SW Sam Jackson Park Rd., #L595, Portland, OR 97239, United States
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR, United States
| |
Collapse
|
48
|
MATOS AO, RANGEL EC, BARÃO VAR, GREGORY RL. Antimicrobial behavior of titanium coating with chlorhexidine-doped thin film exposed to a biofilm supplemented with nicotine. Dent Mater J 2023; 42:228-235. [PMID: 36464292 DOI: 10.4012/dmj.2022-168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
Because nicotine upregulates the growth of most oral bacteria, this in vitro study investigated the antimicrobial effect of chlorhexidine-doped thin film on commercially pure titanium against Fusobacterium nucleatum (F. nucleatum) biofilm supplemented with different concentrations of nicotine (0, 1, and 2 mg/mL). Biofilms were formed on a chlorhexidine-doped thin film on commercially-pure-titanium discs and compared to the control groups. Biofilm viability, total biofilm growth using a spectrophotometer, extracellular polysaccharide content, and pH variations were assessed as dependent variables. Data were submitted to ANOVA and Tukey honest significant difference tests (α=0.05). F. nucleatum biofilm growth was inhibited when exposed to chlorhexidine-doped thin film (p<0.05). Biofilm supplemented with nicotine did not impact the synthesis of EPS on the same type of treatment (p>0.05). The pH values were significantly increased with the increase of nicotine concentration (p<0.05). Chlorhexidine-doped thin film was effective in reducing F. nucleatum biofilm supplemented with nicotine.
Collapse
Affiliation(s)
| | - Elidiane Cipriano RANGEL
- Laboratory of Technological Plasmas (LaPTec), Engineering College, Sao Paulo State University (UNESP)
| | | | - Richard Lee GREGORY
- Department of Biomedical Sciences and Comprehensive Care, Indiana University School of Dentistry
| |
Collapse
|
49
|
Murugkar P, Dimise E, Stewart E, Viala SN, Clardy J, Dewhirst FE, Lewis K. Identification of a growth factor required for culturing specific fastidious oral bacteria. J Oral Microbiol 2023; 15:2143651. [DOI: 10.1080/20002297.2022.2143651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Affiliation(s)
- Pallavi Murugkar
- Antimicrobial Discovery Center, Department of Biology, Northeastern University, 134 Mugar Hall, 360 Huntington Ave 02115, Boston, MA, USA
| | - Eric Dimise
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School 02115, Boston, MA, USA
| | - Eric Stewart
- Antimicrobial Discovery Center, Department of Biology, Northeastern University, 134 Mugar Hall, 360 Huntington Ave 02115, Boston, MA, USA
| | - Stéphane N. Viala
- Department of Microbiology, the Forsyth Institute, Cambridge, MA, USA
| | - Jon Clardy
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School 02115, Boston, MA, USA
| | - Floyd E. Dewhirst
- Department of Microbiology, the Forsyth Institute, Cambridge, MA, USA
- Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA, USA
| | - Kim Lewis
- Antimicrobial Discovery Center, Department of Biology, Northeastern University, 134 Mugar Hall, 360 Huntington Ave 02115, Boston, MA, USA
| |
Collapse
|
50
|
Neilands J, Svensäter G, Boisen G, Robertsson C, Wickström C, Davies JR. Formation and Analysis of Mono-species and Polymicrobial Oral Biofilms in Flow-Cell Models. Methods Mol Biol 2023; 2674:33-54. [PMID: 37258958 DOI: 10.1007/978-1-0716-3243-7_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
The oral microbiota, which is known to include at least 600 different bacterial species, is found on the teeth and mucosal surfaces as multi-species communities or biofilms. The oral surfaces are covered with a pellicle of proteins absorbed from saliva, and biofilm formation is initiated when primary colonizers, which express surface adhesins that bind to specific salivary components, attach to the oral tissues. Further development then proceeds through co-aggregation of additional species. Over time, the composition of oral biofilms, which varies between different sites throughout the oral cavity, is determined by a combination of environmental factors such as the properties of the underlying surface, nutrient availability and oxygen levels, and bacterial interactions within the community. A complex equilibrium between biofilm communities and the host is responsible for the maintenance of a healthy biofilm phenotype (eubiosis). In the face of sustained environmental perturbation, however, biofilm homeostasis can break down giving rise to dysbiosis, which is associated with the development of oral diseases such as caries and periodontitis.In vitro models have an important part to play in increasing our understanding of the complex processes involved in biofilm development in oral health and disease, and the requirements for experimental system, microbial complexity, and analysis techniques will necessarily vary depending on the question posed. In this chapter we describe some current and well-established methods used in our laboratory for studying oral bacteria in biofilm models which can be adapted to suit the needs of individual users.
Collapse
Affiliation(s)
- Jessica Neilands
- Section for Oral Biology and Pathology, Faculty of Odontology, Malmö University, Malmö, Sweden
| | - Gunnel Svensäter
- Section for Oral Biology and Pathology, Faculty of Odontology, Malmö University, Malmö, Sweden
| | - Gabriella Boisen
- Section for Oral Biology and Pathology, Faculty of Odontology, Malmö University, Malmö, Sweden
| | - Carolina Robertsson
- Section for Oral Biology and Pathology, Faculty of Odontology, Malmö University, Malmö, Sweden
| | - Claes Wickström
- Section for Oral Biology and Pathology, Faculty of Odontology, Malmö University, Malmö, Sweden
| | - Julia R Davies
- Section for Oral Biology and Pathology, Faculty of Odontology, Malmö University, Malmö, Sweden.
| |
Collapse
|