1
|
Makkar H, Sriram G. Advances in modeling periodontal host-microbe interactions: insights from organotypic and organ-on-chip systems. LAB ON A CHIP 2025; 25:1342-1371. [PMID: 39963082 PMCID: PMC11833442 DOI: 10.1039/d4lc00871e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 01/27/2025] [Indexed: 02/20/2025]
Abstract
Periodontal disease, a chronic inflammatory condition affecting the supporting structures of teeth, is driven by an imbalanced interaction between the periodontal microbiota and the host inflammatory response. Beyond its local impact, periodontal disease is associated with systemic conditions such as diabetes mellitus, cardiovascular disease, and inflammatory bowel disease, emphasizing the importance of understanding its mechanisms. Traditional pre-clinical models, such as monolayer cultures and animal studies, have provided foundational insights but are limited by their physiological relevance and ethical concerns. Recent advancements in tissue engineering and microfluidic technologies have led to the development of three-dimensional (3D) organotypic culture models and organ-on-chip systems that more closely mimic native tissue microenvironments. This review provides an overview of the evolution of methods to study periodontal host-microbe interactions, from simple 2D monolayer cultures to complex 3D organotypic and microfluidic organ-on-chip (OoC) models. We discuss various fabrication strategies, host-microbe co-culture techniques, and methods for evaluating outcomes in these advanced models. Additionally, we highlight insights gained from gut-on-chip platforms and their potential applications in periodontal research and understanding oral-systemic links of periodontal disease. Through a comprehensive overview of current advancements and future directions, this review provides insights on the transformative potential of OoC technology in periodontal research, offering new avenues for studying disease mechanisms and developing therapeutic strategies.
Collapse
Affiliation(s)
- Hardik Makkar
- Faculty of Dentistry, National University of Singapore, 119085, Singapore.
- Center for Innovation & Precision Dentistry, School of Dental Medicine and School of Engineering, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| | - Gopu Sriram
- Faculty of Dentistry, National University of Singapore, 119085, Singapore.
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, 117583, Singapore
| |
Collapse
|
2
|
Sriram G, Makkar H. Microfluidic organ-on-chip systems for periodontal research: advances and future directions. Front Bioeng Biotechnol 2025; 12:1490453. [PMID: 39840127 PMCID: PMC11747509 DOI: 10.3389/fbioe.2024.1490453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 12/12/2024] [Indexed: 01/23/2025] Open
Abstract
Advances in tissue engineering and microfluidic technologies have enabled the development of sophisticated in vitro models known as organ-on-a-chip (OoC) or microphysiological systems. These systems enable to potential to simulate the dynamic interactions between host tissues and their microenvironment including microbes, biomaterials, mechanical forces, pharmaceutical, and consumer-care products. These fluidic technologies are increasingly being utilized to investigate host-microbe and host-material interactions in oral health and disease. Of interest is their application in understanding periodontal disease, a chronic inflammatory condition marked by the progressive destruction of periodontal tissues, including gingiva, periodontal ligament, and alveolar bone. The pathogenesis of periodontal disease involves a complex interplay between microbial dysbiosis and host immune responses, which can lead to a loss of dental support structures and contribute to systemic conditions such as cardiovascular disease, diabetes, and inflammatory bowel disease. This provides a comprehensive overview of the latest developments in millifluidic and microfluidic systems designed to emulate periodontal host-microbe and host-material interactions. We discuss the critical engineering and biological considerations in designing these platforms, their applications in studying oral biofilms, periodontal tissue responses, and their potential to unravel disease mechanisms and therapeutic targets in periodontal disease.
Collapse
Affiliation(s)
- Gopu Sriram
- Faculty of Dentistry, National University of Singapore, Singapore, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, Singapore
| | - Hardik Makkar
- Faculty of Dentistry, National University of Singapore, Singapore, Singapore
- Center for Innovation & Precision Dentistry, School of Dental Medicine and School of Engineering & Applied Sciences, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
3
|
Ahmad P, Escalante-Herrera A, Marin LM, Siqueira WL. Progression from healthy periodontium to gingivitis and periodontitis: Insights from bioinformatics-driven proteomics - A systematic review with meta-analysis. J Periodontal Res 2025; 60:8-29. [PMID: 38873831 DOI: 10.1111/jre.13313] [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: 10/02/2023] [Revised: 05/23/2024] [Accepted: 05/26/2024] [Indexed: 06/15/2024]
Abstract
AIM The current study aimed to: (1) systematically review the published literature regarding the proteomics analyses of saliva and gingival crevicular fluid (GCF) in healthy humans and gingivitis and/or periodontitis patients; and (2) to identify the differentially expressed proteins (DEPs) based on the systematic review, and comprehensively conduct meta-analyses and bioinformatics analyses. METHODS An online search of Web of Science, Scopus, and PubMed was performed without any restriction on the year and language of publication. After the identification of the DEPs reported by the included human primary studies, gene ontology (GO), the Kyoto encyclopedia of genes and genomes pathway (KEGG), protein-protein interaction (PPI), and meta-analyses were conducted. The risk of bias among the included studies was evaluated using the modified Newcastle-Ottawa quality assessment scale. RESULTS The review identified significant differences in protein expression between healthy individuals and those with gingivitis and periodontitis. In GCF, 247 proteins were upregulated and 128 downregulated in periodontal diseases. Saliva analysis revealed 79 upregulated and 70 downregulated proteins. There were distinct protein profiles between gingivitis and periodontitis, with 159 and 31 unique upregulated proteins in GCF, respectively. Meta-analyses confirmed significant upregulation of various proteins in periodontitis, including ALB and MMP9, while CSTB and GSTP1 were downregulated. AMY1A and SERPINA1 were upregulated in periodontitis saliva. HBD was upregulated in gingivitis GCF, while DEFA3 was downregulated. PPI analysis revealed complex networks of interactions among DEPs. GO and KEGG pathway analyses provided insights into biological processes and pathways associated with periodontal diseases. CONCLUSION The ongoing MS-based proteomics studies emphasize the need for a highly sensitive and specific diagnostic tool for periodontal diseases. Clinician acceptance of the eventual diagnostic method relies on its ability to provide superior or complementary information to current clinical assessment procedures. Future research should prioritize the multiplex measurement of multiple biomarkers simultaneously to enhance diagnostic accuracy and large study cohorts are necessary to ensure the validity and reliability of research findings.
Collapse
Affiliation(s)
- Paras Ahmad
- College of Dentistry, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | | | - Lina M Marin
- College of Dentistry, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Walter L Siqueira
- College of Dentistry, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| |
Collapse
|
4
|
Silbereisen A, Bao K, Wolski W, Nanni P, Kunz L, Afacan B, Emingil G, Bostanci N. Probing the salivary proteome for prognostic biomarkers in response to non-surgical periodontal therapy. J Clin Periodontol 2025; 52:56-67. [PMID: 38660744 PMCID: PMC11671166 DOI: 10.1111/jcpe.13990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 03/04/2024] [Accepted: 04/09/2024] [Indexed: 04/26/2024]
Abstract
AIM This prospective study investigated the salivary proteome before and after periodontal therapy. MATERIALS AND METHODS Ten systemically healthy, non-smoking, stage III, grade C periodontitis patients underwent non-surgical periodontal treatment. Full-mouth periodontal parameters were measured, and saliva (n = 30) collected pre- (T0), and one (T1) and six (T6) months post-treatment. The proteome was investigated by label-free quantitative proteomics. Protein expression changes were modelled over time, with significant protein regulation considered at false discovery rate <0.05. RESULTS Treatment significantly reduced bleeding scores, percentages of sites with pocket depth ≥5 mm, plaque and gingival indexes. One thousand seven hundred and thirteen proteins were identified and 838 proteins (human = 757, bacterial = 81) quantified (≥2 peptides). At T1, 80 (T1 vs. T0: 60↑:20↓), and at T6, 118 human proteins (T6 vs. T0: 67↑:51↓) were regulated. The salivary proteome at T6 versus T1 remained stable. Highest protein activity post- versus pre-treatment was observed for cellular movement and inflammatory response. The small proline-rich protein 3 (T1 vs. T0: 5.4-fold↑) and lymphocyte-specific protein 1 (T6 vs. T0: 4.6-fold↓) were the top regulated human proteins. Proteins from Neisseria mucosa and Treponema socranskii (T1 vs. T0: 8.0-fold↓, 4.9-fold↓) were down-regulated. CONCLUSIONS Periodontal treatment reduced clinical disease parameters and these changes were reflected in the salivary proteome. This underscores the potential of utilizing saliva biomarkers as prognostic tools for monitoring treatment outcomes.
Collapse
Affiliation(s)
- Angelika Silbereisen
- Division of Oral Health and Periodontology, Department of Dental MedicineKarolinska InstitutetStockholmSweden
| | - Kai Bao
- Division of Oral Health and Periodontology, Department of Dental MedicineKarolinska InstitutetStockholmSweden
| | - Witold Wolski
- Functional Genomics Center ZurichETH Zurich and University of ZurichZurichSwitzerland
| | - Paolo Nanni
- Functional Genomics Center ZurichETH Zurich and University of ZurichZurichSwitzerland
| | - Laura Kunz
- Functional Genomics Center ZurichETH Zurich and University of ZurichZurichSwitzerland
| | - Beral Afacan
- Department of Periodontology, Faculty of DentistryAdnan Menderes UniversityAydınTurkey
| | - Gülnur Emingil
- Department of Periodontology, School of DentistryEge UniversityİzmirTurkey
| | - Nagihan Bostanci
- Division of Oral Health and Periodontology, Department of Dental MedicineKarolinska InstitutetStockholmSweden
| |
Collapse
|
5
|
Machla F, Bekiari C, Monou PK, Kofidou E, Theodosaki AM, Katsamenis OL, Zisis V, Kokoti M, Bakopoulou A, Fatouros D, Andreadis D. Development of an Oral Epithelial Ex Vivo Organ Culture Model for Biocompatibility and Permeability Assessment of Biomaterials. Bioengineering (Basel) 2024; 11:1035. [PMID: 39451410 PMCID: PMC11504994 DOI: 10.3390/bioengineering11101035] [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: 09/15/2024] [Revised: 10/10/2024] [Accepted: 10/15/2024] [Indexed: 10/26/2024] Open
Abstract
In the present study, a customized device (Epi-ExPer) was designed and fabricated to facilitate an epithelial organ culture, allowing for controlled exposure to exogenous chemical stimuli and accommodating the evaluation of permeation of the tissue after treatment. The Epi-ExPer system was fabricated using a stereolithography (SLA)-based additive manufacturing (AM) method. Human and porcine oral epithelial mucosa tissues were inserted into the device and exposed to resinous monomers commonly released by dental restorative materials. The effect of these xenobiotics on the morphology, viability, permeability, and expression of relevant markers of the oral epithelium was evaluated. Tissue culture could be performed with the desired orientation of air-liquid interface (ALI) conditions, and exposure to xenobiotics was undertaken in a spatially guarded and reproducible manner. Among the selected monomers, HEMA and TEGDMA reduced tissue viability at high concentrations, while tissue permeability was increased by the latter. Xenobiotics affected the histological image by introducing the vacuolar degeneration of epithelial cells and increasing the expression of panCytokeratin (pCK). Epi-ExPer device offers a simple, precise, and reproducible study system to evaluate interactions of oral mucosa with external stimuli, providing a biocompatibility and permeability assessment tool aiming to an enhanced in vitro/ex vivo-to-in vivo extrapolation (IVIVE) that complies with European Union (EU) and Food and Durg Administration (FDI) policies.
Collapse
Affiliation(s)
- Foteini Machla
- Department of Prosthodontics, Dental and Craniofacial Bioengineering and Applied Biomaterials, School of Dentistry, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece (A.M.T.)
| | - Chrysanthi Bekiari
- Laboratory of Anatomy and Histology, Veterinary School, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (C.B.); (E.K.)
| | - Paraskevi Kyriaki Monou
- Department of Pharmaceutical Technology, School of Pharmacy, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (P.K.M.); (D.F.)
- Center for Interdisciplinary Research and Innovation (CIRI-AUTH), 57001 Thessaloniki, Greece
| | - Evangelia Kofidou
- Laboratory of Anatomy and Histology, Veterinary School, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (C.B.); (E.K.)
| | - Astero Maria Theodosaki
- Department of Prosthodontics, Dental and Craniofacial Bioengineering and Applied Biomaterials, School of Dentistry, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece (A.M.T.)
| | - Orestis L. Katsamenis
- μ-VIS X-ray Imaging Centre, Faculty of Engineering and the Environment, University of Southampton, Southampton SO17 1BJ, UK
- Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
| | - Vasileios Zisis
- Department of Oral Medicine/Pathology, School of Dentistry, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Maria Kokoti
- Department of Prosthodontics, Dental and Craniofacial Bioengineering and Applied Biomaterials, School of Dentistry, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece (A.M.T.)
| | - Athina Bakopoulou
- Department of Prosthodontics, Dental and Craniofacial Bioengineering and Applied Biomaterials, School of Dentistry, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece (A.M.T.)
| | - Dimitrios Fatouros
- Department of Pharmaceutical Technology, School of Pharmacy, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (P.K.M.); (D.F.)
- Center for Interdisciplinary Research and Innovation (CIRI-AUTH), 57001 Thessaloniki, Greece
| | - Dimitrios Andreadis
- Department of Oral Medicine/Pathology, School of Dentistry, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| |
Collapse
|
6
|
Machla F, Monou PK, Bekiari C, Andreadis D, Kofidou E, Panteris E, Katsamenis OL, Kokoti M, Koidis P, About I, Fatouros D, Bakopoulou A. Tissue-Engineered Oral Epithelium for Dental Material Testing: Toward In Vitro Biomimetic Models. Tissue Eng Part C Methods 2024. [PMID: 39302070 DOI: 10.1089/ten.tec.2024.0154] [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: 09/22/2024] Open
Abstract
Tissue-engineered oral epithelium (ΤΕΟΕ) was developed after comparing various culture conditions, including submerged (SUB) and air-liquid interface (ALI) human cell expansion options. Barrier formation was evaluated via transepithelial electrical resistance (TEER) and calcein permeation via spectrofluorometry. TEOE was further assessed for long-term viability via live/dead staining and development of intercellular connections via transmission electron microscopy. Tissue architecture was evaluated via histochemistry and the expression of pancytokeratin (pCK) via immunohistochemistry. The effect of two commonly used dental resinous monomers on TEOE was evaluated for alterations in cell viability and barrier permeability. ALI/keratinocyte growth factor-supplemented (ALI-KGS) culture conditions led to the formation of an 8-20-layer thick, intercellularly connected epithelial barrier. TEER values of ALI-KGS-developed TEOE decreased compared with all other tested conditions, and the established epithelium intensively expressed pCK. Exposure to dental monomers affected the integrity and architecture of TEOE and induced cellular vacuolation, implicating hydropic degeneration. Despite structural modifications, the permeability of TEOE was not substantially affected after exposure to the monomers. In conclusion, the biological properties of the TEOE mimicking the physiological functional conditions and its value as biocompatibility assessment tool for dental materials were characterized.
Collapse
Affiliation(s)
- Foteini Machla
- Department of Prosthodontics, Tissue Engineering Core Unit, School of Dentistry, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Paraskevi Kyriaki Monou
- Department of Pharmaceutical Technology, School of Pharmacy, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
- Center for Interdisciplinary Research and Innovation (CIRI-AUTH), Thessaloniki, Greece
| | - Chrysanthi Bekiari
- Laboratory of Anatomy and Histology, School of Veterinary Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Dimitrios Andreadis
- Department of Oral Medicine/Pathology, School of Dentistry, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Evangelia Kofidou
- Laboratory of Anatomy and Histology, School of Veterinary Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Emmanuel Panteris
- Department of Botany, School of Biology, Faculty of Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Orestis L Katsamenis
- μ-VIS X-ray Imaging Centre, Faculty of Engineering and the Environment, University of Southampton, Southampton, United Kingdom
- Institute for Life Sciences, University of Southampton, Southampton, United Kingdom
| | - Maria Kokoti
- Department of Prosthodontics, Tissue Engineering Core Unit, School of Dentistry, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Petros Koidis
- Department of Prosthodontics, Tissue Engineering Core Unit, School of Dentistry, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Imad About
- Centre National de la Recherche Scientifique, Institute of Movement Sciences, Aix Marseille University, Marseille, France
| | - Dimitrios Fatouros
- Department of Pharmaceutical Technology, School of Pharmacy, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
- Center for Interdisciplinary Research and Innovation (CIRI-AUTH), Thessaloniki, Greece
| | - Athina Bakopoulou
- Department of Prosthodontics, Tissue Engineering Core Unit, School of Dentistry, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
7
|
Shang L, Deng D, Krom BP, Gibbs S. Oral host-microbe interactions investigated in 3D organotypic models. Crit Rev Microbiol 2024; 50:397-416. [PMID: 37166371 DOI: 10.1080/1040841x.2023.2211665] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 04/30/2023] [Accepted: 05/02/2023] [Indexed: 05/12/2023]
Abstract
The oral cavity is inhabited by abundant microbes which continuously interact with the host and influence the host's health. Such host-microbe interactions (HMI) are dynamic and complex processes involving e.g. oral tissues, microbial communities and saliva. Due to difficulties in mimicking the in vivo complexity, it is still unclear how exactly HMI influence the transition between healthy status and disease conditions in the oral cavity. As an advanced approach, three-dimensional (3D) organotypic oral tissues (epithelium and mucosa/gingiva) are being increasingly used to study underlying mechanisms. These in vitro models were designed with different complexity depending on the research questions to be answered. In this review, we summarised the existing 3D oral HMI models, comparing designs and readouts, discussing applications as well as future perspectives.
Collapse
Affiliation(s)
- Lin Shang
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Dongmei Deng
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Bastiaan P Krom
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Susan Gibbs
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Department of Molecular Cell Biology and Immunology, Amsterdam Infection and Immunity Institute, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
8
|
Manoil D, Parga A, Bostanci N, Belibasakis GN. Microbial diagnostics in periodontal diseases. Periodontol 2000 2024; 95:176-193. [PMID: 38797888 DOI: 10.1111/prd.12571] [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: 02/06/2024] [Revised: 03/27/2024] [Accepted: 04/15/2024] [Indexed: 05/29/2024]
Abstract
Microbial analytical methods have been instrumental in elucidating the complex microbial etiology of periodontal diseases, by shaping our understanding of subgingival community dynamics. Certain pathobionts can orchestrate the establishment of dysbiotic communities that can subvert the host immune system, triggering inflammation and tissue destruction. Yet, diagnosis and management of periodontal conditions still rely on clinical and radiographic examinations, overlooking the well-established microbial etiology. This review summarizes the chronological emergence of periodontal etiological models and the co-evolution with technological advances in microbial detection. We additionally review the microbial analytical approaches currently accessible to clinicians, highlighting their value in broadening the periodontal assessment. The epidemiological importance of obtaining culture-based antimicrobial susceptibility profiles of periodontal taxa for antibiotic resistance surveillance is also underscored, together with clinically relevant analytical approaches to guide antibiotherapy choices, when necessary. Furthermore, the importance of 16S-based community and shotgun metagenomic profiling is discussed in outlining dysbiotic microbial signatures. Because dysbiosis precedes periodontal damage, biomarker identification offers early diagnostic possibilities to forestall disease relapses during maintenance. Altogether, this review highlights the underutilized potential of clinical microbiology in periodontology, spotlighting the clinical areas most conductive to its diagnostic implementation for enhancing prevention, treatment predictability, and addressing global antibiotic resistance.
Collapse
Affiliation(s)
- Daniel Manoil
- Division of Cariology and Endodontics, University Clinics of Dental Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Division of Oral Health and Periodontology, Department of Dental Medicine, Karolinska Institutet, Huddinge, Stockholm, Sweden
| | - Ana Parga
- Division of Cariology and Endodontics, University Clinics of Dental Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Department of Microbiology and Parasitology, CIBUS-Faculty of Biology, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Nagihan Bostanci
- Division of Oral Health and Periodontology, Department of Dental Medicine, Karolinska Institutet, Huddinge, Stockholm, Sweden
| | - Georgios N Belibasakis
- Division of Oral Health and Periodontology, Department of Dental Medicine, Karolinska Institutet, Huddinge, Stockholm, Sweden
| |
Collapse
|
9
|
Halstenbach T, Topitsch A, Schilling O, Iglhaut G, Nelson K, Fretwurst T. Mass spectrometry-based proteomic applications in dental implants research. Proteomics Clin Appl 2024; 18:e2300019. [PMID: 38342588 DOI: 10.1002/prca.202300019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 12/07/2023] [Accepted: 12/21/2023] [Indexed: 02/13/2024]
Abstract
Dental implants have been established as successful treatment options for missing teeth with steadily increasing demands. Today, the primary areas of research in dental implantology revolve around osseointegration, soft and hard tissue grafting as well as peri-implantitis diagnostics, prevention, and treatment. This review provides a comprehensive overview of the current literature on the application of MS-based proteomics in dental implant research, highlights how explorative proteomics provided insights into the biology of peri-implant soft and hard tissues and how proteomics facilitated the stratification between healthy and diseased implants, enabling the identification of potential new diagnostic markers. Additionally, this review illuminates technical aspects, and provides recommendations for future study designs based on the current evidence.
Collapse
Affiliation(s)
- Tim Halstenbach
- Department of Oral- and Craniomaxillofacial Surgery/Translational Implantology, Division of Regenerative Oral Medicine, Faculty of Medicine, Medical Center - University of Freiburg, Freiburg, Germany
| | - Annika Topitsch
- Department of Oral- and Craniomaxillofacial Surgery/Translational Implantology, Division of Regenerative Oral Medicine, Faculty of Medicine, Medical Center - University of Freiburg, Freiburg, Germany
- Institute of Surgical Pathology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Oliver Schilling
- Institute of Surgical Pathology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Gerhard Iglhaut
- Department of Oral- and Craniomaxillofacial Surgery/Translational Implantology, Division of Regenerative Oral Medicine, Faculty of Medicine, Medical Center - University of Freiburg, Freiburg, Germany
| | - Katja Nelson
- Department of Oral- and Craniomaxillofacial Surgery/Translational Implantology, Division of Regenerative Oral Medicine, Faculty of Medicine, Medical Center - University of Freiburg, Freiburg, Germany
| | - Tobias Fretwurst
- Department of Oral- and Craniomaxillofacial Surgery/Translational Implantology, Division of Regenerative Oral Medicine, Faculty of Medicine, Medical Center - University of Freiburg, Freiburg, Germany
| |
Collapse
|
10
|
Liu Z, Du X, Xu L, Shi Q, Tang X, Cao Y, Song K. The therapeutic perspective of cold atmospheric plasma in periodontal disease. Oral Dis 2024; 30:938-948. [PMID: 36825384 DOI: 10.1111/odi.14547] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/31/2023] [Accepted: 02/21/2023] [Indexed: 02/25/2023]
Abstract
OBJECTIVES Periodontal disease (PD) is one of the most common infectious diseases with complex inflammatory conditions, having irreversibly destructive impacts on the periodontal supporting tissues. The application of cold atmospheric plasma (CAP) is a promising adjuvant therapy modality for PD. However, the mechanism of CAP in PD treatment is still poorly understood. The review motivates to outline the latest researches concerning the applications of CAP in PD treatment. METHODS We searched CAP-related literature through utilizing the well-established databases of Pubmed, Scopus and Web of Science according to the following keywords related to periodontal disease (periodontal, gingival, gingivitis, gingiva, periodontium, periodontitis). RESULTS A total of 18 concerning original studies were found. These studies could be classified according to three pathophysiological perspectives of PD. The therapeutic mechanisms of CAP may be attributed to the oxidative stress-related cell death of periodontal bacteria, the suppression of periodontal inflammation and pro-inflammatory cytokine secretion, as well as the acceleration of periodontal soft tissue wound healing and hard tissue reconstruction. CONCLUSIONS Cold atmospheric plasma has potential therapeutic effects on PD through three mechanisms: antimicrobial effect, inflammation attenuation, and tissue remodeling. This review hopefully provides a comprehensive perspective into the potential of CAP in PD therapy.
Collapse
Affiliation(s)
- Zhixin Liu
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Prosthodontics and Implantology, School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Xijin Du
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Prosthodontics and Implantology, School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Lianyi Xu
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Prosthodontics and Implantology, School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Qi Shi
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Prosthodontics and Implantology, School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Xuezhi Tang
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Prosthodontics and Implantology, School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Yingguang Cao
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Prosthodontics and Implantology, School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Ke Song
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Prosthodontics and Implantology, School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| |
Collapse
|
11
|
Muniraj G, Tan RHS, Dai Y, Wu R, Alberti M, Sriram G. Microphysiological Modeling of Gingival Tissues and Host-Material Interactions Using Gingiva-on-Chip. Adv Healthc Mater 2023; 12:e2301472. [PMID: 37758297 PMCID: PMC11468103 DOI: 10.1002/adhm.202301472] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 09/14/2023] [Indexed: 10/03/2023]
Abstract
Gingiva plays a crucial barrier role at the interface of teeth, tooth-supporting structures, microbiome, and external agents. To mimic this complex microenvironment, an in vitro microphysiological platform and biofabricated full-thickness gingival equivalents (gingiva-on-chip) within a vertically stacked microfluidic device is developed. This design allowed long-term and air-liquid interface culture, and host-material interactions under flow conditions. Compared to static cultures, dynamic cultures on-chip enabled the biofabrication of gingival equivalents with stable mucosal matrix, improved epithelial morphogenesis, and barrier features. Additionally, a diseased state with disrupted barrier function representative of gingival/oral mucosal ulcers is modeled. The apical flow feature is utilized to emulate the mechanical action of mouth rinse and integrate the assessment of host-material interactions and transmucosal permeation of oral-care formulations in both healthy and diseased states. Although the gingiva-on-chip cultures have thicker and more mature epithelium, the flow of oral-care formulations induced increased tissue disruption and cytotoxic features compared to static conditions. The realistic emulation of mouth rinsing action facilitated a more physiological assessment of mucosal irritation potential. Overall, this microphysiological system enables biofabrication of human gingiva equivalents in intact and ulcerated states, providing a miniaturized and integrated platform for downstream host-material and host-microbiome applications in gingival and oral mucosa research.
Collapse
Affiliation(s)
- Giridharan Muniraj
- Faculty of DentistryNational University of SingaporeSingapore119085Singapore
| | - Rachel Hui Shuen Tan
- Singapore Institute of Manufacturing Technology (SIMTech)Agency for Science, Technology and Research (A*STAR)Singapore138634Singapore
| | - Yichen Dai
- Faculty of DentistryNational University of SingaporeSingapore119085Singapore
| | - Ruige Wu
- Singapore Institute of Manufacturing Technology (SIMTech)Agency for Science, Technology and Research (A*STAR)Singapore138634Singapore
| | - Massimo Alberti
- Singapore Institute of Manufacturing Technology (SIMTech)Agency for Science, Technology and Research (A*STAR)Singapore138634Singapore
- REVIVO BioSystems Pte. Ltd.Singapore138623Singapore
| | - Gopu Sriram
- Faculty of DentistryNational University of SingaporeSingapore119085Singapore
- ORCHIDS: Oral Care Health Innovations and Designs SingaporeNational University of SingaporeSingapore119085Singapore
- NUS Centre for Additive Manufacturing (AM.NUS)National University of SingaporeSingapore117602Singapore
| |
Collapse
|
12
|
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
|
13
|
Halstenbach T, Nelson K, Iglhaut G, Schilling O, Fretwurst T. Impact of peri-implantitis on the proteome biology of crevicular fluid: A pilot study. J Periodontol 2023; 94:835-847. [PMID: 36585920 DOI: 10.1002/jper.22-0461] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 11/22/2022] [Accepted: 12/15/2022] [Indexed: 01/01/2023]
Abstract
BACKGROUND The proteome of the peri-implant crevicular fluid (PICF) has not been systematically investigated. The aim of the present study was to reveal the proteome biology of dental implants affected with peri-implantitis. METHODS Patients with at least one diseased implant were included (probing depth ≥6 mm, ≥3 mm peri-implant radiological bone loss). Using sterile paper strips, samples were collected from healthy implants (I), healthy teeth (T) and peri-implantitis affected implants (P). Proteome analysis was performed using liquid chromatography - tandem mass spectrometry (LC-MS/MS) and data independent acquisition, allowing the identification and quantification of human and bacterial proteins as well as semi-specific peptides. RESULTS A total of 38 samples from 14 patients were included in the study; 2332 different human proteins were identified across all samples. No differentially expressed proteins between T and I were found. Comparing P to I, 59 proteins were found upregulated and 31 downregulated in P with significance. Upregulated proteins included proinflammatory proteins such as immunoglobulins, dysferlin, and S100P, as well as antimicrobial proteins, for example, myeloperoxidase or azurocidin. Gene ontology analysis further revealed higher activity of immunological pathways. Proteolytic patterns indicated the activity of inflammatory proteins such as cathepsin G. A total of 334 bacterial proteins were identified and quantified. Peri-implantitis showed elevated proteolytic activity. CONCLUSION I and T share similarities in their proteome, while diseased implants deviate strongly from healthy conditions. The PICF proteome of peri-implantitis affected sites exhibits an inflammatory fingerprint, dominated by neutrophil activity when compared with healthy implants.
Collapse
Affiliation(s)
- Tim Halstenbach
- Department of Oral- and Craniomaxillofacial Surgery/Translational Implantology, Division of Regenerative Oral Medicine, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg im Breisgau, Germany
- Institute of Surgical Pathology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg im Breisgau, Germany
| | - Katja Nelson
- Department of Oral- and Craniomaxillofacial Surgery/Translational Implantology, Division of Regenerative Oral Medicine, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg im Breisgau, Germany
| | - Gerhard Iglhaut
- Department of Oral- and Craniomaxillofacial Surgery/Translational Implantology, Division of Regenerative Oral Medicine, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg im Breisgau, Germany
| | - Oliver Schilling
- Institute of Surgical Pathology, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg im Breisgau, Germany
| | - Tobias Fretwurst
- Department of Oral- and Craniomaxillofacial Surgery/Translational Implantology, Division of Regenerative Oral Medicine, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg im Breisgau, Germany
| |
Collapse
|
14
|
Ardila CM, Jiménez-Arbeláez GA, Vivares-Builes AM. Potential Clinical Application of Organs-on-a-Chip in Periodontal Diseases: A Systematic Review of In Vitro Studies. Dent J (Basel) 2023; 11:158. [PMID: 37504224 PMCID: PMC10378380 DOI: 10.3390/dj11070158] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 06/15/2023] [Accepted: 06/23/2023] [Indexed: 07/29/2023] Open
Abstract
The periodontium is a unique organ from the standpoint of building an organ-on-a-chip (OoC) since it is a system that is continually threatened by microorganisms, their noxious compounds, and antigenic components. At the same time, periodontal health depends on a balanced connection between the host and the bacteria in the oral cavity, which is a complex micro-ecological environment. The objective of this systematic review of in vitro studies is to revise the potential clinical application of OoC in periodontal diseases. PRISMA was used to guide this analysis. The review framework made use of several databases, including SCOPUS, PubMed/MEDLINE, SCIELO, and LILACS as well as the gray literature. This systematic review comprised seven studies. The clinical efficacy of OoC in periodontal diseases was observed in models of the gingival crevice for the research of periodontitis, periodontal medication analysis, the interaction of multiple microbial species, pH measurements in in situ-grown biofilm, testing antimicrobial reagents, evaluation of mucosal interactions with microorganisms, and a device for quantitative exploration of microorganisms. OoC has the potential to advance our understanding of periodontal diseases by providing a more accurate representation of the oral microenvironment and enabling the development of new treatments.
Collapse
Affiliation(s)
- Carlos M. Ardila
- Basic Studies Department, School of Dentistry, Universidad de Antioquia UdeA, Medellín 050010, Colombia
| | - Gustavo A. Jiménez-Arbeláez
- School of Dentistry, Institución Universitaria Visión de Las Américas, Medellín 050031, Colombia; (G.A.J.-A.); (A.M.V.-B.)
| | - Annie Marcela Vivares-Builes
- School of Dentistry, Institución Universitaria Visión de Las Américas, Medellín 050031, Colombia; (G.A.J.-A.); (A.M.V.-B.)
| |
Collapse
|
15
|
Danella EB, Costa de Medeiros M, D'Silva NJ. Cytokines secreted by inflamed oral mucosa: implications for oral cancer progression. Oncogene 2023; 42:1159-1165. [PMID: 36879116 DOI: 10.1038/s41388-023-02649-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 02/10/2023] [Accepted: 02/23/2023] [Indexed: 03/08/2023]
Abstract
The oral mucosa has an essential role in protecting against physical, microbial, and chemical harm. Compromise of this barrier triggers a wound healing response. Key events in this response such as immune infiltration, re-epithelialization, and stroma remodeling are coordinated by cytokines that promote cellular migration, invasion, and proliferation. Cytokine-mediated cellular invasion and migration are also essential features in cancer dissemination. Therefore, exploration of cytokines that regulate each stage of oral wound healing will provide insights about cytokines that are exploited by oral squamous cell carcinoma (SCC) to promote tumor development and progression. This will aid in identifying potential therapeutic targets to constrain SCC recurrence and increase patient survival. In this review, we discuss cytokines that overlap in oral wounds and SCC, emphasizing how these cytokines promote cancer progression.
Collapse
Affiliation(s)
- Erika B Danella
- Periodontics and Oral Medicine, University of Michigan School of Dentistry, 1011 N. University Ave, Ann Arbor, MI, USA
| | - Marcell Costa de Medeiros
- Periodontics and Oral Medicine, University of Michigan School of Dentistry, 1011 N. University Ave, Ann Arbor, MI, USA
| | - Nisha J D'Silva
- Periodontics and Oral Medicine, University of Michigan School of Dentistry, 1011 N. University Ave, Ann Arbor, MI, USA. .,Pathology, University of Michigan Medical School, 1500 E Medical Center Dr, Ann Arbor, MI, USA. .,Rogel Cancer Center, 1500 E Medical Center Dr, Ann Arbor, MI, USA.
| |
Collapse
|
16
|
Makkar H, Zhou Y, Tan KS, Lim CT, Sriram G. Modeling Crevicular Fluid Flow and Host-Oral Microbiome Interactions in a Gingival Crevice-on-Chip. Adv Healthc Mater 2023; 12:e2202376. [PMID: 36398428 DOI: 10.1002/adhm.202202376] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 11/07/2022] [Indexed: 11/21/2022]
Abstract
Gingival crevice and gingival crevicular fluid (GCF) flow play a crucial role at the gingiva-oral microbiome interface which contributes toward maintaining the balance between gingival health and periodontal disease. Interstitial flow of GCF strongly impacts the host-microbiome interactions and tissue responses. However, currently available in vitro preclinical models largely disregard the dynamic nature of gingival crevicular microenvironment, thus limiting the progress in the development of periodontal therapeutics. Here, a proof-of-principle "gingival crevice-on-chip" microfluidic platform to culture gingival connective tissue equivalent (CTE) under dynamic interstitial fluid flow mimicking the GCF is described. On-chip co-culture using oral symbiont (Streptococcus oralis) shows the potential to recapitulate microbial colonization, formation of biofilm-like structures at the tissue-microbiome interface, long-term co-culture, and bacterial clearance secondary to simulated GCF (s-GCF) flow. Further, on-chip exposure of the gingival CTEs to the toll-like receptor-2 (TLR-2) agonist or periodontal pathogen Fusobacterium nucleatum demonstrates the potential to mimic early gingival inflammation. In contrast to direct exposure, the induction of s-GCF flow toward the bacterial front attenuates the secretion of inflammatory mediators demonstrating the protective effect of GCF flow. This proposed in vitro platform offers the potential to study complex host-microbe interactions in periodontal disease and the development of periodontal therapeutics under near-microphysiological conditions.
Collapse
Affiliation(s)
- Hardik Makkar
- Faculty of Dentistry, National University of Singapore, Singapore, 119085, Singapore
| | - Ying Zhou
- Institute for Health Innovation and Technology (iHealthtech), National University of Singapore, Singapore, 117599, 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
| | - 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
| | - 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
|
17
|
Makkar H, Atkuru S, Tang YL, Sethi T, Lim CT, Tan KS, Sriram G. Differential immune responses of 3D gingival and periodontal connective tissue equivalents to microbial colonization. J Tissue Eng 2022; 13:20417314221111650. [PMID: 35923175 PMCID: PMC9340411 DOI: 10.1177/20417314221111650] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 06/20/2022] [Indexed: 11/15/2022] Open
Abstract
Gingival and periodontal ligament fibroblasts are functionally distinct cell
types within the dento-gingival unit that participate in host immune response.
Their microenvironment influences the behavior and immune response to microbial
challenge. We developed three-dimensional gingival and periodontal connective
tissue equivalents (CTEs) using human fibrin-based matrix. The CTEs were
characterized, and the heterogeneity in their innate immune response was
investigated. The CTEs demonstrated no to minimal response to planktonic
Streptococcus mitis and Streptococcus
oralis, while their biofilms elicited a moderate increase in IL-6
and IL-8 production. In contrast, Fusobacterium nucleatum
provoked a substantial increase in IL-6 and IL-8 production. Interestingly, the
gingival CTEs secreted significantly higher IL-6, while periodontal counterparts
produced higher IL-8. In conclusion, the gingival and periodontal CTEs exhibited
differential responses to various bacterial challenges. This gives insights into
the contribution of tissue topography and fibroblast heterogeneity in rendering
protective and specific immune responses toward early biofilm colonizers.
Collapse
Affiliation(s)
- Hardik Makkar
- Faculty of Dentistry, National University of Singapore, Singapore
| | - Srividya Atkuru
- Faculty of Dentistry, National University of Singapore, Singapore
| | - Yi Ling Tang
- Faculty of Dentistry, National University of Singapore, Singapore
| | - Tanya Sethi
- Faculty of Dentistry, National University of Singapore, Singapore
| | - Chwee Teck Lim
- Institute for Health Innovation and Technology (iHealthtech), National University of Singapore, Singapore
| | - Kai Soo Tan
- Faculty of Dentistry, National University of Singapore, Singapore
| | - Gopu Sriram
- Faculty of Dentistry, National University of Singapore, Singapore
| |
Collapse
|
18
|
Validation and verification of predictive salivary biomarkers for oral health. Sci Rep 2021; 11:6406. [PMID: 33742017 PMCID: PMC7979790 DOI: 10.1038/s41598-021-85120-w] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 02/24/2021] [Indexed: 02/06/2023] Open
Abstract
Oral health is important not only due to the diseases emerging in the oral cavity but also due to the direct relation to systemic health. Thus, early and accurate characterization of the oral health status is of utmost importance. There are several salivary biomarkers as candidates for gingivitis and periodontitis, which are major oral health threats, affecting the gums. These need to be verified and validated for their potential use as differentiators of health, gingivitis and periodontitis status, before they are translated to chair-side for diagnostics and personalized monitoring. We aimed to measure 10 candidates using high sensitivity ELISAs in a well-controlled cohort of 127 individuals from three groups: periodontitis (60), gingivitis (31) and healthy (36). The statistical approaches included univariate statistical tests, receiver operating characteristic curves (ROC) with the corresponding Area Under the Curve (AUC) and Classification and Regression Tree (CART) analysis. The main outcomes were that the combination of multiple biomarker assays, rather than the use of single ones, can offer a predictive accuracy of > 90% for gingivitis versus health groups; and 100% for periodontitis versus health and periodontitis versus gingivitis groups. Furthermore, ratios of biomarkers MMP-8, MMP-9 and TIMP-1 were also proven to be powerful differentiating values compared to the single biomarkers.
Collapse
|
19
|
Abstract
Plasma is an electrically conducting medium that responds to electric and magnetic fields. It consists of large quantities of highly reactive species, such as ions, energetic electrons, exited atoms and molecules, ultraviolet photons, and metastable and active radicals. Non-thermal or cold plasmas are partially ionized gases whose electron temperatures usually exceed several tens of thousand degrees K, while the ions and neutrals have much lower temperatures. Due to the presence of reactive species at low temperature, the biological effects of non-thermal plasmas have been studied for application in the medical area with promising results. This review outlines the application of cold atmospheric pressure plasma (CAPP) in dentistry for the control of several pathogenic microorganisms, induction of anti-inflammatory, tissue repair effects and apoptosis of cancer cells, with low toxicity to healthy cells. Therefore, CAPP has potential to be applied in many areas of dentistry such as cariology, periodontology, endodontics and oral oncology.
Collapse
|
20
|
Fragkioudakis I, Riggio MP, Apatzidou DA. Understanding the microbial components of periodontal diseases and periodontal treatment-induced microbiological shifts. J Med Microbiol 2020; 70. [PMID: 33295858 DOI: 10.1099/jmm.0.001247] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
In the mid-1960s the microbial aetiology of periodontal diseases was introduced based on classical experimental gingivitis studies . Since then, numerous studies have addressed the fundamental role that oral microbiota plays in the initiation and progression of periodontal diseases. Recent advances in laboratory identification techniques have contributed to a better understanding of the complexity of the oral microbiome in both health and disease. Modern culture-independent methods such as human oral microbial identification microarray and next-generation sequencing have been used to identify a wide variety of microbial taxa residing in the gingival sulcus and the periodontal pocket. The first theory of the 'non-specific plaque' hypothesis gave rise to the 'ecological plaque' hypothesis and more recently to the 'polymicrobial synergy and dysbiosis hypothesis'. Periodontitis is now considered to be a multimicrobial inflammatory disease in which the various bacterial species within the dental biofilm are in a dysbiotic state and this imbalance favours the establishment of chronic inflammatory conditions and ultimately the destruction of tooth-supporting tissues. Apart from the known putative periodontal pathogens, the whole biofilm community is now considered to play a role in the establishment of inflammation and the initiation and progression of periodontitis in a susceptible host. Treatment is unlikely to eliminate putative pathogens but, when it is thoroughly performed it has the potential to establish a healthy ecosystem by altering the microbial community in numbers and composition and also contribute to the maturation of the host immune response.
Collapse
Affiliation(s)
- Ioannis Fragkioudakis
- Department of Preventive Dentistry, Periodontology and Implant Biology, Faculty of Dentistry, Aristotle University of Thessaloniki, Greece
| | - Marcello P Riggio
- Oral Sciences Research Group, Dental School, College of Medical Veterinary and Life Sciences, University of Glasgow, UK
| | - Danae Anastasia Apatzidou
- Department of Preventive Dentistry, Periodontology and Implant Biology, Faculty of Dentistry, Aristotle University of Thessaloniki, Greece
| |
Collapse
|
21
|
Seneviratne CJ, Suriyanarayanan T, Widyarman AS, Lee LS, Lau M, Ching J, Delaney C, Ramage G. Multi-omics tools for studying microbial biofilms: current perspectives and future directions. Crit Rev Microbiol 2020; 46:759-778. [PMID: 33030973 DOI: 10.1080/1040841x.2020.1828817] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The advent of omics technologies has greatly improved our understanding of microbial biology, particularly in the last two decades. The field of microbial biofilms is, however, relatively new, consolidated in the 1980s. The morphogenic switching by microbes from planktonic to biofilm phenotype confers numerous survival advantages such as resistance to desiccation, antibiotics, biocides, ultraviolet radiation, and host immune responses, thereby complicating treatment strategies for pathogenic microorganisms. Hence, understanding the mechanisms governing the biofilm phenotype can result in efficient treatment strategies directed specifically against molecular markers mediating this process. The application of omics technologies for studying microbial biofilms is relatively less explored and holds great promise in furthering our understanding of biofilm biology. In this review, we provide an overview of the application of omics tools such as transcriptomics, proteomics, and metabolomics as well as multi-omics approaches for studying microbial biofilms in the current literature. We also highlight how the use of omics tools directed at various stages of the biological information flow, from genes to metabolites, can be integrated via multi-omics platforms to provide a holistic view of biofilm biology. Following this, we propose a future artificial intelligence-based multi-omics platform that can predict the pathways associated with different biofilm phenotypes.
Collapse
Affiliation(s)
- Chaminda J Seneviratne
- Singapore Oral Microbiomics Initiative (SOMI), National Dental Research Institute Singapore, National Dental Centre, Singapore, Singapore.,Duke NUS Medical School, Singapore, Singapore
| | - Tanujaa Suriyanarayanan
- Singapore Oral Microbiomics Initiative (SOMI), National Dental Research Institute Singapore, National Dental Centre, Singapore, Singapore.,Duke NUS Medical School, Singapore, Singapore
| | - Armelia Sari Widyarman
- Department of Microbiology, Faculty of Dentistry, Trisakti University, Grogol, West Jakarta, Indonesia
| | - Lye Siang Lee
- Duke-NUS Medical School, Metabolomics Lab, Cardiovascular and Metabolic Disorders, Singapore, Singapore
| | - Matthew Lau
- Singapore Oral Microbiomics Initiative (SOMI), National Dental Research Institute Singapore, National Dental Centre, Singapore, Singapore
| | - Jianhong Ching
- Duke-NUS Medical School, Metabolomics Lab, Cardiovascular and Metabolic Disorders, Singapore, Singapore
| | - Christopher Delaney
- School of Medicine, Dentistry & Nursing, Glasgow Dental Hospital & School, University of Glasgow, Glasgow, UK
| | - Gordon Ramage
- School of Medicine, Dentistry & Nursing, Glasgow Dental Hospital & School, University of Glasgow, Glasgow, UK
| |
Collapse
|
22
|
Bostanci N, Silbereisen A, Bao K, Grossmann J, Nanni P, Fernandez C, Nascimento GG, Belibasakis GN, Lopez R. Salivary proteotypes of gingivitis tolerance and resilience. J Clin Periodontol 2020; 47:1304-1316. [PMID: 32777086 PMCID: PMC7692908 DOI: 10.1111/jcpe.13358] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 07/19/2020] [Accepted: 08/05/2020] [Indexed: 12/18/2022]
Abstract
Aim This study aimed to characterize the salivary proteome during the induction and resolution of gingival inflammation in the course of human experimental gingivitis (EG), and to cluster the proteomic profiles based on the clinically defined “slow” and “fast” response patterns. Materials and Methods A total of 50 unstimulated whole saliva were obtained from the EG model which was induced over 21 days (days 0, 7, 14 and 21), followed by a two‐week resolution phase (day 35). Label‐free quantitative proteomics using liquid chromatography–tandem mass spectrometry was applied. Regulated proteins were subject to Gene Ontology enrichment analysis. Results A total of 804 human proteins were quantified by ≥ 2 peptides. Principal component analysis depicted significant differences between “fast” and “slow” responders. Despite gingival and plaque scores being similar at baseline among the two groups, “fast” responders presented with 48 proteins that were at > 4‐fold higher levels than “slow” responders. These up‐regulated proteins showed enrichment in “antigen presentation” and “proteolysis.” Conclusions Together, these findings highlight the utility of integrative systems‐level quantitative proteomic approaches to unravel the molecular basis of “salivary proteotypes” associated with gingivitis dubbed as “fast” and “slow” responders. Hence, these differential responses may help prognosticate individual susceptibility to gingival inflammation.
Collapse
Affiliation(s)
- Nagihan Bostanci
- Section of Periodontology and Dental Prevention, Division of Oral Diseases, Department of Dental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Angelika Silbereisen
- Section of Periodontology and Dental Prevention, Division of Oral Diseases, Department of Dental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Kai Bao
- Section of Periodontology and Dental Prevention, Division of Oral Diseases, Department of Dental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Jonas Grossmann
- Functional Genomic Center Zurich, ETH Zurich and University of Zurich, Zurich, Switzerland
| | - Paolo Nanni
- Functional Genomic Center Zurich, ETH Zurich and University of Zurich, Zurich, Switzerland
| | - Claudia Fernandez
- Functional Genomic Center Zurich, ETH Zurich and University of Zurich, Zurich, Switzerland
| | - Gustavo G Nascimento
- Section of Periodontology, Department of Dentistry and Oral Health, Aarhus University, Aarhus, Denmark
| | - Georgios N Belibasakis
- Section of Periodontology and Dental Prevention, Division of Oral Diseases, Department of Dental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Rodrigo Lopez
- Section of Periodontology, Department of Dentistry and Oral Health, Aarhus University, Aarhus, Denmark
| |
Collapse
|
23
|
Rizal MI, Soeroso Y, Sulijaya B, Assiddiq BF, Bachtiar EW, Bachtiar BM. Proteomics approach for biomarkers and diagnosis of periodontitis: systematic review. Heliyon 2020; 6:e04022. [PMID: 32529063 PMCID: PMC7276445 DOI: 10.1016/j.heliyon.2020.e04022] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 04/12/2020] [Accepted: 05/15/2020] [Indexed: 01/03/2023] Open
Abstract
Quantitative proteomic workflow based on mass spectrometry (MS) is recently developed by the researchers to screen for biomarkers in periodontal diseases comprising periodontitis. Periodontitis is known for chronic inflammatory disease characterized by progressive destruction of the tooth-supporting apparatus, yet has a lack of clear pathobiology based on a discrepancy between specified categories and diagnostic vagueness. The objective of this review was to outlined the accessible information related to proteomics studies on periodontitis. The Preferred Reporting Items for Systematical Reviews and Meta-Analysis (PRISMA) statement guides to acquaint proteomic analysis on periodontal diseases was applied. Three databases were used in this study, such as Pubmed, ScienceDirect and Biomed Central from 2009 up to November 2019. Proteomics analysis platforms that used in the studies were outlined. Upregulated and downregulated proteins findings data were found, in which could be suitable as candidate biomarkers for this disease.
Collapse
Affiliation(s)
- Muhammad Ihsan Rizal
- Oral Science Research Center, Faculty of Dentistry, Universitas Indonesia, Jakarta, Indonesia
| | - Yuniarti Soeroso
- Department of Periodontology, Faculty of Dentistry, Universitas Indonesia, Jakarta, Indonesia
| | - Benso Sulijaya
- Department of Periodontology, Faculty of Dentistry, Universitas Indonesia, Jakarta, Indonesia
| | | | - Endang W. Bachtiar
- Oral Science Research Center, Faculty of Dentistry, Universitas Indonesia, Jakarta, Indonesia
- Department of Oral Biology, Faculty of Dentistry, Universitas Indonesia, Jakarta, Indonesia
| | - Boy M. Bachtiar
- Oral Science Research Center, Faculty of Dentistry, Universitas Indonesia, Jakarta, Indonesia
- Department of Oral Biology, Faculty of Dentistry, Universitas Indonesia, Jakarta, Indonesia
| |
Collapse
|
24
|
Bikker FJ, Nascimento GG, Nazmi K, Silbereisen A, Belibasakis GN, Kaman WE, Lopez R, Bostanci N. Salivary Total Protease Activity Based on a Broad-Spectrum Fluorescence Resonance Energy Transfer Approach to Monitor Induction and Resolution of Gingival Inflammation. Mol Diagn Ther 2020; 23:667-676. [PMID: 31372941 PMCID: PMC6775538 DOI: 10.1007/s40291-019-00421-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
OBJECTIVE Salivary total protease and chitinase activities were measured by a broad-spectrum fluorescence resonance energy transfer approach as predictors of induction and resolution of gingival inflammation in healthy individuals by applying an experimental human gingivitis model. METHODS Dental biofilm accumulated (21 days, Induction Phase) by omitting oral hygiene practices followed by a 2-week Resolution Phase to restore gingival health in an experimental gingivitis study. Plaque accumulation, as assessed by the Turesky Modification of the Quigley-Hein Plaque Index (TQHPI), and gingival inflammation, assessed using the Modified Gingival Index (MGI), scores were recorded and unstimulated saliva was collected weekly. Saliva was analysed for total protein, albumin, total protease activity and chitinase activity (n = 18). RESULTS The TQHPI and MGI scores, as well as total protease activity, increased until day 21. After re-establishment of oral hygiene, gingival inflammation levels returned to values similar to baseline (day 0). Levels of protease activity decreased significantly, but not to baseline values. Furthermore, 'fast' responders, who responded immediately to plaque, exhibited significantly higher proteolytic activity throughout the experimental course than 'slow' responders, who showed a lagged inflammatory response. CONCLUSION The results indicate that differential inflammatory responses encompass inherent variations in total salivary proteolytic activities, which could be further utilised in contemporary diagnostic, prognostic and treatment modalities for periodontal diseases.
Collapse
Affiliation(s)
- Floris J Bikker
- Department of Oral Biochemistry, Academic Centre for Dentistry Amsterdam (ACTA), Free University of Amsterdam and University of Amsterdam, Amsterdam, The Netherlands.
| | - Gustavo G Nascimento
- Section of Periodontology, Department of Dentistry and Oral Health, Aarhus University, Aarhus, Denmark
| | - Kamran Nazmi
- Department of Oral Biochemistry, Academic Centre for Dentistry Amsterdam (ACTA), Free University of Amsterdam and University of Amsterdam, Amsterdam, The Netherlands
| | - Angelika Silbereisen
- Section of Periodontology and Dental Prevention, Division of Oral Diseases, Department of Dental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Georgios N Belibasakis
- Section of Periodontology and Dental Prevention, Division of Oral Diseases, Department of Dental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Wendy E Kaman
- Department of Oral Biochemistry, Academic Centre for Dentistry Amsterdam (ACTA), Free University of Amsterdam and University of Amsterdam, Amsterdam, The Netherlands.,Department of Medical Microbiology and Infectious Diseases, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Rodrigo Lopez
- Section of Periodontology, Department of Dentistry and Oral Health, Aarhus University, Aarhus, Denmark
| | - Nagihan Bostanci
- Section of Periodontology and Dental Prevention, Division of Oral Diseases, Department of Dental Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
25
|
Nguyen T, Sedghi L, Ganther S, Malone E, Kamarajan P, Kapila YL. Host-microbe interactions: Profiles in the transcriptome, the proteome, and the metabolome. Periodontol 2000 2020; 82:115-128. [PMID: 31850641 DOI: 10.1111/prd.12316] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Periodontal studies using transcriptomics, proteomics, and metabolomics encompass the collection of mRNA transcripts, proteins, and small-molecule chemicals in the context of periodontal health and disease. The number of studies using these approaches has significantly increased in the last decade and they have provided new insight into the pathogenesis and host-microbe interactions that define periodontal diseases. This review provides an overview of current molecular findings using -omic approaches that underlie periodontal disease, including modulation of the host immune response, tissue homeostasis, and complex metabolic processes of the host and the oral microbiome. Integration of these -omic approaches will broaden our perspective of the molecular mechanisms involved in periodontal disease, advancing and improving the diagnosis and treatment of various stages and forms of periodontal disease.
Collapse
Affiliation(s)
- Trang Nguyen
- School of Dentistry, University of California San Francisco, San Francisco, California, USA
| | - Lea Sedghi
- Department of Orofacial Sciences, School of Dentistry, University of California San Francisco, San Francisco, California, USA
| | - Sean Ganther
- Department of Orofacial Sciences, School of Dentistry, University of California San Francisco, San Francisco, California, USA
| | - Erin Malone
- Department of Orofacial Sciences, School of Dentistry, University of California San Francisco, San Francisco, California, USA
| | - Pachiyappan Kamarajan
- Department of Orofacial Sciences, School of Dentistry, University of California San Francisco, San Francisco, California, USA
| | - Yvonne L Kapila
- Department of Orofacial Sciences, School of Dentistry, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
26
|
A salivary metabolite signature that reflects gingival host-microbe interactions: instability predicts gingivitis susceptibility. Sci Rep 2020; 10:3008. [PMID: 32080300 PMCID: PMC7033112 DOI: 10.1038/s41598-020-59988-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Accepted: 02/04/2020] [Indexed: 11/08/2022] Open
Abstract
Several proteins and peptides in saliva were shown to stimulate gingival wound repair, but the role of salivary metabolites in this process remains unexplored. In vitro gingival re-epithelialization kinetics were determined using unstimulated saliva samples from healthy individuals collected during an experimental gingivitis study. Elastic net regression with stability selection identified a specific metabolite signature in a training dataset that was associated with the observed re-epithelialization kinetics and enabled its prediction for all saliva samples obtained in the clinical study. This signature encompassed ten metabolites, including plasmalogens, diacylglycerol and amino acid derivatives, which reflect enhanced host-microbe interactions. This association is in agreement with the positive correlation of the metabolite signature with the individual’s gingival bleeding index. Remarkably, intra-individual signature-variation over time was associated with elevated risk for gingivitis development. Unravelling how these metabolites stimulate wound repair could provide novel avenues towards therapeutic approaches in patients with impaired wound healing capacity.
Collapse
|
27
|
Brown JL, Johnston W, Delaney C, Rajendran R, Butcher J, Khan S, Bradshaw D, Ramage G, Culshaw S. Biofilm-stimulated epithelium modulates the inflammatory responses in co-cultured immune cells. Sci Rep 2019; 9:15779. [PMID: 31673005 PMCID: PMC6823452 DOI: 10.1038/s41598-019-52115-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 10/14/2019] [Indexed: 12/13/2022] Open
Abstract
The gingival epithelium is a physical and immunological barrier to the microbiota of the oral cavity, which interact through soluble mediators with the immune cells that patrol the tissue at the gingival epithelium. We sought to develop a three-dimensional gingivae-biofilm interface model using a commercially available gingival epithelium to study the tissue inflammatory response to oral biofilms associated with “health”, “gingivitis” and “periodontitis”. These biofilms were developed by sequential addition of microorganisms to mimic the formation of supra- and sub-gingival plaque in vivo. Secondly, to mimic the interactions between gingival epithelium and immune cells in vivo, we integrated peripheral blood mononuclear cells and CD14+ monocytes into our three-dimensional model and were able to assess the inflammatory response in the immune cells cultured with and without gingival epithelium. We describe a differential inflammatory response in immune cells cultured with epithelial tissue, and more so following incubation with epithelium stimulated by “gingivitis-associated” biofilm. These results suggest that gingival epithelium-derived soluble mediators may control the inflammatory status of immune cells in vitro, and therefore targeting of the epithelial response may offer novel therapies. This multi-cellular interface model, both of microbial and host origin, offers a robust in vitro platform to investigate host-pathogens at the epithelial surface.
Collapse
Affiliation(s)
- Jason L Brown
- Institute of Biomedical and Environmental Health Research, School of Science and Sport, University of the West of Scotland, Paisley, PA1 2BE, UK.,Oral Sciences Research Group, Glasgow Dental School, School of Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8TA, UK
| | - William Johnston
- Oral Sciences Research Group, Glasgow Dental School, School of Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8TA, UK
| | - Chris Delaney
- Institute of Biomedical and Environmental Health Research, School of Science and Sport, University of the West of Scotland, Paisley, PA1 2BE, UK
| | - Ranjith Rajendran
- Oral Sciences Research Group, Glasgow Dental School, School of Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8TA, UK
| | - John Butcher
- Institute of Biomedical and Environmental Health Research, School of Science and Sport, University of the West of Scotland, Paisley, PA1 2BE, UK.,Department of Life Sciences, School of Health and Life Sciences, Glasgow Caledonian University, Glasgow, G4 0BA, UK
| | - Shaz Khan
- Oral Healthcare R&D, GlaxoSmithKline Consumer Healthcare, Weybridge, KT13 0DE, UK
| | - David Bradshaw
- Oral Healthcare R&D, GlaxoSmithKline Consumer Healthcare, Weybridge, KT13 0DE, UK
| | - Gordon Ramage
- Oral Sciences Research Group, Glasgow Dental School, School of Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8TA, UK.
| | - Shauna Culshaw
- Oral Sciences Research Group, Glasgow Dental School, School of Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8TA, UK.
| |
Collapse
|
28
|
Bai F, Cai Z, Yang L. Recent progress in experimental and human disease-associated multi-species biofilms. Comput Struct Biotechnol J 2019; 17:1234-1244. [PMID: 31921390 PMCID: PMC6944735 DOI: 10.1016/j.csbj.2019.09.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 09/18/2019] [Accepted: 09/21/2019] [Indexed: 12/16/2022] Open
Abstract
Human bodies are colonized by trillions of microorganisms, which are often referred to as human microbiota and play important roles in human health. Next generation sequencing studies have established links between the genetic content of human microbiota and various human diseases. However, it remains largely unknown about the spatial organizations and interspecies interactions of individual species within the human microbiota. Bacterial cells tend to form surface-attached biofilms in many natural environments, which enable intercellular communications and interactions in a microbial ecosystem. In this review, we summarize the recent progresses on the experimental and human disease-associated multi-species biofilm studies. We hypothesize that engineering biofilm structures and interspecies interactions might provide a tool for manipulating the composition and function of human microbiota.
Collapse
Affiliation(s)
- Fang Bai
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China
| | - Zhao Cai
- Singapore Centre for Environmental Life Sciences Engineering (SCELSE), Nanyang Technology University, Singapore
| | - Liang Yang
- School of Medicine, Southern University of Science and Technology (SUSTech), Shenzhen, Guangdong, China
| |
Collapse
|
29
|
Binti Badlishah Sham NI, Lewin SD, Grant MM. Proteomic Investigations of In Vitro and In Vivo Models of Periodontal Disease. Proteomics Clin Appl 2019; 14:e1900043. [PMID: 31419032 DOI: 10.1002/prca.201900043] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 07/29/2019] [Indexed: 12/14/2022]
Abstract
Proteomics has currently been a developing field in periodontal diseases to obtain protein information of certain samples. Periodontal disease is an inflammatory disorder that attacks the teeth, connective tissues, and alveolar bone within the oral cavity. Proteomics information can provide proteins that are differentially expressed in diseased or healthy samples. This review provides insight into approaches researching single species, multi species, bacteria, non-human, and human models of periodontal disease for proteomics information. The approaches that have been taken include gel electrophoresis and qualitative and quantitative mass spectrometry. This review is carried out by extracting information about in vitro and in vivo studies of proteomics in models of periodontal diseases that have been carried out in the past two decades. The research has concentrated on a relatively small but well-known group of microorganisms. A wide range of models has been reviewed and conclusions across the breadth of these studies are presented in this review.
Collapse
Affiliation(s)
- Nurul Iman Binti Badlishah Sham
- School of Dentistry, Institute of Clinical Sciences, University of Birmingham, 5 Mill Pool Way, Edgbaston, Birmingham, B5 7EG, UK.,Faculty of Dentistry , Universiti Sains Islam Malaysia, 55100, Kuala Lumpur, Malaysia
| | - Sean D Lewin
- School of Dentistry, Institute of Clinical Sciences, University of Birmingham, 5 Mill Pool Way, Edgbaston, Birmingham, B5 7EG, UK
| | - Melissa M Grant
- School of Dentistry, Institute of Clinical Sciences, University of Birmingham, 5 Mill Pool Way, Edgbaston, Birmingham, B5 7EG, UK
| |
Collapse
|
30
|
Bostanci N, Bao K, Greenwood D, Silbereisen A, Belibasakis GN. Periodontal disease: From the lenses of light microscopy to the specs of proteomics and next-generation sequencing. Adv Clin Chem 2019; 93:263-290. [PMID: 31655732 DOI: 10.1016/bs.acc.2019.07.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Periodontal disease entails the inflammatory destruction of the tooth supporting (periodontal) tissues as a result of polymicrobial colonization of the tooth surface in the form of biofilms. Extensive data collected over the past decades on this chronic disease demonstrate that its progression is infrequent and episodic, and the susceptibility to it can vary among individuals. Physical assessments of previously occurring damage to periodontal tissues remain the cornerstone of detection and diagnosis, whereas traditionally used diagnostic procedures do neither identify susceptible individuals nor distinguish between disease-active and disease-inactive periodontal sites. Thus, more sensitive and accurate "measurable biological indicators" of periodontal diseases are needed in order to place diagnosis (e.g., the presence or stage) and management of the disease on a more rational less empirical basis. Contemporary "omics" technologies may help unlock the path to this quest. High throughput nucleic acid sequencing technologies have enabled us to examine the taxonomic distribution of microbial communities in oral health and disease, whereas proteomic technologies allowed us to decipher the molecular state of the host in disease, as well as the interactive cross-talk of the host with the microbiome. The newly established field of metaproteomics has enabled the identification of the repertoire of proteins that oral microorganisms use to compete or co-operate with each other. Vast such data is derived from oral biological fluids, including gingival crevicular fluid and saliva, which is progressively completed and catalogued as the analytical technologies and bioinformatics tools progressively advance. This chapter covers the current "omics"-derived knowledge on the microbiome, the host and their "interactome" with regard to periodontal diseases, and addresses challenges and opportunities ahead.
Collapse
Affiliation(s)
- Nagihan Bostanci
- Section of Periodontology and Dental Prevention, Division of Oral Diseases, Department of Dental Medicine, Karolinska Institutet, Stockholm, Sweden.
| | - Kai Bao
- Section of Periodontology and Dental Prevention, Division of Oral Diseases, Department of Dental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - David Greenwood
- Section of Periodontology and Dental Prevention, Division of Oral Diseases, Department of Dental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Angelika Silbereisen
- Section of Periodontology and Dental Prevention, Division of Oral Diseases, Department of Dental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Georgios N Belibasakis
- Section of Periodontology and Dental Prevention, Division of Oral Diseases, Department of Dental Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
31
|
Carvalho-Filho PC, Moura-Costa LF, Pimentel ACM, Lopes MPP, Freitas SA, Miranda PM, Costa RS, Figueirêdo CAV, Meyer R, Gomes-Filho IS, Olczak T, Xavier MT, Trindade SC. Apoptosis Transcriptional Profile Induced by Porphyromonas gingivalis HmuY. Mediators Inflamm 2019; 2019:6758159. [PMID: 31011284 PMCID: PMC6442302 DOI: 10.1155/2019/6758159] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 12/27/2018] [Accepted: 01/23/2019] [Indexed: 11/30/2022] Open
Abstract
This study aimed at evaluating the transcriptional profile of apoptosis-related genes after in vitro stimulation of peripheral blood mononuclear cells (PBMCs) derived from individuals with periodontitis (P) and healthy nonperiodontitis (NP) control subjects with P. gingivalis HmuY protein. PBMCs from the P and NP groups were stimulated with HmuY P. gingivalis protein, and the expression of genes related to apoptosis was assessed by custom real-time polymerase chain reaction array (Custom RT2 PCR Array). Compared with the NP group, the P group showed low relative levels of apoptosis-related gene expression, downregulated for FAS, FAS ligand, TNFSF10 (TRAIL), BAK1, CASP9, and APAF1 after P. gingivalis HmuY protein stimulation. Furthermore, the P group exhibited low levels of relative gene expression, downregulated for CASP7 when the cells were not stimulated. Our data suggest that P. gingivalis HmuY protein might participate differently in the modulation of the intrinsic and extrinsic apoptosis pathways.
Collapse
Affiliation(s)
- Paulo C. Carvalho-Filho
- Department of Immunology, Federal University of Bahia, Bahia, Brazil
- Dental School, Bahiana School of Medicine and Public Health, Brazil
| | | | | | - Mabel P. P. Lopes
- Department of Immunology, Federal University of Bahia, Bahia, Brazil
| | | | | | - Ryan S. Costa
- Department of Immunology, Federal University of Bahia, Bahia, Brazil
| | | | - Roberto Meyer
- Department of Immunology, Federal University of Bahia, Bahia, Brazil
| | | | - Teresa Olczak
- Laboratory of Medical Biology, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
| | - Márcia T. Xavier
- Dental School, Bahiana School of Medicine and Public Health, Brazil
| | - Soraya C. Trindade
- Department of Immunology, Federal University of Bahia, Bahia, Brazil
- Department of Periodontics, Feira de Santana State University, Bahia, Brazil
| |
Collapse
|
32
|
Groeger S, Meyle J. Oral Mucosal Epithelial Cells. Front Immunol 2019; 10:208. [PMID: 30837987 PMCID: PMC6383680 DOI: 10.3389/fimmu.2019.00208] [Citation(s) in RCA: 239] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 01/23/2019] [Indexed: 12/14/2022] Open
Abstract
Cellular Phenotype and Apoptosis: The function of epithelial tissues is the protection of the organism from chemical, microbial, and physical challenges which is indispensable for viability. To fulfill this task, oral epithelial cells follow a strongly regulated scheme of differentiation that results in the formation of structural proteins that manage the integrity of epithelial tissues and operate as a barrier. Oral epithelial cells are connected by various transmembrane proteins with specialized structures and functions. Keratin filaments adhere to the plasma membrane by desmosomes building a three-dimensional matrix. Cell-Cell Contacts and Bacterial Influence: It is known that pathogenic oral bacteria are able to affect the expression and configuration of cell-cell junctions. Human keratinocytes up-regulate immune-modulatory receptors upon stimulation with bacterial components. Periodontal pathogens including P. gingivalis are able to inhibit oral epithelial innate immune responses through various mechanisms and to escape from host immune reaction, which supports the persistence of periodontitis and furthermore is able to affect the epithelial barrier function by altering expression and distribution of cell-cell interactions including tight junctions (TJs) and adherens junctions (AJs). In the pathogenesis of periodontitis a highly organized biofilm community shifts from symbiosis to dysbiosis which results in destructive local inflammatory reactions. Cellular Receptors: Cell-surface located toll like receptors (TLRs) and cytoplasmatic nucleotide-binding oligomerization domain (NOD)-like receptors (NLRs) belong to the pattern recognition receptors (PRRs). PRRs recognize microbial parts that represent pathogen-associated molecular patterns (PAMPs). A multimeric complex of proteins known as inflammasome, which is a subset of NLRs, assembles after activation and proceeds to pro-inflammatory cytokine release. Cytokine Production and Release: Cytokines and bacterial products may lead to host cell mediated tissue destruction. Keratinocytes are able to produce diverse pro-inflammatory cytokines and chemokines, including interleukin (IL)-1, IL-6, IL-8 and tumor necrosis factor (TNF)-α. Infection by pathogenic bacteria such as Porphyromonas gingivalis (P. gingivalis) and Aggregatibacter actinomycetemcomitans (A. actinomycetemcomitans) can induce a differentiated production of these cytokines. Immuno-modulation, Bacterial Infection, and Cancer Cells: There is a known association between bacterial infection and cancer. Bacterial components are able to up-regulate immune-modulatory receptors on cancer cells. Interactions of bacteria with tumor cells could support malignant transformation an environment with deficient immune regulation. The aim of this review is to present a set of molecular mechanisms of oral epithelial cells and their reactions to a number of toxic influences.
Collapse
Affiliation(s)
- Sabine Groeger
- Department of Periodontology, Justus-Liebig-University of Giessen, Giessen, Germany
| | - Joerg Meyle
- Department of Periodontology, Justus-Liebig-University of Giessen, Giessen, Germany
| |
Collapse
|
33
|
Bostanci N, Selevsek N, Wolski W, Grossmann J, Bao K, Wahlander A, Trachsel C, Schlapbach R, Öztürk VÖ, Afacan B, Emingil G, Belibasakis GN. Targeted Proteomics Guided by Label-free Quantitative Proteome Analysis in Saliva Reveal Transition Signatures from Health to Periodontal Disease. Mol Cell Proteomics 2018; 17:1392-1409. [PMID: 29610270 DOI: 10.1074/mcp.ra118.000718] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Revised: 03/28/2018] [Indexed: 12/18/2022] Open
Abstract
Periodontal diseases are among the most prevalent worldwide, but largely silent, chronic diseases. They affect the tooth-supporting tissues with multiple ramifications on life quality. Their early diagnosis is still challenging, due to lack of appropriate molecular diagnostic methods. Saliva offers a non-invasively collectable reservoir of clinically relevant biomarkers, which, if utilized efficiently, could facilitate early diagnosis and monitoring of ongoing disease. Despite several novel protein markers being recently enlisted by discovery proteomics, their routine diagnostic application is hampered by the lack of validation platforms that allow for rapid, accurate and simultaneous quantification of multiple proteins in large cohorts. Here we carried out a pipeline of two proteomic platforms; firstly, we applied open ended label-free quantitative (LFQ) proteomics for discovery in saliva (n = 67, including individuals with health, gingivitis, and periodontitis), followed by selected-reaction monitoring (SRM)-targeted proteomics for validation in an independent cohort (n = 82). The LFQ platform led to the discovery of 119 proteins with at least 2-fold significant difference between health and disease. The 65 proteins chosen for the subsequent SRM platform included 50 functionally related proteins derived from the significantly enriched processes of the LFQ data, 11 from literature-mining, and four house-keeping ones. Among those, 60 were reproducibly quantifiable proteins (92% success rate), represented by a total of 143 peptides. Machine-learning modeling led to a narrowed-down panel of five proteins of high predictive value for periodontal diseases with maximum area under the receiver operating curve >0.97 (higher in disease: Matrix metalloproteinase-9, Ras-related protein-1, Actin-related protein 2/3 complex subunit 5; lower in disease: Clusterin, Deleted in Malignant Brain Tumors 1). This panel enriches the pool of credible clinical biomarker candidates for diagnostic assay development. Yet, the quantum leap brought into the field of periodontal diagnostics by this study is the application of the biomarker discovery-through-verification pipeline, which can be used for validation in further cohorts.
Collapse
Affiliation(s)
- Nagihan Bostanci
- From the ‡Division of Oral Diseases, Department of Dental Medicine, Karolinska Institutet, Stockholm, Sweden;
| | - Nathalie Selevsek
- §Functional Genomics Center Zürich, University of Zürich/ETH Zürich, Zürich, Switzerland
| | - Witold Wolski
- §Functional Genomics Center Zürich, University of Zürich/ETH Zürich, Zürich, Switzerland
| | - Jonas Grossmann
- §Functional Genomics Center Zürich, University of Zürich/ETH Zürich, Zürich, Switzerland
| | - Kai Bao
- From the ‡Division of Oral Diseases, Department of Dental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Asa Wahlander
- ¶AstraZeneca Translational Biomarkers and Bioanalysis, Drug Safety and Metabolism, Innovative Medicines, Mölndal, Sweden
| | - Christian Trachsel
- §Functional Genomics Center Zürich, University of Zürich/ETH Zürich, Zürich, Switzerland
| | - Ralph Schlapbach
- §Functional Genomics Center Zürich, University of Zürich/ETH Zürich, Zürich, Switzerland
| | - Veli Özgen Öztürk
- ‖Department of Periodontology, School of Dentistry, Adnan Menderes University, Aydin, Turkey
| | - Beral Afacan
- ‖Department of Periodontology, School of Dentistry, Adnan Menderes University, Aydin, Turkey
| | - Gulnur Emingil
- **Department of Periodontology, School of Dentistry, Ege University, Izmir, Turkey
| | - Georgios N Belibasakis
- From the ‡Division of Oral Diseases, Department of Dental Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
34
|
Thurnheer T, Belibasakis GN. Streptococcus oralis maintains homeostasis in oral biofilms by antagonizing the cariogenic pathogen Streptococcus mutans. Mol Oral Microbiol 2018; 33:234-239. [PMID: 29327482 DOI: 10.1111/omi.12216] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/08/2018] [Indexed: 12/24/2022]
Abstract
Bacteria residing in oral biofilms live in a state of dynamic equilibrium with one another. The intricate synergistic or antagonistic interactions between them are crucial for determining this balance. Using the six-species Zürich "supragingival" biofilm model, this study aimed to investigate interactions regarding growth and localization of the constituent species. As control, an inoculum containing all six strains was used, whereas in each of the further five inocula one of the bacterial species was alternately absent, and in the last, both streptococci were absent. Biofilms were grown anaerobically on hydroxyapatite disks, and after 64 h they were harvested and quantified by culture analyses. For visualization, fluorescence in situ hybridization and confocal laser scanning microscopy were used. Compared with the control, no statistically significant difference of total colony-forming units was observed in the absence of any of the biofilm species, except for Fusobacterium nucleatum, whose absence caused a significant decrease in total bacterial numbers. Absence of Streptococcus oralis resulted in a significant decrease in Actinomyces oris, and increase in Streptococcus mutans (P < .001). Absence of A. oris, Veillonella dispar or S. mutans did not cause any changes. The structure of the biofilm with regards to the localization of the species did not result in observable changes. In summary, the most striking observation of the present study was that absence of S. oralis resulted in limited growth of commensal A. oris and overgrowth of S. mutans. These data establish highlight S. oralis as commensal keeper of homeostasis in the biofilm by antagonizing S. mutans, so preventing a caries-favoring dysbiotic state.
Collapse
Affiliation(s)
- T Thurnheer
- Clinic of Preventive Dentistry, Periodontology and Cariology, Divison of Oral Microbiology and Immunology, Center of Dental Medicine, University of Zürich, Zürich, Switzerland
| | - G N Belibasakis
- Division of Oral Diseases, Department of Dental Medicine, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
35
|
Guo W, Wang P, Liu ZH, Ye P. Analysis of differential expression of tight junction proteins in cultured oral epithelial cells altered by Porphyromonas gingivalis, Porphyromonas gingivalis lipopolysaccharide, and extracellular adenosine triphosphate. Int J Oral Sci 2018; 10:e8. [PMID: 29319048 PMCID: PMC5795020 DOI: 10.1038/ijos.2017.51] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/31/2017] [Indexed: 12/20/2022] Open
Abstract
Tight junctions (TJs) are the most apical intercellular junctions of epithelial cells formed by occludin, claudins, junctional adhesion molecules (JAMs), and zonula occludens (ZO). Tight junction proteins can sense the presence of bacteria and regulate the transcription of target genes that encode effectors and regulators of the immune response. The aim of this study was to determine the impact of TJ proteins in response to Porphyromonas gingivalis (P. gingivalis), P. gingivalis lipopolysaccharide (P. gingivalis LPS), and extracellular adenosine triphosphate (ATP) in the oral epithelial cell culture model. Quantified real time-polymerase chain reaction (RT-PCR), immunoblots, and immunostaining were performed to assess the gene and protein expression in TJs. It was found that P. gingivalis infection led to transient upregulation of the genes encoding occludin, claudin-1, and claudin-4 but not JAM-A, claudin-15, or ZO-1, while P. gingivalis LPS increased claudin-1, claudin-15, and ZO-1 and decreased occludin, JAM-A, and claudin-4. Tight junction proteins showed significant upregulation in the above two groups when cells were pretreated with ATP for 3 h. The findings indicated that P. gingivalis induced the host defence responses at an early stage. P. gingivalis LPS exerted a more powerful stimulatory effect on the disruption of the epithelial barrier than P. gingivalis. ATP stimulation enhanced the reaction of TJ proteins to P. gingivalis invasion and LPS destruction of the epithelium.
Collapse
Affiliation(s)
- Wei Guo
- Department of Endodontics, Yantai Stomatological Hospital, Binzhou Medical University, Yantai, China
| | - Peng Wang
- Department of Pediatrics, Yantai Stomatological Hospital, Binzhou Medical University, Yantai, China
| | - Zhong-Hao Liu
- Department of Implant, Yantai Stomatological Hospital, Binzhou Medical University, Yantai, China
| | - Ping Ye
- Institute of Dental Research, Centre for Oral Health, Westmead Hospital, Westmead, Australia.,Faculty of Dentistry, the University of Sydney, Sydney, Australia
| |
Collapse
|
36
|
The making of a miscreant: tobacco smoke and the creation of pathogen-rich biofilms. NPJ Biofilms Microbiomes 2017; 3:26. [PMID: 29081982 PMCID: PMC5655325 DOI: 10.1038/s41522-017-0033-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 08/29/2017] [Accepted: 09/25/2017] [Indexed: 02/03/2023] Open
Abstract
We have previously reported that oral biofilms in clinically healthy smokers are pathogen-rich, and that this enrichment occurs within 24 h of biofilm formation. The present investigation aimed to identify a mechanism by which smoking creates this altered community structure. By combining in vitro microbial–mucosal interface models of commensal (consisting of Streptococcus oralis, Streptococcus sanguis, Streptococcus mitis, Actinomyces naeslundii, Neisseria mucosa and Veillonella parvula) and pathogen-rich (comprising S.oralis, S.sanguis, S.mitis, A.naeslundii, N.mucosa and V.parvula, Fusobacterium nucleatum, Porphyromonas gingivalis, Filifactor alocis, Dialister pneumosintes, Selenonomas sputigena, Selenominas noxia, Catonella morbi, Parvimonas micra and Tannerella forsythia) communities with metatranscriptomics, targeted proteomics and fluorescent microscopy, we demonstrate that smoke exposure significantly downregulates essential metabolic functions within commensal biofilms, while significantly increasing expression of virulence genes, notably lipopolysaccharide (LPS), flagella and capsule synthesis. By contrast, in pathogen-rich biofilms several metabolic pathways were over-expressed in response to smoke exposure. Under smoke-rich conditions, epithelial cells mounted an early and amplified pro-inflammatory and oxidative stress response to these virulence-enhanced commensal biofilms, and a muted early response to pathogen-rich biofilms. Commensal biofilms also demonstrated early and widespread cell death. Similar results were observed when smoke-free epithelial cells were challenged with smoke-conditioned biofilms, but not vice versa. In conclusion, our data suggest that smoke-induced transcriptional shifts in commensal biofilms triggers a florid pro-inflammatory response, leading to early commensal death, which may preclude niche saturation by these beneficial organisms. The cytokine-rich, pro-oxidant, anaerobic environment sustains inflammophilic bacteria, and, in the absence of commensal antagonism, may promote the creation of pathogen-rich biofilms in smokers. Tobacco smoke inhibits the metabolism of beneficial bacteria in biofilms, while activating specific genes in pathogenic bacteria. This suggests a mechanism to explain how smoking quickly leads to the formation of damaging biofilms in the mouth and respiratory tract. Purnima Kumar and colleagues at Ohio State University, USA studied the effect of tobacco smoke on cultured biofilms used to model those that form on mucous membranes. They detected specific and varied changes in the activity of genes, proteins and metabolism that allowed pathogenic bacteria to displace beneficial “commensal” bacteria. The research suggests the transition toward pathogen-rich biofilms may contribute to the health effects of smoking by causing increased inflammation of mucous membranes and the production of damaging oxidant chemicals. Further research should investigate the chemical constituents of smoke responsible for these effects.
Collapse
|
37
|
Rudick CP, Miyamoto T, Lang MS, Agrawal DK. Triggering receptor expressed on myeloid cells in the pathogenesis of periodontitis: potential novel treatment strategies. Expert Rev Clin Immunol 2017; 13:1189-1197. [PMID: 29027827 DOI: 10.1080/1744666x.2017.1392855] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
INTRODUCTION Periodontal diseases are polymicrobial inflammatory disorders of the tissue, ligament, and bone structures supporting teeth. Periodontitis (inflammation with corresponding loss of attachment) affects 40-50% of adults. Recently, members of the Triggering Receptor on Myeloid Cell (TREM) family have been studied to determine their relationship to these diseases. Areas covered: TREM-1 is a receptor expressed on the surface of PMNs, monocytes, macrophages, dendritic cells, vascular smooth muscle cells, and keratinocytes upregulated in the presence of periodontal inflammation. TREM-1 expression can be upregulated by oral bacterium Porphyromonas gingivalis that can be abrogated by a sub-antimicrobial dose of doxycycline. When cleaved from the cell surface, a soluble form of TREM-1 (sTREM-1) can be used as a biomarker of inflammation and might also provide a link between oral and systemic inflammation. While less understood, TREM-2 has a role in osteoclastogenesis which could contribute to the alveolar bone destruction seen in more advanced periodontitis. Expert commentary: Additional studies to simulate biofilm microenvironment in TREM research are warranted. Longitudinal studies determining TREM-1, sTREM-1, and TREM-2 levels in tissues over time and progression of periodontal diseases would provide valuable information in the role of TREM receptors as indicators of or contributors to the disease process.
Collapse
Affiliation(s)
- Courtney P Rudick
- a Department of Clinical & Translational Science , Creighton University School of Medicine , Omaha , NE , USA
| | - Takanari Miyamoto
- b Department of Periodontology , Creighton University School of Medicine , Omaha , NE , USA
| | - Melissa S Lang
- b Department of Periodontology , Creighton University School of Medicine , Omaha , NE , USA
| | - Devendra K Agrawal
- a Department of Clinical & Translational Science , Creighton University School of Medicine , Omaha , NE , USA
| |
Collapse
|
38
|
Buskermolen JK, Janus MM, Roffel S, Krom BP, Gibbs S. Saliva-Derived Commensal and Pathogenic Biofilms in a Human Gingiva Model. J Dent Res 2017; 97:201-208. [PMID: 28892653 PMCID: PMC6429568 DOI: 10.1177/0022034517729998] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
In vitro models that closely mimic human host-microbiome interactions can be a
powerful screening tool for antimicrobials and will hold great potential for
drug validation and discovery. The aim of this study was to develop an
organotypic oral mucosa model that could be exposed to in vitro cultured
commensal and pathogenic biofilms in a standardized and scalable manner. The
oral mucosa model consisted of a tissue-engineered human gingiva equivalent
containing a multilayered differentiated gingiva epithelium (keratinocytes)
grown on a collagen hydrogel, containing gingiva fibroblasts, which represented
the lamina propria. Keratinocyte and fibroblast telomerase reverse
transcriptase–immortalized cell lines were used to overcome the limitations of
isolating cells from small biopsies when scalable culture experiments were
required. The oral biofilms were grown under defined conditions from human
saliva to represent 3 distinct phenotypes: commensal, gingivitis, and
cariogenic. The in vitro grown biofilms contained physiologic numbers of
bacterial species, averaging >70 operational taxonomic units, including 20
differentiating operational taxonomic units. When the biofilms were applied
topically to the gingiva equivalents for 24 h, the gingiva epithelium increased
its expression of elafin, a protease inhibitor and antimicrobial protein. This
increased elafin expression was observed as a response to all 3 biofilm types,
commensal as well as pathogenic (gingivitis and cariogenic). Biofilm exposure
also increased secretion of the antimicrobial cytokine CCL20 and inflammatory
cytokines IL-6, CXCL8, and CCL2 from gingiva equivalents. This inflammatory
response was far greater after commensal biofilm exposure than after pathogenic
biofilm exposure. These results show that pathogenic oral biofilms have early
immune evasion properties as compared with commensal oral biofilms. The novel
host-microbiome model provides an ideal tool for future investigations of
gingiva responses to commensal and pathogenic biofilms and for testing novel
therapeutics.
Collapse
Affiliation(s)
- J K Buskermolen
- 1 Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - M M Janus
- 2 Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - S Roffel
- 1 Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - B P Krom
- 2 Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - S Gibbs
- 1 Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.,3 Department of Dermatology, VU University Medical Centre, Amsterdam, The Netherlands
| |
Collapse
|
39
|
Bostanci N, Bao K. Contribution of proteomics to our understanding of periodontal inflammation. Proteomics 2017; 17. [DOI: 10.1002/pmic.201500518] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 11/15/2016] [Accepted: 12/15/2016] [Indexed: 12/27/2022]
Affiliation(s)
- Nagihan Bostanci
- Department of Dental Medicine; Karolinska Institute; Huddinge Sweden
| | - Kai Bao
- Division of Oral Microbiology and Immunology; Institute of Oral Biology; Center of Dental Medicine; University of Zürich; Zürich Switzerland
| |
Collapse
|
40
|
Belibasakis GN, Kast JI, Thurnheer T, Akdis CA, Bostanci N. The expression of gingival epithelial junctions in response to subgingival biofilms. Virulence 2016; 6:704-9. [PMID: 26305580 PMCID: PMC4720238 DOI: 10.1080/21505594.2015.1081731] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Periodontitis is an infectious inflammatory disease that destroys the tooth-supporting tissues. It is caused by the formation of subgingival biofilms on the surface of the tooth. Characteristic bacteria associated with subgingival biofilms are the Gram-negative anaerobes Porphyromonas gingivalis, Tannerella forsythia and Treponema denticola, collectively known as the “red complex” species. Inter-epithelial junctions ensure the barrier integrity of the gingival epithelium. This may however be disrupted by the biofilm challenge. The aim of this in vitro study was to investigate the effect of subgingival biofilms on the expression of inter-epithelial junctions by gingival epithelia, and evaluate the relative role of the red complex. Multi-layered human gingival epithelial cultures were challenged with a 10-species in vitro subgingival biofilm model, or its variant without the red complex, for 3 h and 24 h. A low-density array microfluidic card platform was then used for analyzing the expression of 62 genes encoding for tight junctions, gap junctions, adherens junctions, and desmosomes. Although there was a limited effect of the biofilms on the expression of tight, adherens and gap junctions, the expression of a number of desmosomal components was affected. In particular, Desmoglein-1 displayed a limited and transient up-regulation in response to the biofilm. In contrast, Desmocollin-2, Desmoplakin and Plakoglobin were down-regulated equally by both biofilm variants, after 24 h. In conclusion, this subgingival biofilm model may down-regulate selected desmosomal junctions in the gingival epithelium, irrespective of the presence of the “red complex.” In turn, this could compromise the structural integrity of the gingival tissue, favoring bacterial invasion and chronic infection.
Collapse
Affiliation(s)
- Georgios N Belibasakis
- a Oral Microbiology and Immunology; Institute of Oral Biology; Center of Dental Medicine; University of Zürich ; Zürich , Switzerland
| | - Jeannette I Kast
- b Swiss Institute of Allergy and Asthma Research (SIAF); University of Zürich ; Davos , Switzerland
| | - Thomas Thurnheer
- a Oral Microbiology and Immunology; Institute of Oral Biology; Center of Dental Medicine; University of Zürich ; Zürich , Switzerland
| | - Cezmi A Akdis
- b Swiss Institute of Allergy and Asthma Research (SIAF); University of Zürich ; Davos , Switzerland
| | - Nagihan Bostanci
- c Oral Translational Research; Institute of Oral Biology; Center of Dental Medicine; University of Zürich ; Zürich , Switzerland
| |
Collapse
|
41
|
Watters C, Fleming D, Bishop D, Rumbaugh KP. Host Responses to Biofilm. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2016; 142:193-239. [PMID: 27571696 DOI: 10.1016/bs.pmbts.2016.05.007] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
From birth to death the human host immune system interacts with bacterial cells. Biofilms are communities of microbes embedded in matrices composed of extracellular polymeric substance (EPS), and have been implicated in both the healthy microbiome and disease states. The immune system recognizes many different bacterial patterns, molecules, and antigens, but these components can be camouflaged in the biofilm mode of growth. Instead, immune cells come into contact with components of the EPS matrix, a diverse, hydrated mixture of extracellular DNA (bacterial and host), proteins, polysaccharides, and lipids. As bacterial cells transition from planktonic to biofilm-associated they produce small molecules, which can increase inflammation, induce cell death, and even cause necrosis. To survive, invading bacteria must overcome the epithelial barrier, host microbiome, complement, and a variety of leukocytes. If bacteria can evade these initial cell populations they have an increased chance at surviving and causing ongoing disease in the host. Planktonic cells are readily cleared, but biofilms reduce the effectiveness of both polymorphonuclear neutrophils and macrophages. In addition, in the presence of these cells, biofilm formation is actively enhanced, and components of host immune cells are assimilated into the EPS matrix. While pathogenic biofilms contribute to states of chronic inflammation, probiotic Lactobacillus biofilms cause a negligible immune response and, in states of inflammation, exhibit robust antiinflammatory properties. These probiotic biofilms colonize and protect the gut and vagina, and have been implicated in improved healing of damaged skin. Overall, biofilms stimulate a unique immune response that we are only beginning to understand.
Collapse
Affiliation(s)
- C Watters
- Wound Infections Department, Naval Medical Research Center, Silver Spring, MD, United States
| | - D Fleming
- Department of Surgery, Texas Tech University Health Sciences Center, Lubbock, TX, United States; Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - D Bishop
- Wound Infections Department, Naval Medical Research Center, Silver Spring, MD, United States
| | - K P Rumbaugh
- Department of Surgery, Texas Tech University Health Sciences Center, Lubbock, TX, United States; Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX, United States.
| |
Collapse
|
42
|
Proteomic profiling of host-biofilm interactions in an oral infection model resembling the periodontal pocket. Sci Rep 2015; 5:15999. [PMID: 26525412 PMCID: PMC4630604 DOI: 10.1038/srep15999] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 10/07/2015] [Indexed: 12/28/2022] Open
Abstract
Periodontal infections cause inflammatory destruction of the tooth supporting tissues. We recently developed a dynamic, in vitro periodontal organotypic tissue model in a perfusion bioreactor system, in co-culture with an 11-species subgingival biofilm, which may recapitulate early events during the establishment of periodontal infections. This study aimed to characterize the global proteome regulations in this host-biofilm interaction model. Semi-quantitative shotgun proteomics were applied for protein identification and quantification in the co-culture supernatants (human and bacterial) and the biofilm lysates (bacterial). A total of 896 and 3363 proteins were identified as secreted in the supernatant and expressed in the biofilm lysate, respectively. Enriched gene ontology analysis revealed that the regulated secreted human tissue proteins were related to processes of cytoskeletal rearrangement, stress responses, apoptosis, and antigen presentation, all of which are commensurate with deregulated host responses. Most secreted bacterial biofilm proteins derived from their cytoplasmic domain. In the presence of the tissue, the levels of Fusobacterium nucleatum, Actinomyces oris and Campylobacter rectus proteins were significantly regulated. The functions of the up-regulated intracellular (biofilm lysate) proteins were associated with cytokinesis. In conclusion, the proteomic overview of regulated pathways in this host-biofilm interaction model provides insights to the early events of periodontal pathogenesis.
Collapse
|