Oral Biofilms

A special issue of Microorganisms (ISSN 2076-2607). This special issue belongs to the section "Biofilm".

Deadline for manuscript submissions: closed (30 November 2020) | Viewed by 67925

Special Issue Editor


E-Mail Website
Guest Editor
School of Medicine, New York Medical College, Valhalla, NY 10595, USA
Interests: biofilm development and cell dispersion; COPD exacerbation; anti-microbial agent discovery; oral and systemic health linkage
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear Colleagues,

Microbial species are present in nature either as planktonic cells or incorporated into biofilms. Biofilms evolve from the planktonic state and are characterized as dense micro-communities that grow on inert surfaces and encapsulate themselves with secreted polymers. When organisms form a biofilm, they are able to adapt to environmental change by altering their gene expression patterns. The biofilm structure and corresponding change in gene expression can protect the microbes from disinfectant agents or antibiotics. The resultant biofilm can pose a serious public health issue. The physical nature of biofilms and the survival mechanisms they possess, whether phenotypic adaptability or genetic resistance, leave them impervious to antibiotic treatment. Given the lack of response to traditional antimicrobial therapy, biofilm infections currently pose a great challenge to the world of medicine.

The majority of the oral microbes are commensal organisms. Those that are pathogenic microbes can result in oral infections and at times initiate systemic diseases. This Special Issue focused on oral biofilms will address: (1). The development and life cycle of oral biofilms; (2). innovative approaches to study oral biofilms; and (3). methods to control the pathogenesis associated with biofilms.

Prof. Dr. Zvi Loewy
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. Microorganisms 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

Published Papers (15 papers)

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

Research

Jump to: Review, Other

19 pages, 2525 KiB  
Article
Probiotic Alternative to Chlorhexidine in Periodontal Therapy: Evaluation of Clinical and Microbiological Parameters
by Andrea Butera, Simone Gallo, Carolina Maiorani, Domenico Molino, Alessandro Chiesa, Camilla Preda, Francesca Esposito and Andrea Scribante
Microorganisms 2021, 9(1), 69; https://doi.org/10.3390/microorganisms9010069 - 29 Dec 2020
Cited by 91 | Viewed by 5631
Abstract
Periodontitis consists of a progressive destruction of tooth-supporting tissues. Considering that probiotics are being proposed as a support to the gold standard treatment Scaling-and-Root-Planing (SRP), this study aims to assess two new formulations (toothpaste and chewing-gum). 60 patients were randomly assigned to three [...] Read more.
Periodontitis consists of a progressive destruction of tooth-supporting tissues. Considering that probiotics are being proposed as a support to the gold standard treatment Scaling-and-Root-Planing (SRP), this study aims to assess two new formulations (toothpaste and chewing-gum). 60 patients were randomly assigned to three domiciliary hygiene treatments: Group 1 (SRP + chlorhexidine-based toothpaste) (control), Group 2 (SRP + probiotics-based toothpaste) and Group 3 (SRP + probiotics-based toothpaste + probiotics-based chewing-gum). At baseline (T0) and after 3 and 6 months (T1–T2), periodontal clinical parameters were recorded, along with microbiological ones by means of a commercial kit. As to the former, no significant differences were shown at T1 or T2, neither in controls for any index, nor in the experimental groups for adherent gingiva and gingival recession. Conversely, some significant differences were found in Group 2 and 3 for the other clinical indexes tested. Considering microbiological parameters, no significant differences were detected compared to baseline values for any group, except in Group 2 and 3 at T2 only for the percentage of the orange complex pathogens and for the copies/microliter of Prevotella intermedia and Fusobacterium nucleatum. Accordingly, although colonization of probiotic bacteria has not been assessed in this study, the probiotics tested represent a valid support to SRP with a benefit on several clinical indexes and on specific periodontopathogens. Despite this promising action, the relationship between the use of probiotics and improvement in clinical parameters is still unclear and deserves to be further explored. Full article
(This article belongs to the Special Issue Oral Biofilms)
Show Figures

Figure 1

13 pages, 2874 KiB  
Article
Interactions between Candida albicans and Enterococcus faecalis in an Organotypic Oral Epithelial Model
by Akshaya Lakshmi Krishnamoorthy, Alex A. Lemus, Adline Princy Solomon, Alex M. Valm and Prasanna Neelakantan
Microorganisms 2020, 8(11), 1771; https://doi.org/10.3390/microorganisms8111771 - 11 Nov 2020
Cited by 15 | Viewed by 3617
Abstract
Candida albicans as an opportunistic pathogen exploits the host immune system and causes a variety of life-threatening infections. The polymorphic nature of this fungus gives it tremendous advantage to breach mucosal barriers and cause oral and disseminated infections. Similar to C. albicans, [...] Read more.
Candida albicans as an opportunistic pathogen exploits the host immune system and causes a variety of life-threatening infections. The polymorphic nature of this fungus gives it tremendous advantage to breach mucosal barriers and cause oral and disseminated infections. Similar to C. albicans, Enterococcus faecalis is a major opportunistic pathogen, which is of critical concern in immunocompromised patients. There is increasing evidence that E. faecalis co-exists with C. albicans in the human body in disease samples. While the interactive profiles between these two organisms have been studied on abiotic substrates and mouse models, studies on their interactions on human oral mucosal surfaces are non-existent. Here, for the first time, we comprehensively characterized the interactive profiles between laboratory and clinical isolates of C. albicans (SC5314 and BF1) and E. faecalis (OG1RF and P52S) on an organotypic oral mucosal model. Our results demonstrated that the dual species biofilms resulted in profound surface erosion and significantly increased microbial invasion into mucosal compartments, compared to either species alone. Notably, several genes of C. albicans involved in tissue adhesion, hyphal formation, fungal invasion, and biofilm formation were significantly upregulated in the presence of E. faecalis. By contrast, E. faecalis genes involved in quorum sensing, biofilm formation, virulence, and mammalian cell invasion were downregulated. This study highlights the synergistic cross-kingdom interactions between E. faecalis and C. albicans in mucosal tissue invasion. Full article
(This article belongs to the Special Issue Oral Biofilms)
Show Figures

Graphical abstract

13 pages, 2977 KiB  
Article
Probiotic Effects on Multispecies Biofilm Composition, Architecture, and Caries Activity In Vitro
by Zhihui Chen, Sebastian Schlafer, Gerd Göstemeyer and Falk Schwendicke
Microorganisms 2020, 8(9), 1272; https://doi.org/10.3390/microorganisms8091272 - 21 Aug 2020
Cited by 8 | Viewed by 2724
Abstract
While probiotics have been tested for their anti-caries effect in vitro and also clinically, there is a lack of understanding of their effects on complex dental biofilms. We assessed two probiotics, Lactobacillus reuteri and Streptococcus oligofermentans, on a continuous-cultured model containing Streptococcus [...] Read more.
While probiotics have been tested for their anti-caries effect in vitro and also clinically, there is a lack of understanding of their effects on complex dental biofilms. We assessed two probiotics, Lactobacillus reuteri and Streptococcus oligofermentans, on a continuous-cultured model containing Streptococcus mutans, Lactobacillus rhamnosus and Actinomyces naeslundii. Cariogenic biofilms were grown on bovine enamel specimens and daily challenged with L. reuteri or S. oligofermentans whole culture (LC/SC) or cell-free supernatant (LS/SS) or medium only (negative control, NC) (n = 21/group) for 10 days. Biofilm was assessed via counting colony-forming units, quantitative polymerase chain reaction, and fluorescence in situ hybridization. Caries activity was determined by pH measurements and by assessing mineral loss (ΔZ) using transverse microradiography. Both LC and SC significantly reduced total and strain-specific cariogenic bacterial numbers (p < 0.05). ΔZ was reduced in LC (mean ± SD: 1846.67 ± 317.89) and SC (3315.87 ± 617.30) compared to NC (4681.48 ± 495.18, p < 0.05). No significant reductions in bacterial numbers and ΔZ was induced by supernatants. Biofilm architecture was not considerably affected by probiotic applications. Viable probiotics L. reuteri and S. oligofermentans, but not their culture supernatants, could reduce the caries activity of multi-species biofilms in vitro. Full article
(This article belongs to the Special Issue Oral Biofilms)
Show Figures

Figure 1

12 pages, 2902 KiB  
Article
A Novel Small Molecule, 1,3-di-m-tolyl-urea, Inhibits and Disrupts Multispecies Oral Biofilms
by Shanthini Kalimuthu, Becky P.K. Cheung, Joyce Y.Y. Yau, Karthi Shanmugam, Adline Princy Solomon and Prasanna Neelakantan
Microorganisms 2020, 8(9), 1261; https://doi.org/10.3390/microorganisms8091261 - 20 Aug 2020
Cited by 7 | Viewed by 2947
Abstract
An imbalance of homeostasis between the microbial communities and the host system leads to dysbiosis in oral micro flora. DMTU (1,3-di-m-tolyl-urea) is a biocompatible compound that was shown to inhibit Streptococcus mutans biofilm by inhibiting its communication system (quorum sensing). Here, we hypothesized [...] Read more.
An imbalance of homeostasis between the microbial communities and the host system leads to dysbiosis in oral micro flora. DMTU (1,3-di-m-tolyl-urea) is a biocompatible compound that was shown to inhibit Streptococcus mutans biofilm by inhibiting its communication system (quorum sensing). Here, we hypothesized that DMTU is able to inhibit multispecies biofilms. We developed a multispecies oral biofilm model, comprising an early colonizer Streptococcus gordonii, a bridge colonizer Fusobacterium nucleatum, and late colonizers Porphyromonas gingivalis and Aggregatibacter actinomycetemcomitans. We performed comprehensive investigations to demonstrate the effect of DMTU on planktonic cells and biofilms. Our findings showed that DMTU inhibits and disrupts multispecies biofilms without bactericidal effects. Mechanistic studies revealed a significant down regulation of biofilm and virulence-related genes in P. gingivalis. Taken together, our study highlights the potential of DMTU to inhibit polymicrobial biofilm communities and their virulence. Full article
(This article belongs to the Special Issue Oral Biofilms)
Show Figures

Figure 1

16 pages, 2638 KiB  
Article
Characterization of Scardovia wiggsiae Biofilm by Original Scanning Electron Microscopy Protocol
by Maurizio Bossù, Laura Selan, Marco Artini, Michela Relucenti, Giuseppe Familiari, Rosanna Papa, Gianluca Vrenna, Patrizia Spigaglia, Fabrizio Barbanti, Alessandro Salucci, Gianni Di Giorgio, Julietta V. Rau and Antonella Polimeni
Microorganisms 2020, 8(6), 807; https://doi.org/10.3390/microorganisms8060807 - 27 May 2020
Cited by 30 | Viewed by 4724
Abstract
Early childhood caries (ECC) is a severe manifestation of carious pathology with rapid and disruptive progression. The ECC microbiota includes a wide variety of bacterial species, among which is an anaerobic newly named species, Scardovia wiggsiae, a previously unidentified Bifidobacterium. Our aim [...] Read more.
Early childhood caries (ECC) is a severe manifestation of carious pathology with rapid and disruptive progression. The ECC microbiota includes a wide variety of bacterial species, among which is an anaerobic newly named species, Scardovia wiggsiae, a previously unidentified Bifidobacterium. Our aim was to provide the first ultrastructural characterization of S. wiggsiae and its biofilm by scanning electron microscopy (SEM) using a protocol that faithfully preserved the biofilm architecture and allowed an investigation at very high magnifications (order of nanometers) and with the appropriate resolution. To accomplish this task, we analyzed Streptococcus mutans’ biofilm by conventional SEM and VP-SEM protocols, in addition, we developed an original procedure, named OsO4-RR-TA-IL, which avoids dehydration, drying and sputter coating. This innovative protocol allowed high-resolution and high-magnification imaging (from 10000× to 35000×) in high-vacuum and high-voltage conditions. After comparing three methods, we chose OsO4-RR-TA-IL to investigate S. wiggsiae. It appeared as a fusiform elongated bacterium, without surface specialization, arranged in clusters and submerged in a rich biofilm matrix, which showed a well-developed micro-canalicular system. Our results provide the basis for the development of innovative strategies to quantify the effects of different treatments, in order to establish the best option to counteract ECC in pediatric patients. Full article
(This article belongs to the Special Issue Oral Biofilms)
Show Figures

Figure 1

13 pages, 2556 KiB  
Article
Anti-Inflammatory Effects of Vitamin E in Response to Candida albicans
by Silvana Barros, Ana Paula D. Ribeiro, Steven Offenbacher and Zvi G. Loewy
Microorganisms 2020, 8(6), 804; https://doi.org/10.3390/microorganisms8060804 - 26 May 2020
Cited by 5 | Viewed by 2985
Abstract
Oral mucositis, inflammation, and ulceration that occur in the oral cavity can manifest in significant pain. A formulation was designed to investigate the potential of vitamin E to ameliorate inflammation resulting from Candida albicans in cell-based systems. Human gingival fibroblasts and THP1 cells [...] Read more.
Oral mucositis, inflammation, and ulceration that occur in the oral cavity can manifest in significant pain. A formulation was designed to investigate the potential of vitamin E to ameliorate inflammation resulting from Candida albicans in cell-based systems. Human gingival fibroblasts and THP1 cells were stimulated with heat killed C. albicans and Porphyromonas gingivalis LPS (agonists). Unstimulated cells were included as controls. Cells were also simultaneously treated with a novel denture adhesive formulation that contains vitamin E (antagonist). The experimental conditions included cells exposed to the experimental formulation or the vehicle for 2 h for mRNA extraction and analysis, and cells left for 24 h under those experimental conditions for analysis of protein expression by ELISA. ssAffymetrix expression microarray pathway analyses demonstrated that the tested formulation exhibited a statistically significant (p < 0.05) inhibition of the following key inflammatory pathways: TLR 6, IL-1 signaling (IRAK, A20), NF-kappaB, IL-6 signaling (gp130, JK2 and GRB2), TNF signaling (TNF receptor) and Arachidonic acid metabolism (PLA2). Quantitative PCR array analysis confirmed the downregulation of key inflammatory genes when cells under adhesive treatment were challenged with heat killed C. albicans. PGE2 secretion was inhibited by the tested formulation only on THP1 cells after 24 h stimulation with C. albicans. These results suggest that the active formulation containing vitamin E acetate can modulate inflammatory responses, through anti-inflammatory actions as indicated by in vitro experimental conditions. Full article
(This article belongs to the Special Issue Oral Biofilms)
Show Figures

Figure 1

23 pages, 6067 KiB  
Article
Endodontic-Like Oral Biofilms as Models for Multispecies Interactions in Endodontic Diseases
by Dejana Lukic, Lamprini Karygianni, Manuela Flury, Thomas Attin and Thomas Thurnheer
Microorganisms 2020, 8(5), 674; https://doi.org/10.3390/microorganisms8050674 - 06 May 2020
Cited by 20 | Viewed by 3360
Abstract
Oral bacteria possess the ability to form biofilms on solid surfaces. After the penetration of oral bacteria into the pulp, the contact between biofilms and pulp tissue may result in pulpitis, pulp necrosis and/or periapical lesion. Depending on the environmental conditions and the [...] Read more.
Oral bacteria possess the ability to form biofilms on solid surfaces. After the penetration of oral bacteria into the pulp, the contact between biofilms and pulp tissue may result in pulpitis, pulp necrosis and/or periapical lesion. Depending on the environmental conditions and the availability of nutrients in the pulp chamber and root canals, mainly Gram-negative anaerobic microorganisms predominate and form the intracanal endodontic biofilm. The objective of the present study was to investigate the role of different substrates on biofilm formation as well as the separate and collective incorporation of six endodontic pathogens, namely Enterococcus faecalis, Staphylococcus aureus, Prevotella nigrescens, Selenomonas sputigena, Parvimonas micra and Treponema denticola into a nine-species “basic biofilm”. This biofilm was formed in vitro as a standard subgingival biofilm, comprising Actinomyces oris, Veillonella dispar, Fusobacterium nucleatum, Streptococcus anginosus, Streptococcus oralis, Prevotella intermedia, Campylobacter rectus, Porphyromonas gingivalis, and Tannerella forsythia. The resulting endodontic-like biofilms were grown 64 h under the same conditions on hydroxyapatite and dentin discs. After harvesting the endodontic-like biofilms, the bacterial growth was determined using quantitative real-time PCR, were labeled using fluorescence in situ hybridization (FISH) and analyzed by confocal laser scanning microscopy (CLSM). The addition of six endodontic pathogens to the “basic biofilm” induced a decrease in the cell number of the “basic” species. Interestingly, C. rectus counts increased in biofilms containing E. faecalis, S. aureus, P. nigrescens and S. sputigena, respectively, both on hydroxyapatite and on dentin discs, whereas P. intermedia counts increased only on dentin discs by addition of E. faecalis. The growth of E. faecalis on hydroxyapatite discs and of E. faecalis and S. aureus on dentin discs were significantly higher in the biofilm containing all species than in the “basic biofilm”. Contrarily, the counts of P. nigrescens, S. sputigena and P. micra on hydroxyapatite discs as well as counts of P. micra and T. denticola on dentin discs decreased in the all-species biofilm. Overall, all bacterial species associated with endodontic infections were successfully incorporated into the standard multispecies biofilm model both on hydroxyapatite and dentin discs. Thus, future investigations on endodontic infections can rely on this newly established endodontic-like multispecies biofilm model. Full article
(This article belongs to the Special Issue Oral Biofilms)
Show Figures

Figure 1

12 pages, 3446 KiB  
Article
Proteolytic Activity of Bacillus subtilis upon κ-Casein Undermines Its “Caries-Safe” Effect
by Danielle Duanis-Assaf, Eli Kenan, Ronit Sionov, Doron Steinberg and Moshe Shemesh
Microorganisms 2020, 8(2), 221; https://doi.org/10.3390/microorganisms8020221 - 06 Feb 2020
Cited by 9 | Viewed by 4485
Abstract
Milk is believed to be a relatively “caries-safe” food. This belief relies on the fact that caseins, which constitute around 80% of milk’s protein content, were found to inhibit the adhesion of Streptococcus mutans to enamel and, therefore, decrease biofilm formation. While S. [...] Read more.
Milk is believed to be a relatively “caries-safe” food. This belief relies on the fact that caseins, which constitute around 80% of milk’s protein content, were found to inhibit the adhesion of Streptococcus mutans to enamel and, therefore, decrease biofilm formation. While S. mutans is considered a leading cause of dental disorders, Bacillus subtilis is a non-pathogenic foodborne bacterium, frequently contaminating milk and its products. This study aimed to investigate the effects of dairy-associated foodborne bacteria such as B. subtilis on biofilm formation by S. mutans in the presence of casein proteins. Our results indicate that there is a significant decrease in total biofilm formation by S. mutans exposed to a casein protein mixture in a mono-species culture, whereas, in the co-culture with B. subtilis, an inhibitory effect of the caseins mixture on S. mutans biofilm formation was observed. Proteolytic activity analysis suggested that B. subtilis is capable of breaking down milk proteins, especially κ-casein, which enables biofilm formation by S. mutans in the presence of milk caseins. Therefore, these findings may challenge the assumption that milk is “caries-safe”, especially in a complex microbial environment. Full article
(This article belongs to the Special Issue Oral Biofilms)
Show Figures

Figure 1

13 pages, 1572 KiB  
Article
Methylcellulose Hydrogel with Melissa officinalis Essential Oil as a Potential Treatment for Oral Candidiasis
by Elisa Serra, Fabien Saubade, Cosimo Ligorio, Kathryn Whitehead, Alastair Sloan, David W. Williams, Araida Hidalgo-Bastida, Joanna Verran and Sladjana Malic
Microorganisms 2020, 8(2), 215; https://doi.org/10.3390/microorganisms8020215 - 06 Feb 2020
Cited by 29 | Viewed by 5636
Abstract
Candida spp. are the most prevalent fungi of the human microbiota and are opportunistic pathogens that can cause oral candidiasis. Management of such infections is limited due to the low number of antifungal drugs available, their relatively high toxicity and the emergence of [...] Read more.
Candida spp. are the most prevalent fungi of the human microbiota and are opportunistic pathogens that can cause oral candidiasis. Management of such infections is limited due to the low number of antifungal drugs available, their relatively high toxicity and the emergence of antifungal resistance. Therefore, much interest in the antimicrobial potential of natural compounds has recently been evident. The use of hydrogels in the delivery of biocides has been explored due to their biocompatibility, ease with drug encapsulation, and due to their potential to confer mechanical and structural properties similar to biological tissue. Methylcellulose hydrogels (10% (w/v)) with 1% (v/v) and 2% (v/v) Melissa officinalis oil were synthesised. The rheological properties and gelation time of the hydrogels were evaluated. Antimicrobial action, the antifungal potential and ability to displace Candida were determined. Rheological tests revealed that the hydrogel jellified in three minutes at 37 °C. Loaded hydrogels successfully inhibited Candida albicans growth as evident by zone of inhibition and time-kill assays. A significant reduction in retained C. albicans was demonstrated with the hydrogel at 2% Melissa officinalis concentration. This work demonstrated that an essential oil-loaded hydrogel had the potential to provide a novel antimicrobial therapy for the treatment of oral candidiasis. Full article
(This article belongs to the Special Issue Oral Biofilms)
Show Figures

Figure 1

15 pages, 2850 KiB  
Article
Streptococcus mutans and Actinomyces naeslundii Interaction in Dual-Species Biofilm
by Rosa Virginia Dutra de Oliveira, Fernanda Salloume Sampaio Bonafé, Denise Madalena Palomari Spolidorio, Cristiane Yumi Koga-Ito, Aline Leite de Farias, Kelly R. Kirker, Garth A. James and Fernanda Lourenção Brighenti
Microorganisms 2020, 8(2), 194; https://doi.org/10.3390/microorganisms8020194 - 31 Jan 2020
Cited by 17 | Viewed by 4486
Abstract
The study of bacterial interaction between Streptococcus mutans and Actinomyces naeslundii may disclose important features of biofilm interspecies relationships. The aim of this study was to characterize—with an emphasis on biofilm formation and composition and metabolic activity—single- and dual-species biofilms of S. mutans [...] Read more.
The study of bacterial interaction between Streptococcus mutans and Actinomyces naeslundii may disclose important features of biofilm interspecies relationships. The aim of this study was to characterize—with an emphasis on biofilm formation and composition and metabolic activity—single- and dual-species biofilms of S. mutans or A. naeslundii, and to use a drip flow reactor (DFR) to evaluate biofilm stress responses to 0.2% chlorhexidine diacetate (CHX). Single- and dual-species biofilms were grown for 24 h. The following factors were evaluated: cell viability, biomass and total proteins in the extracellular matrix, 2,3-bis(2-methoxy-4-nitro-5-sulfophenyl)-2H-tetrazolium-5-carboxanilide—“XTT”—reduction and lactic acid production. To evaluate stress response, biofilms were grown in DFR. Biofilms were treated with CHX or 0.9% sodium chloride (NaCl; control). Biofilms were plated for viability assessment. Confocal laser-scanning microscopy (CLSM) was also performed. Data analysis was carried out at 5% significance level. S. mutans viability and lactic acid production in dual-species biofilms were significantly reduced. S. mutans showed a higher resistance to CHX in dual-species biofilms. Total protein content, biomass and XTT reduction showed no significant differences between single- and dual-species biofilms. CLSM images showed the formation of large clusters in dual-species biofilms. In conclusion, dual-species biofilms reduced S. mutans viability and lactic acid production and increased S. mutans’ resistance to chlorhexidine. Full article
(This article belongs to the Special Issue Oral Biofilms)
Show Figures

Figure 1

17 pages, 3199 KiB  
Article
Role of FAD-I in Fusobacterial Interspecies Interaction and Biofilm Formation
by Bhumika Shokeen, Jane Park, Emily Duong, Sonam Rambhia, Manash Paul, Aaron Weinberg, Wenyuan Shi and Renate Lux
Microorganisms 2020, 8(1), 70; https://doi.org/10.3390/microorganisms8010070 - 02 Jan 2020
Cited by 7 | Viewed by 2860 | Correction
Abstract
RadD, a major adhesin of oral fusobacteria, is part of a four-gene operon encoding the small lipoprotein FAD-I and two currently uncharacterized small proteins encoded by the rapA and rapB genes. Previously, we described a role for FAD-I in the induction of human [...] Read more.
RadD, a major adhesin of oral fusobacteria, is part of a four-gene operon encoding the small lipoprotein FAD-I and two currently uncharacterized small proteins encoded by the rapA and rapB genes. Previously, we described a role for FAD-I in the induction of human B-defensin 2 (hBD2) upon contact with oral epithelial cells. Here, we investigated potential roles for fad-I, rapA, and rapB in interspecies interaction and biofilm formation. Gene inactivation mutants were generated for each of these genes in the nucleatum and polymorphum subspecies of Fusobacterium nucleatum and characterized for their adherence to partner species, biofilm formation, and operon transcription. Binding to Streptococcus gordonii was increased in all mutant strains with Δfad-I having the most significant effect. This increased adherence was directly proportional to elevated radD transcript levels and resulted in significantly different architecture and height of the biofilms formed by Δfad-I and S. gordonii compared to the wild-type parent. In conclusion, FAD-I is important for fusobacterial interspecies interaction as its lack leads to increased production of the RadD adhesin suggesting a role of FAD-I in its regulation. This regulatory effect does not require the presence of functional RadD. Full article
(This article belongs to the Special Issue Oral Biofilms)
Show Figures

Figure 1

23 pages, 5202 KiB  
Article
PgRsp Is a Novel Redox-Sensing Transcription Regulator Essential for Porphyromonas gingivalis Virulence
by Michał Śmiga and Teresa Olczak
Microorganisms 2019, 7(12), 623; https://doi.org/10.3390/microorganisms7120623 - 28 Nov 2019
Cited by 5 | Viewed by 2872
Abstract
Porphyromonas gingivalis is one of the etiological agents of chronic periodontitis. Both heme and oxidative stress impact expression of genes responsible for its survival and virulence. Previously we showed that P. gingivalis ferric uptake regulator homolog affects expression of a gene encoding a [...] Read more.
Porphyromonas gingivalis is one of the etiological agents of chronic periodontitis. Both heme and oxidative stress impact expression of genes responsible for its survival and virulence. Previously we showed that P. gingivalis ferric uptake regulator homolog affects expression of a gene encoding a putative Crp/Fnr superfamily member, termed P. gingivalis redox-sensing protein (PgRsp). Although PgRsp binds heme and shows the highest similarity to proteins assigned to the CooA family, it could be a member of a novel, separate family of proteins with unknown function. Expression of the pgrsp gene is autoregulated and iron/heme dependent. Genes encoding proteins engaged in the oxidative stress response were upregulated in the pgrsp mutant (TO11) strain compared with the wild-type strain. The TO11 strain showed higher biomass production, biofilm formation, and coaggregation ability with Tannerella forsythia and Prevotella intermedia. We suggest that PgRsp may regulate production of virulence factors, proteases, Hmu heme acquisition system, and FimA protein. Moreover, we observed growth retardation of the TO11 strain under oxidative conditions and decreased survival ability of the mutant cells inside macrophages. We conclude that PgRsp protein may play a role in the oxidative stress response using heme as a ligand for sensing changes in redox status, thus regulating the alternative pathway of the oxidative stress response alongside OxyR. Full article
(This article belongs to the Special Issue Oral Biofilms)
Show Figures

Figure 1

Review

Jump to: Research, Other

15 pages, 647 KiB  
Review
Porphyromonas gingivalis, a Long-Range Pathogen: Systemic Impact and Therapeutic Implications
by Hannah Mulhall, Olivier Huck and Salomon Amar
Microorganisms 2020, 8(6), 869; https://doi.org/10.3390/microorganisms8060869 - 09 Jun 2020
Cited by 34 | Viewed by 12376
Abstract
Periodontitis is an inflammatory disease associated with a dysbiosis of the oral flora characterized by a chronic sustained inflammation leading to destruction of tooth-supporting tissues. Over the last decade, an association between periodontitis and systemic disorders such as cardiovascular diseases, rheumatoid arthritis and [...] Read more.
Periodontitis is an inflammatory disease associated with a dysbiosis of the oral flora characterized by a chronic sustained inflammation leading to destruction of tooth-supporting tissues. Over the last decade, an association between periodontitis and systemic disorders such as cardiovascular diseases, rheumatoid arthritis and obesity has been demonstrated. The role of periodontal pathogens, notably Porphyromonas gingivalis (P. gingivalis), in the onset or exacerbation of systemic diseases has been proposed. P. gingivalis expresses several virulence factors that promote its survival, spreading, and sustaining systemic inflammation. Recently, the impact of periodontitis on gut dysbiosis has also been suggested as a potential mechanism underlying the systemic influence of periodontitis. New therapeutic strategies for periodontitis and other dysbiotic conditions, including the use of beneficial microbes to restore healthy microbial flora, may pave the way to improved therapeutic outcomes and more thorough patient management. Full article
(This article belongs to the Special Issue Oral Biofilms)
Show Figures

Figure 1

25 pages, 3078 KiB  
Review
The Microbiome of Peri-Implantitis: A Systematic Review and Meta-Analysis
by Philipp Sahrmann, Fabienne Gilli, Daniel B. Wiedemeier, Thomas Attin, Patrick R. Schmidlin and Lamprini Karygianni
Microorganisms 2020, 8(5), 661; https://doi.org/10.3390/microorganisms8050661 - 01 May 2020
Cited by 82 | Viewed by 6600
Abstract
This review aimed to systematically compare microbial profiles of peri-implantitis to those of periodontitis and healthy implants. Therefore, an electronic search in five databases was conducted. For inclusion, studies assessing the microbiome of peri-implantitis in otherwise healthy patients were considered. Literature was assessed [...] Read more.
This review aimed to systematically compare microbial profiles of peri-implantitis to those of periodontitis and healthy implants. Therefore, an electronic search in five databases was conducted. For inclusion, studies assessing the microbiome of peri-implantitis in otherwise healthy patients were considered. Literature was assessed for consistent evidence of exclusive or predominant peri-implantitis microbiota. Of 158 potentially eligible articles, data of 64 studies on 3730 samples from peri-implant sites were included in this study. Different assessment methods were described in the studies, namely bacterial culture, PCR-based assessment, hybridization techniques, pyrosequencing, and transcriptomic analyses. After analysis of 13 selected culture-dependent studies, no microbial species were found to be specific for peri-implantitis. After assessment of 28 studies using PCR-based methods and a meta-analysis on 19 studies, a higher prevalence of Aggregatibacter actinomycetemcomitans and Prevotella intermedia (log-odds ratio 4.04 and 2.28, respectively) was detected in peri-implantitis biofilms compared with healthy implants. Actinomyces spp., Porphyromonas spp. and Rothia spp. were found in all five pyrosequencing studies in healthy-, periodontitis-, and peri-implantitis samples. In conclusion, the body of evidence does not show a consistent specific profile. Future studies should focus on the assessment of sites with different diagnosis for the same patient, and investigate the complex host-biofilm interaction. Full article
(This article belongs to the Special Issue Oral Biofilms)
Show Figures

Figure 1

Other

Jump to: Research, Review

1 pages, 159 KiB  
Correction
Correction: Shokeen, B., et al. Role of FAD-I in Fusobacterial Interspecies Interaction and Biofilm Formation. Microorganisms 2020, 8, 70
by Bhumika Shokeen, Jane Park, Emily Duong, Sonam Rambhia, Manash Paul, Aaron Weinberg, Wenyuan Shi and Renate Lux
Microorganisms 2021, 9(1), 63; https://doi.org/10.3390/microorganisms9010063 - 29 Dec 2020
Cited by 2 | Viewed by 1395
Abstract
The authors wish to make the following corrections to this paper [...] Full article
(This article belongs to the Special Issue Oral Biofilms)
Back to TopTop