Insights in Oral Microbiota

A special issue of Pathogens (ISSN 2076-0817). This special issue belongs to the section "Bacterial Pathogens".

Deadline for manuscript submissions: closed (31 December 2023) | Viewed by 22914

Special Issue Editor


E-Mail Website
Guest Editor
State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China School of Stomatology, Sichuan University, Chengdu 610064, China
Interests: oral microbiome; oral diseases; drug development; drug resistance

Special Issue Information

Dear Colleagues,

Oral microbiota is the most diverse microbiota after gut microbiota in humans, harboring bacteria, viruses, fungi, and protozoa. Oral microbiota is essential for the maintenance of oral and systemic health. The shift of oral microbiota contributes greatly to the development of oral and systemic diseases. Understanding the functional mechanisms of oral microbiota and identifying the key microbes will shed light on new concepts and directions for the prevention, diagnosis, and treatment of certain oral and systemic diseases. In this Special Issue, we will unveil the diversity and structure of oral microbiota in health and disease, decipher the functions of oral microbiota/microbe in the development of oral and systemic diseases, and explore the new strategies to regulate/treat the oral microbiota or the key microbes. The main goal of this Special Issue is to provide the clinical relevance of oral microbiota in health and diseases and the new therapeutic strategies or technologies, such as new antimicrobial agents, probiotics, etc.

Prof. Dr. Biao Ren
Guest Editor

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Pathogens is an international peer-reviewed open access monthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2700 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • Oral microbiota
  • Key microbes
  • Oral and systemic diseases
  • Antimicrobial agents
  • Probiotics

Published Papers (9 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

Jump to: Review

12 pages, 2864 KiB  
Article
Oral Microbial Signatures of Tobacco Chewers and Oral Cancer Patients in India
by Shriya Sawant, Jinesh Dugad, Deepak Parikh, Sathiyaraj Srinivasan and Harinder Singh
Pathogens 2023, 12(1), 78; https://doi.org/10.3390/pathogens12010078 - 03 Jan 2023
Cited by 7 | Viewed by 2232
Abstract
Dysbiosis of the oral microbiome has been found to play a key role in the genesis and progression of oral cancer (OC). Tobacco chewing, a risk factor for oral cancer, is also associated with oral dysbiosis. Since tobacco chewing is a lifestyle habit [...] Read more.
Dysbiosis of the oral microbiome has been found to play a key role in the genesis and progression of oral cancer (OC). Tobacco chewing, a risk factor for oral cancer, is also associated with oral dysbiosis. Since tobacco chewing is a lifestyle habit in the South Asian subcontinent, including India, and contributes to one-third of the global oral cancer burden; we aimed to identify the oral bacterial diversity of Indian oral cancer patients and tobacco chewers. We used 16S rRNA amplicon sequencing to study the composition of oral microbiota in OC patients and tobacco chewers in India and compared it with healthy controls. The abundance of predominant phyla, Firmicutes, and Bacteroidetes varied between the study groups. Our study identified Leptotrichia, Treponema, Lautropia, and Cardiobacterium as significantly enriched in tobacco chewers, whereas genera Pseudomonas, Capnocytophaga, and Mycoplasma were enriched in oral cancer, which could be potential biomarkers for the Indian population. Furthermore, the functional prediction revealed that genes involved in lipid biosynthesis and fatty acid elongation were upregulated in the oral cancer group, whereas those for the reductive TCA cycle were upregulated in the tobacco group. As the role of bacteria in oral cancer is becoming more evident, identification of bacterial diversity and biomarkers for tobacco chewers and OC patients can aid in the early diagnosis of OC in high-risk individuals. Full article
(This article belongs to the Special Issue Insights in Oral Microbiota)
Show Figures

Figure 1

12 pages, 3408 KiB  
Article
Culture Supernatant of Enterococcus faecalis Promotes the Hyphal Morphogenesis and Biofilm Formation of Candida albicans
by Qingsong Jiang, Qi Jing, Biao Ren, Lei Cheng, Xuedong Zhou, Wenli Lai, Jinzhi He and Mingyun Li
Pathogens 2022, 11(10), 1177; https://doi.org/10.3390/pathogens11101177 - 12 Oct 2022
Cited by 5 | Viewed by 1626
Abstract
(1) Background: Enterococcus faecalis and Candida albicans are often isolated from infected root canals. The interaction between these two species is not clear enough. Therefore, the objective of this study was to investigate the effect of E. faecalis on the biofilm formation, hyphal [...] Read more.
(1) Background: Enterococcus faecalis and Candida albicans are often isolated from infected root canals. The interaction between these two species is not clear enough. Therefore, the objective of this study was to investigate the effect of E. faecalis on the biofilm formation, hyphal morphogenesis and virulence gene expression of C. albicans. (2) Methods: We used the culture supernatant of E. faecalis (CSE) to treat the biofilms of C. albicans. Then, crystal violet staining and colony-forming unit (CFU) counting were performed to evaluate biofilm biomass. Scanning electron microscopy (SEM) and confocal laser scanning microscope (CLSM) were applied to observe fungal morphology. Subsequently, exopolymeric substances (EPS) production, cellular surface hydrophobicity (CSH) and adhesion force of biofilms were investigated by CLSM, water–hydrocarbon two-phase assay and atomic force microscopy (AFM), respectively. Finally, the expression of C. albicans virulence genes (ALS1, ALS3, HWP1 and EFG1) were measured by RT-qPCR assay. (3) Results: The exposure of CSE promoted the biofilm formation and hyphal morphogenesis of C. albicans, increased the EPS production, CSH and adhesion force of C. albicans biofilms, and increased the expression level of EFG1. (4) Conclusions: Our data indicated that CSE promoted the hyphal morphogenesis and biofilm formation of C. albicans. Full article
(This article belongs to the Special Issue Insights in Oral Microbiota)
Show Figures

Figure 1

14 pages, 2317 KiB  
Article
Treponema denticola Induces Neuronal Apoptosis by Promoting Amyloid-β Accumulation in Mice
by Linrui Wu, Xinyi Su, Zhiqun Tang, Lixiang Jian, He Zhu, Xingqun Cheng and Hongkun Wu
Pathogens 2022, 11(10), 1150; https://doi.org/10.3390/pathogens11101150 - 05 Oct 2022
Cited by 6 | Viewed by 2245
Abstract
Background: Neuronal apoptosis is a major contributor to Alzheimer’s disease (AD). Periodontitis is a significant risk factor for AD. The periodontal pathogens Porphyromonas gingivalis and Treponema denticola have been shown to initiate the hallmark pathologies and behavioral symptoms of AD. Studies have found [...] Read more.
Background: Neuronal apoptosis is a major contributor to Alzheimer’s disease (AD). Periodontitis is a significant risk factor for AD. The periodontal pathogens Porphyromonas gingivalis and Treponema denticola have been shown to initiate the hallmark pathologies and behavioral symptoms of AD. Studies have found that T. denticola infection induced Tau hyperphosphorylation and amyloid β accumulation in the hippocampi of mice. Aβ accumulation is closely associated with neuronal apoptosis. However, the roles of T. denticola in neuronal apoptosis remain unclear and its roles in AD pathology need further study. Objective: This study aimed to investigate whether oral infection with T. denticola induced alveolar bone loss and neuronal apoptosis in mice. Methods: C57BL/6 mice were orally administered with T. denticola, Micro-CT was employed to assess the alveolar bone resorption. Western blotting, quantitative PCR, and TUNEL staining were utilized to detect the apoptosis-associated changes in mouse hippocampi. N2a were co-cultured with T. denticola to verify in vivo results. Results: Mice infected with T. denticola exhibited more alveolar bone loss compared with the control mice. T. denticola oral infection induced neuronal apoptosis in hippocampi of mice. Consistent results of the apoptosis-associated protein expression were observed in N2a cells treated with T. denticola and Aβ1–42 in vitro. However, the Aβ inhibitor reversed these results, suggesting that Aβ1–42 mediates T. denticola infection-induced neuronal apoptosis. Conclusions: This study found that oral infected T. denticola caused alveolar bone loss, and induced neuronal apoptosis by promoting Aβ accumulation in mice, providing evidence for the link between periodontitis and AD. Full article
(This article belongs to the Special Issue Insights in Oral Microbiota)
Show Figures

Figure 1

14 pages, 10479 KiB  
Article
Characterization of Bacterial Differences Induced by Cleft-Palate-Related Spatial Heterogeneity
by Fangjie Zhou, Zhifei Su, Qinyang Li, Renke Wang, Ying Liao, Min Zhang and Jiyao Li
Pathogens 2022, 11(7), 771; https://doi.org/10.3390/pathogens11070771 - 05 Jul 2022
Cited by 3 | Viewed by 2034
Abstract
Background: Cleft palate (CP) patients have a higher prevalence of oral and respiratory tract bacterial infections than the general population. Nevertheless, characteristics of bacterial differences induced by CP-related anatomical heterogeneity are unknown. Methods: In this study, we systematically described the characteristics of [...] Read more.
Background: Cleft palate (CP) patients have a higher prevalence of oral and respiratory tract bacterial infections than the general population. Nevertheless, characteristics of bacterial differences induced by CP-related anatomical heterogeneity are unknown. Methods: In this study, we systematically described the characteristics of bacteria in the oral and nasal niches in healthy children, CP children, healthy adolescents, CP adolescents, and postoperative adolescents by 454-pyrosequencing technology (V3–V6) to determine bacterial differences induced by CP. Results: Due to the CP-induced variations in spatial structure, the early establishment of microecology in CP children was different from that in healthy children. Nasal bacterial composition showed greater changes than in the saliva. Moreover, such discrepancy also appeared in CP and postoperative adolescents who had even undergone surgery > 10 years previously. Interestingly, we found by Lefse analysis that part of bacterial biomarkers in the nasal cavity of CP subjects was common oral flora, suggesting bacterial translocation between the oral and nasal niches. Therefore, we defined the oral–nasal translocation bacteria as O-N bac. By comparing multiple groups, we took the intersection sets of O-N bacs selected from CP children, CP adolescents, and postoperative adolescents as TS O-N bacs with time–character, including Streptococcus, Gemella, Alloprevotella, Neisseria, Rothia, Actinomyces, and Veillonella. These bacteria were at the core of the nasal bacterial network in CP subjects, and some were related to infectious diseases. Conclusions: CP would lead to significant and long-term differences in oral and nasal flora. TS O-N bacs migrating from the oral to the nasal might be the key stone causing nasal flora dysbiosis in the CP patients. Full article
(This article belongs to the Special Issue Insights in Oral Microbiota)
Show Figures

Figure 1

18 pages, 4854 KiB  
Article
Novel Giomers Incorporated with Antibacterial Quaternary Ammonium Monomers to Inhibit Secondary Caries
by Yandi Chen, Bina Yang, Lei Cheng, Hockin H. K. Xu, Hao Li, Yuyao Huang, Qiong Zhang, Xuedong Zhou, Jingou Liang and Jing Zou
Pathogens 2022, 11(5), 578; https://doi.org/10.3390/pathogens11050578 - 14 May 2022
Cited by 4 | Viewed by 2290
Abstract
The objective of this study was to develop novel modified giomers by incorporating the antibacterial quaternary ammonium monomers (QAMs), dimethylaminododecyl methacrylate (DMADDM) or dimethylaminohexadecyl methacrylate (DMAHDM) into a commercial giomer. The material performances including mechanical properties, surface characteristics, color data, cytotoxicity and fluoride [...] Read more.
The objective of this study was to develop novel modified giomers by incorporating the antibacterial quaternary ammonium monomers (QAMs), dimethylaminododecyl methacrylate (DMADDM) or dimethylaminohexadecyl methacrylate (DMAHDM) into a commercial giomer. The material performances including mechanical properties, surface characteristics, color data, cytotoxicity and fluoride release of the novel giomers were evaluated. Antibacterial activity against severe early childhood caries (S-ECC) saliva-derived biofilms was assessed by lactic acid production measurement, MTT assay, biofilm staining and 16S rRNA sequencing. A rat model was developed and the anti-caries effect was investigated by micro-CT scanning and modified Keyes’ scoring. The results showed that the material properties of the QAMs groups were comparable to those of the control group. The novel giomers significantly inhibited lactic acid production and biofilm viability of S-ECC saliva-derived biofilms. Furthermore, caries-related genera such as Streptococcus and Lactobacillus reduced in QAMs groups, which showed their potential to change the microbial compositions. In the rat model, lesion depth, mineral loss and scoring of the QAMs groups were significantly reduced, without side effects on oral tissues. In conclusion, the novel giomers incorporated with antibacterial QAMs could inhibit the cariogenic biofilms and help prevent secondary caries, with great potential for future application in restorative treatment. Full article
(This article belongs to the Special Issue Insights in Oral Microbiota)
Show Figures

Figure 1

13 pages, 4101 KiB  
Article
Regulatory Effect of Irresistin-16 on Competitive Dual-Species Biofilms Composed of Streptococcus mutans and Streptococcus sanguinis
by Xiangyu Hu, Min Wang, Yan Shen, Lingjun Zhang, Yihuai Pan, Yan Sun and Keke Zhang
Pathogens 2022, 11(1), 70; https://doi.org/10.3390/pathogens11010070 - 06 Jan 2022
Cited by 4 | Viewed by 2144
Abstract
Based on the ecological plaque hypothesis, suppressing opportunistic pathogens within biofilms, rather than killing microbes indiscriminately, could be a biofilm control strategy for managing dental caries. The present study aimed to evaluate the effects of irresistin-16 (IRS-16) on competitive dual-species biofilms, which consisted [...] Read more.
Based on the ecological plaque hypothesis, suppressing opportunistic pathogens within biofilms, rather than killing microbes indiscriminately, could be a biofilm control strategy for managing dental caries. The present study aimed to evaluate the effects of irresistin-16 (IRS-16) on competitive dual-species biofilms, which consisted of the conditional cariogenic agent Streptococcus mutans (S. mutans) and oral commensal bacteria Streptococcus sanguinis (S. sanguinis). Bacterial growth and biofilm formation were monitored using growth curve and crystal violet staining, respectively. The microbial proportion was determined using fluorescence in situ hybridization. A 2, 5-diphenyltetrazolium bromide assay was used to measure the metabolic activity of biofilms. Bacterial/extracellular polysaccharide (EPS) dyeing, together with water-insoluble EPS measurements, were used to estimate EPS synthesis. A lactic acid assay was performed to detect lactic acid generation in biofilms. The cytotoxicity of IRS-16 was evaluated in mouse fibroblast L929 cells using a live/dead cell viability assay and cell counting kit-8 assay. Our results showed that IRS-16 exhibited selective anti-biofilm activity, leading to a remarkable survival disadvantage of S. mutans within competitive dual-species biofilms. In addition, the metabolic activity, EPS synthesis, and acid generation of dual-species biofilms were significantly reduced by IRS-16. Moreover, IRS-16 showed minimal cytotoxicity against mouse fibroblast L929 cells. In conclusion, IRS-16 exhibited remarkable regulatory effects on dual-species biofilms composed of S. mutans and S. sanguinis with low cytotoxicity, suggesting that it may have potential for use in caries management through ecological biofilm control. Full article
(This article belongs to the Special Issue Insights in Oral Microbiota)
Show Figures

Figure 1

Review

Jump to: Research

19 pages, 1219 KiB  
Review
Periodontopathogens Porphyromonas gingivalis and Fusobacterium nucleatum and Their Roles in the Progression of Respiratory Diseases
by Tao Shi, Jiale Wang, Jiajia Dong, Pingyue Hu and Qiang Guo
Pathogens 2023, 12(9), 1110; https://doi.org/10.3390/pathogens12091110 - 30 Aug 2023
Cited by 4 | Viewed by 2499
Abstract
The intricate interplay between oral microbiota and the human host extends beyond the confines of the oral cavity, profoundly impacting the general health status. Both periodontal diseases and respiratory diseases show high prevalence worldwide and have a marked influence on the quality of [...] Read more.
The intricate interplay between oral microbiota and the human host extends beyond the confines of the oral cavity, profoundly impacting the general health status. Both periodontal diseases and respiratory diseases show high prevalence worldwide and have a marked influence on the quality of life for the patients. Accumulating studies are establishing a compelling association between periodontal diseases and respiratory diseases. Here, in this review, we specifically focus on the key periodontal pathogenic bacteria Porphyromonas gingivalis and Fusobacterium nucleatum and dissect their roles in the onset and course of respiratory diseases, mainly pneumonia, chronic obstructive pulmonary disease, lung cancer, and asthma. The mechanistic underpinnings and molecular processes on how P. gingivalis and F. nucleatum contribute to the progression of related respiratory diseases are further summarized and analyzed, including: induction of mucus hypersecretion and chronic airway inflammation; cytotoxic effects to disrupt the morphology and function of respiratory epithelial cells; synergistic pathogenic effects with respiratory pathogens like Streptococcus pneumoniae and Pseudomonas aeruginosa. By delving into the complex relationship to periodontal diseases and periodontopathogens, this review helps unearth novel insights into the etiopathogenesis of respiratory diseases and inspires the development of potential therapeutic avenues and preventive strategies. Full article
(This article belongs to the Special Issue Insights in Oral Microbiota)
Show Figures

Figure 1

21 pages, 744 KiB  
Review
Application of Fluorescence In Situ Hybridization (FISH) in Oral Microbial Detection
by Junjie Gu, Huayu Wang, Mengye Zhang, Yichen Xiong, Lei Yang, Biao Ren and Ruijie Huang
Pathogens 2022, 11(12), 1450; https://doi.org/10.3390/pathogens11121450 - 01 Dec 2022
Cited by 2 | Viewed by 3545
Abstract
Varieties of microorganisms reside in the oral cavity contributing to the occurrence and development of microbes associated with oral diseases; however, the distribution and in situ abundance in the biofilm are still unclear. In order to promote the understanding of the ecosystem of [...] Read more.
Varieties of microorganisms reside in the oral cavity contributing to the occurrence and development of microbes associated with oral diseases; however, the distribution and in situ abundance in the biofilm are still unclear. In order to promote the understanding of the ecosystem of oral microbiota and the diagnosis of oral diseases, it is necessary to monitor and compare the oral microorganisms from different niches of the oral cavity in situ. The fluorescence in situ hybridization (FISH) has proven to be a powerful tool for representing the status of oral microorganisms in the oral cavity. FISH is one of the most routinely used cytochemical techniques for genetic detection, identification, and localization by a fluorescently labeled nucleic acid probe, which can hybridize with targeted nucleic acid sequences. It has the advantages of rapidity, safety, high sensitivity, and specificity. FISH allows the identification and quantification of different oral microorganisms simultaneously. It can also visualize microorganisms by combining with other molecular biology technologies to represent the distribution of each microbial community in the oral biofilm. In this review, we summarized and discussed the development of FISH technology and the application of FISH in oral disease diagnosis and oral ecosystem research, highlighted its advantages in oral microbiology, listed the existing problems, and provided suggestions for future development.. Full article
(This article belongs to the Special Issue Insights in Oral Microbiota)
Show Figures

Graphical abstract

16 pages, 11423 KiB  
Review
Small Molecule Compounds, A Novel Strategy against Streptococcus mutans
by Sirui Yang, Jin Zhang, Ran Yang and Xin Xu
Pathogens 2021, 10(12), 1540; https://doi.org/10.3390/pathogens10121540 - 25 Nov 2021
Cited by 8 | Viewed by 2684
Abstract
Dental caries, as a common oral infectious disease, is a worldwide public health issue. Oral biofilms are the main cause of dental caries. Streptococcus mutans (S. mutans) is well recognized as the major causative factor of dental caries within oral biofilms. [...] Read more.
Dental caries, as a common oral infectious disease, is a worldwide public health issue. Oral biofilms are the main cause of dental caries. Streptococcus mutans (S. mutans) is well recognized as the major causative factor of dental caries within oral biofilms. In addition to mechanical removal such as tooth brushing and flossing, the topical application of antimicrobial agents is necessarily adjuvant to the control of caries particularly for high-risk populations. The mainstay antimicrobial agents for caries such as chlorhexidine have limitations including taste confusions, mucosal soreness, tooth discoloration, and disruption of an oral microbial equilibrium. Antimicrobial small molecules are promising in the control of S. mutans due to good antimicrobial activity, good selectivity, and low toxicity. In this paper, we discussed the application of antimicrobial small molecules to the control of S. mutans, with a particular focus on the identification and development of active compounds and their modes of action against the growth and virulence of S. mutans. Full article
(This article belongs to the Special Issue Insights in Oral Microbiota)
Back to TopTop