1
|
Adelfio M, Callen GE, Diaz AR, Paster BJ, He X, Hasturk H, Ghezzi CE. Underscoring long-term host-microbiome interactions in a physiologically relevant gingival tissue model. NPJ Biofilms Microbiomes 2025; 11:9. [PMID: 39789014 PMCID: PMC11718163 DOI: 10.1038/s41522-024-00641-2] [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: 04/08/2024] [Accepted: 12/25/2024] [Indexed: 01/12/2025] Open
Abstract
The human body houses many distinct and interconnecting microbial populations with long-lasting systemic effects, where the oral cavity serves as a pathogens' reservoir. The correlation of different disease states strongly supports the need to understand the interplay between the oral tissue niche and microbiome. Despite efforts, the recapitulation of gingival architecture and physiological characteristics of the periodontal niche has yet to be accomplished by traditional cultural strategies. Here, we are showing for the first time the investigation of host-microbiome interactions in healthy conditions within a human oral tissue model over seven days. Our results indicated long-term host and microbiome viability, host barrier integrity, phenotypic functional response, and preservation of healthy microbial populations and interbacterial dialogs. This in vitro platform can maintain tissue homeostasis at the interface of the periodontal niche, thus, offering opportunities to identify predictive disease biomarkers and to develop intervention strategies to promote oral and overall health.
Collapse
Affiliation(s)
- M Adelfio
- Department of Biomedical Engineering, University of Massachusetts Lowell, Lowell, 01854, MA, USA
| | - G E Callen
- Department of Biomedical Engineering, University of Massachusetts Lowell, Lowell, 01854, MA, USA
| | - A R Diaz
- Department of Biomedical Engineering, University of Massachusetts Lowell, Lowell, 01854, MA, USA
| | - B J Paster
- The ADA Forsyth Institute, 245 First St, Cambridge, 02142, MA, USA
| | - X He
- The ADA Forsyth Institute, 245 First St, Cambridge, 02142, MA, USA
| | - H Hasturk
- The ADA Forsyth Institute, 245 First St, Cambridge, 02142, MA, USA
| | - C E Ghezzi
- Department of Biomedical Engineering, University of Massachusetts Lowell, Lowell, 01854, MA, USA.
| |
Collapse
|
2
|
Dai Y, Wang P, Mishra A, You K, Zong Y, Lu WF, Chow EKH, Preshaw PM, Huang D, Chew JRJ, Ho D, Sriram G. 3D Bioprinting and Artificial Intelligence-Assisted Biofabrication of Personalized Oral Soft Tissue Constructs. Adv Healthc Mater 2024:e2402727. [PMID: 39690752 DOI: 10.1002/adhm.202402727] [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: 10/09/2024] [Indexed: 12/19/2024]
Abstract
Regeneration of oral soft tissue defects, including mucogingival defects associated with the recession or loss of gingival and/or mucosal tissues around teeth and implants, is crucial for restoring oral tissue form, function, and health. This study presents a novel approach using three-dimensional (3D) bioprinting to fabricate individualized grafts with precise size, shape, and layer-by-layer cellular organization. A multicomponent polysaccharide/fibrinogen-based bioink is developed, and bioprinting parameters are optimized to create shape-controlled oral soft tissue (gingival) constructs. Rheological, printability, and shape-fidelity assays, demonstrated the influence of thickener concentration and print parameters on print resolution and shape fidelity. Artificial intelligence (AI)-derived tool enabled streamline the iterative bioprinting parameter optimization and analysis of the interaction between the bioprinting parameters. The cell-laden polysaccharide/fibrinogen-based bioinks exhibited excellent cellular viability and shape fidelity of shape-controlled, full-thickness gingival tissue constructs over the 18-day culture period. While variations in thickener concentrations within the bioink minimally impact the cellular organization and morphogenesis (gingival epithelial, connective tissue, and basement membrane markers), they influence the shape fidelity of the bioprinted constructs. This study represents a significant step toward the biofabrication of personalized soft tissue grafts, offering potential applications in the repair and regeneration of mucogingival defects associated with periodontal disease and dental implants.
Collapse
Affiliation(s)
- Yichen Dai
- Faculty of Dentistry, National University of Singapore, Singapore, 119085, Singapore
| | - Peter Wang
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, 117583, Singapore
- Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore, 117456, Singapore
| | - Apurva Mishra
- Faculty of Dentistry, National University of Singapore, Singapore, 119085, Singapore
| | - Kui You
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, 117583, Singapore
- Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore, 117456, Singapore
| | - Yuheng Zong
- Faculty of Dentistry, National University of Singapore, Singapore, 119085, Singapore
| | - Wen Feng Lu
- Department of Mechanical Engineering, National University of Singapore, Singapore, 117602, Singapore
- NUS Centre for Additive Manufacturing (AM.NUS), National University of Singapore, Singapore, 117602, Singapore
| | - Edward Kai-Hua Chow
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, 117583, Singapore
- Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore, 117456, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
| | - Philip M Preshaw
- Faculty of Dentistry, National University of Singapore, Singapore, 119085, Singapore
- School of Dentistry, University of Dundee, Dundee, DD1 4HN, UK
| | - Dejian Huang
- Department of Food, Science and Technology, National University of Singapore, Singapore, 117542, Singapore
| | - Jacob Ren Jie Chew
- Faculty of Dentistry, National University of Singapore, Singapore, 119085, Singapore
- National University Centre for Oral Health Singapore, National University Hospital, Singapore, 119085, Singapore
| | - Dean Ho
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, 117583, Singapore
- Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore, 117456, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- The Bia-Echo Asia Centre for Reproductive Longevity and Equality (ACRLE), National University of Singapore, Singapore, 117456, Singapore
| | - Gopu Sriram
- Faculty of Dentistry, National University of Singapore, Singapore, 119085, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, 117583, Singapore
- NUS Centre for Additive Manufacturing (AM.NUS), National University of Singapore, Singapore, 117602, Singapore
| |
Collapse
|
3
|
Chakraborty R, Ray P, Barik S, Banik O, Mahapatra C, Banoth E, Kumar P. A Review on Microengineering of Epithelial Barriers for Biomedical and Pharmaceutical Research. ACS APPLIED BIO MATERIALS 2024; 7:8107-8125. [PMID: 39565389 DOI: 10.1021/acsabm.4c00813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2024]
Abstract
Epithelial tissue forms a barrier around the human body and visceral organs, providing protection, permeation, sensation, and secretion. It is vital for our sustenance as it protects the tissue from harm and injury by restricting the entry of foreign bodies inside. Furthermore, it is a strong barrier to drugs, nutrients, and other essential deliverables. This layer also houses a large consortium of microbes, which thrive in tandem with human tissue, providing several health benefits. Moreover, the complex interplay of the microbiome with the barrier tissue is poorly understood. Therefore, replicating these barrier tissues on microdevices to generate physiological and pathophysiological models has been a huge interest for researchers over the last few decades. The artificially engineered reconstruction of these epithelial cellular barriers on microdevices could help underpin the host-microbe interaction, generating a physiological understanding of the tissue, tissue remodeling, receptor-based selective diffusion, drug testing, and others. In addition, these devices could reduce the burden of animal sacrifices for similar research and minimize the failure rate in drug discovery due to the use of primary human cells and others. This review discusses the nature of the epithelial barrier at different tissue sites, the recent developments in creating engineered barrier models, and their applications in pathophysiology, host-microbe interactions, drug discovery, and cytotoxicity. The review aims to provide know-how and knowledge behind engineered epithelial barrier tissue to bioengineers, biotechnologists, and scientists in allied fields.
Collapse
Affiliation(s)
- Ruchira Chakraborty
- Biodesign and Medical Devices Laboratory, Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela 769008, Odisha, India
| | - Pragyan Ray
- Biodesign and Medical Devices Laboratory, Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela 769008, Odisha, India
| | - Swagatika Barik
- Biodesign and Medical Devices Laboratory, Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela 769008, Odisha, India
- Opto-Biomedical Microsystem Laboratory, Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela, Sector-1, Rourkela 769008, Odisha, India
| | - Oindrila Banik
- Biodesign and Medical Devices Laboratory, Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela 769008, Odisha, India
- Opto-Biomedical Microsystem Laboratory, Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela, Sector-1, Rourkela 769008, Odisha, India
| | - Chinmaya Mahapatra
- Department of Biotechnology, National Institute of Technology, Raipur-492010 Chhattisgarh, India
| | - Earu Banoth
- Opto-Biomedical Microsystem Laboratory, Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela, Sector-1, Rourkela 769008, Odisha, India
| | - Prasoon Kumar
- Biodesign and Medical Devices Laboratory, Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela 769008, Odisha, India
| |
Collapse
|
4
|
Kunath BJ, De Rudder C, Laczny CC, Letellier E, Wilmes P. The oral-gut microbiome axis in health and disease. Nat Rev Microbiol 2024; 22:791-805. [PMID: 39039286 DOI: 10.1038/s41579-024-01075-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/25/2024] [Indexed: 07/24/2024]
Abstract
The human body hosts trillions of microorganisms throughout many diverse habitats with different physico-chemical characteristics. Among them, the oral cavity and the gut harbour some of the most dense and diverse microbial communities. Although these two sites are physiologically distinct, they are directly connected and can influence each other in several ways. For example, oral microorganisms can reach and colonize the gastrointestinal tract, particularly in the context of gut dysbiosis. However, the mechanisms of colonization and the role that the oral microbiome plays in causing or exacerbating diseases in other organs have not yet been fully elucidated. Here, we describe recent advances in our understanding of how the oral and intestinal microbiota interplay in relation to their impact on human health and disease.
Collapse
Affiliation(s)
- Benoit J Kunath
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg.
| | - Charlotte De Rudder
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Cedric C Laczny
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Elisabeth Letellier
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Belvaux, Luxembourg
| | - Paul Wilmes
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg.
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, Belvaux, Luxembourg.
| |
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
|
Bankar N, Latta L, Loretz B, Reda B, Dudek J, Hähl H, Hannig M, Lehr CM. Antimicrobial and antibiotic-potentiating effect of calcium peroxide nanoparticles on oral bacterial biofilms. NPJ Biofilms Microbiomes 2024; 10:106. [PMID: 39406727 PMCID: PMC11480382 DOI: 10.1038/s41522-024-00569-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 09/15/2024] [Indexed: 10/18/2024] Open
Abstract
Bacterial biofilms represent a prominent biological barrier against physical and chemical attacks. Disturbing the anaerobic microenvironment within biofilms by co-delivery of oxygen appears as a promising strategy to enhance the activity of an antibiotic. Here, we report the effect of oxygen-producing calcium peroxide nanoparticles (CaO2 NP) in combination with tobramycin sulfate (Tob). On Pseudomonas aeruginosa PAO1 biofilms in vitro, the additive effect of CaO2 NP towards Tob activity enhanced biofilm eradication by 2 log compared to Tob alone. For natural biofilms grown in the oral cavity of human volunteers in situ, treatment by CaO2 NP alone slightly increased the fraction of dead bacteria from 44% in various controls, including Tob alone, to 57%. However, the combination of CaO2 NP with Tob further increased the fraction of dead bacteria to 69%. These data confirm the intrinsic antimicrobial and antibiotic-potentiating effect of CaO2 NP also in a clinically relevant setting.
Collapse
Affiliation(s)
- Neha Bankar
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarland University, 66123, Saarbrücken, Germany
- Department of Pharmacy, Saarland University, 66123, Saarbrücken, Germany
| | - Lorenz Latta
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarland University, 66123, Saarbrücken, Germany
| | - Brigitta Loretz
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarland University, 66123, Saarbrücken, Germany.
| | - Bashar Reda
- Clinic of Operative Dentistry, Periodontology and Preventive Dentistry, University Hospital, Saarland University, Building 73, 66421, Homburg/Saar, Germany
- Department of Periodontology, School of Dentistry, Al- Shahbaa Private University, 66123, Aleppo, Syria
| | - Johanna Dudek
- Clinic of Operative Dentistry, Periodontology and Preventive Dentistry, University Hospital, Saarland University, Building 73, 66421, Homburg/Saar, Germany
| | - Hendrik Hähl
- Experimental Physics and Center for Biophysics, Saarland University, 66123, Saarbrücken, Germany
| | - Matthias Hannig
- Clinic of Operative Dentistry, Periodontology and Preventive Dentistry, University Hospital, Saarland University, Building 73, 66421, Homburg/Saar, Germany
| | - Claus-Michael Lehr
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarland University, 66123, Saarbrücken, Germany.
- Department of Pharmacy, Saarland University, 66123, Saarbrücken, Germany.
| |
Collapse
|
7
|
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
|
8
|
Zhang Y, Shang L, Roffel S, Spiekstra SW, Deng D, Gibbs S. Streptococcus mitis enhances metal-induced apoptosis in reconstructed human gingiva but not skin. Toxicol In Vitro 2024; 100:105913. [PMID: 39079590 DOI: 10.1016/j.tiv.2024.105913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 07/11/2024] [Accepted: 07/26/2024] [Indexed: 08/03/2024]
Abstract
BACKGROUND Commensal bacteria colonizing oral mucosa and skin play an essential role in maintaining host-microbiome homeostasis. It is unknown whether cytotoxicity resulting from metal ions leaching from medical devices may be influenced by commensal microbes. OBJECTIVE Determine whether the extent of apoptosis triggered by nickel or titanium ions is influenced by Streptococcus mitis and whether apoptosis occurs via the intrinsic or extrinsic apoptosis pathway. METHODS Reconstructed Human Gingiva (RHG) and Skin (RHS) were topically exposed to titanium or nickel salts in the presence or absence of S. mitis. Cytotoxicity and apoptosis were assessed by histology, immunohistochemistry, TUNEL assay, and Western Blot. RESULTS S. mitis alone resulted in negligible cytotoxicity. After metal exposure, localized apoptosis was observed in the epithelium and fibroblasts within the lamina propria hydrogel of both RHG and RHS. S. mitis enhanced metal-mediated apoptosis in gingiva but not in skin. Apoptosis was mediated via the extrinsic pathway caspase 8. Activation of the execution phase of apoptosis occurred via caspases 3 and 7, and PARP-1. CONCLUSION Our study supports the finding that metals have irritant, cytotoxic properties resulting in apoptosis when leaching into skin or gingiva. Particularly for gingiva, commensal microbes exaggerate this detrimental effect.
Collapse
Affiliation(s)
- Yan Zhang
- 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 & Immunity Institute, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands; Department of Orthodontic, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, China
| | - Lin Shang
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Sanne Roffel
- 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 & Immunity Institute, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Sander W Spiekstra
- Department of Molecular Cell Biology and Immunology, Amsterdam Infection & Immunity Institute, Amsterdam UMC, 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
| | - 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 & Immunity Institute, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
9
|
Gelin A, Masson-Meyers D, Amini F, Moharamzadeh K, Tayebi L. Collagen: The superior material for full-thickness oral mucosa tissue engineering. J Oral Biosci 2024; 66:511-518. [PMID: 38909983 DOI: 10.1016/j.job.2024.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 06/19/2024] [Accepted: 06/20/2024] [Indexed: 06/25/2024]
Abstract
BACKGROUND Tissue engineering has significantly progressed in developing full-thickness oral mucosa constructs designed to replicate the natural oral mucosa. These constructs serve as valuable in vitro models for biocompatibility testing and oral disease modeling and hold clinical potential for replacing damaged or lost oral soft tissue. However, one of the major challenges in tissue engineering of the oral mucosa is the identification of an appropriate scaffold with optimal porosity, interconnected porous networks, biodegradability, and biocompatibility. These characteristics facilitate cell migration, nutrient delivery, and vascularization. Various biomaterials have been investigated for constructing tissue-engineered oral mucosa models; collagen has demonstrated superior outcomes compared with other materials. HIGHLIGHT This review discusses the different types of tissue-engineered oral mucosa developed using various materials and includes articles published between January 2000 and December 2022 in PubMed and Google Scholar. The review focuses on the superiority of collagen-based scaffolds for tissue engineering of oral mucosa, explores in vitro applications, and discusses potential clinical applications. CONCLUSION Among the various scaffold materials used for engineering the connective tissue of the oral mucosa, collagen-based scaffolds possess excellent biological properties, offering high-quality oral mucosa constructs and high resemblance to the native human oral mucosa in terms of histology and expression of various differentiation markers.
Collapse
Affiliation(s)
- Alexandra Gelin
- Marquette University School of Dentistry, Milwaukee, WI, 53233, USA
| | | | - Fatemeh Amini
- School of Dentistry, Shahed University of Medical Sciences, Tehran, Iran
| | - Keyvan Moharamzadeh
- Hamdan Bin Mohammed College of Dental Medicine (HBMCDM), Mohammed Bin Rashid University of Medicine and Health Sciences (MBRU), Dubai, United Arab Emirates; School of Clinical Dentistry, University of Sheffield, Sheffield, United Kingdom
| | - Lobat Tayebi
- Marquette University School of Dentistry, Milwaukee, WI, 53233, USA.
| |
Collapse
|
10
|
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
|
11
|
Yang Z, Zhang S, Ying L, Zhang W, Chen X, Liang Y, Chen R, Yao K, Li C, Yu C, Jamilian P, Zarezadeh M, Kord-Varkaneh H, Wang J, Li H. The effect of probiotics supplementation on cancer-treatment complications: a critical umbrella review of interventional meta-analyses. Crit Rev Food Sci Nutr 2024:1-26. [PMID: 39002141 DOI: 10.1080/10408398.2024.2372880] [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: 07/15/2024]
Abstract
Cancer-related complications pose significant challenges in the management and treatment of patients with malignancies. Several meta-analyses have indicated improving effects of probiotics on cancer complications, while some studies have reported contentious findings. The purpose of the present study was to evaluate the efficacy of probiotics in addressing cancer complications, including diarrhea, mucositis, and infections, following chemotherapy, radiotherapy, and surgery. Relevant studies were searched in the PubMed, Scopus, Embase and Web of Science databases and Google Scholar up to September 2023. All meta-analyses addressing the effects of probiotics on all cancer treatments-induced complications including infection, diarrhea and oral mucositis were included. The pooled results were calculated using a random-effects model. Analyses of subgroups, sensitivity and publication bias were also conducted. The results revealed that the probiotics supplementation was effective on reduction of total cancer complications (OR:0.53; 95% CI: 0.44, 0.62, p < 0.001; I2=79.0%, p < 0.001), total infection rate (OR:0.47; 95%CI: 0.41, 0.52, p < 0.001; I2= 48.8%, p < 0.001); diarrhea (OR:0.50; 95%CI: 0.44, 0.57, p < 0.001; I2=44.4%, p = 0.023) and severe diarrhea (OR: 0.4; 95%CI: 0.27, 0.56, p < 0.001; I2=31.3%, p = 0.178), oral mucositis (OR: 0.76; 95%CI: 0.58, 0.94, p < 0.001; I2=95.5%, p < 0.001) and severe oral mucositis (OR:0.65, 95%CI: 0.58, 0.72 p < 0.001; I2=22.1%, p = 0.274). Multi strain probiotic (OR:0.49; 95%CI: 0.32, 0.65, p < 0.001; I2=90.7%, p < 0.001) were more efficacious than single strain (OR:0.73; 95%CI: 0.66, 0.81, p < 0.001; I2=0.00%, p = 0.786). The findings of the current umbrella meta-analysis provide strong evidence that probiotic supplementation can reduce cancer complications.
Collapse
Affiliation(s)
- Zhao Yang
- College of Life Science and Technology, Innovation Center of Molecular Diagnostics, Beijing University of Chemical Technology, Beijing, China
- College of Life Science and Technology, Key Laboratory of Protection and Utilization of Biological Resources in Tarim Basin of Xinjiang Production and Construction Corps, Tarim University, Xinjiang, China
| | - Shijie Zhang
- Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Xinchang Pharmaceutical Factory, Zhejiang Medicine Co., Ltd, Shaoxing, China
| | - Lu Ying
- College of Life Science and Technology, Innovation Center of Molecular Diagnostics, Beijing University of Chemical Technology, Beijing, China
- College of Life Science and Technology, Key Laboratory of Protection and Utilization of Biological Resources in Tarim Basin of Xinjiang Production and Construction Corps, Tarim University, Xinjiang, China
| | - Wenjing Zhang
- College of Life Science and Technology, Innovation Center of Molecular Diagnostics, Beijing University of Chemical Technology, Beijing, China
| | - Xiaoyang Chen
- College of Life Science and Technology, Innovation Center of Molecular Diagnostics, Beijing University of Chemical Technology, Beijing, China
| | - Youfeng Liang
- College of Life Science and Technology, Innovation Center of Molecular Diagnostics, Beijing University of Chemical Technology, Beijing, China
| | - Ruolan Chen
- College of Life Science and Technology, Innovation Center of Molecular Diagnostics, Beijing University of Chemical Technology, Beijing, China
| | - Keying Yao
- College of Life Science and Technology, Innovation Center of Molecular Diagnostics, Beijing University of Chemical Technology, Beijing, China
| | - Chunhui Li
- College of Life Science and Technology, Innovation Center of Molecular Diagnostics, Beijing University of Chemical Technology, Beijing, China
| | - Changyuan Yu
- College of Life Science and Technology, Innovation Center of Molecular Diagnostics, Beijing University of Chemical Technology, Beijing, China
| | - Parmida Jamilian
- School of Pharmacy and Bioengineering, Keele University, Staffordshire, UK
| | - Meysam Zarezadeh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamed Kord-Varkaneh
- Department of Nutrition and Food Hygiene, School of Medicine, Nutrition Health Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Jianfeng Wang
- Department of Urology, China-Japan Friendship Hospital, Beijing, China
| | - Hanmin Li
- Institute of Liver Diseases, Hubei Provincial Hospital of Traditional Chinese Medicine (Hubei University of Traditional Chinese Medicine Affiliated Hospital), Wuhan, Hubei Province, China
- Theory and Application Research of Liver and Kidney in Traditional Chinese Medicine, Hubei Provincial Key Laboratory, Wuhan, Hubei Province, China
| |
Collapse
|
12
|
Annunziato A, Vacca M, Cristofori F, Dargenio VN, Celano G, Francavilla R, De Angelis M. Celiac Disease: The Importance of Studying the Duodenal Mucosa-Associated Microbiota. Nutrients 2024; 16:1649. [PMID: 38892582 PMCID: PMC11174386 DOI: 10.3390/nu16111649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 05/24/2024] [Accepted: 05/24/2024] [Indexed: 06/21/2024] Open
Abstract
There is increasing evidence indicating that changes in both the composition and functionality of the intestinal microbiome are closely associated with the development of several chronic inflammatory diseases, with celiac disease (CeD) being particularly noteworthy. Thanks to the advent of culture-independent methodologies, the ability to identify and quantify the diverse microbial communities residing within the human body has been significantly improved. However, in the context of CeD, a notable challenge lies in characterizing the specific microbiota present on the mucosal surfaces of the intestine, rather than relying solely on fecal samples, which may not fully represent the relevant microbial populations. Currently, our comprehension of the composition and functional importance of mucosa-associated microbiota (MAM) in CeD remains an ongoing field of research because the limited number of available studies have reported few and sometimes contradictory results. MAM plays a crucial role in the development and progression of CeD, potentially acting as both a trigger and modulator of the immune response within the intestinal mucosa, given its proximity to the epithelial cells and direct interaction. According to this background, this review aims to consolidate the existing literature specifically focused on MAM in CeD. By elucidating the complex interplay between the host immune system and the gut microbiota, we aim to pave the way for new interventions based on novel therapeutic targets and diagnostic biomarkers for MAM in CeD.
Collapse
Affiliation(s)
- Alessandro Annunziato
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, Via Amendola 165/a, 70126 Bari, Italy; (A.A.); (G.C.); (M.D.A.)
| | - Mirco Vacca
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, Via Amendola 165/a, 70126 Bari, Italy; (A.A.); (G.C.); (M.D.A.)
| | - Fernanda Cristofori
- Interdisciplinary Department of Medicine, Pediatric Section, Children’s Hospital ‘Giovanni XXIII’, University of Bari Aldo Moro, 70126 Bari, Italy; (F.C.); (V.N.D.); (R.F.)
| | - Vanessa Nadia Dargenio
- Interdisciplinary Department of Medicine, Pediatric Section, Children’s Hospital ‘Giovanni XXIII’, University of Bari Aldo Moro, 70126 Bari, Italy; (F.C.); (V.N.D.); (R.F.)
| | - Giuseppe Celano
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, Via Amendola 165/a, 70126 Bari, Italy; (A.A.); (G.C.); (M.D.A.)
| | - Ruggiero Francavilla
- Interdisciplinary Department of Medicine, Pediatric Section, Children’s Hospital ‘Giovanni XXIII’, University of Bari Aldo Moro, 70126 Bari, Italy; (F.C.); (V.N.D.); (R.F.)
| | - Maria De Angelis
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, Via Amendola 165/a, 70126 Bari, Italy; (A.A.); (G.C.); (M.D.A.)
| |
Collapse
|
13
|
Mishra Y, Ranjan A, Mishra V, Chattaraj A, Aljabali AAA, El-Tanani M, Hromić-Jahjefendić A, Uversky VN, Tambuwala MM. The role of the gut microbiome in gastrointestinal cancers. Cell Signal 2024; 115:111013. [PMID: 38113978 DOI: 10.1016/j.cellsig.2023.111013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 12/06/2023] [Accepted: 12/13/2023] [Indexed: 12/21/2023]
Abstract
The gut microbiota present in the human digestive system is incredibly varied and is home to trillions of microorganisms. The gut microbiome is shaped at birth, while numerous genetic, dietary, and environmental variables primarily influence the microbiome composition. The importance of gut microbiota on host health is becoming more widely acknowledged. Digestion, intestinal permeability, and immunological and metabolism responses can all be affected by changes in the composition and function of the gut microbiota. There is mounting evidence that the microbial population's complex traits are important biomarkers and indicators of patient outcomes in cancer and its therapies. Numerous studies have demonstrated that changed commensal gut microorganisms contribute to the development and spread of cancer through various routes. Despite the ongoing controversy surrounding the gut microbiome and gastrointestinal cancer, accumulating evidence points to a potentially far more intricate connection than a simple cause-and-effect relationship. SIMPLE SUMMARY: Due to their high frequency and fatality rate, gastrointestinal cancers are regarded as a severe public health issue with complex medical and economic burdens. The gut microbiota may directly or indirectly interact with existing therapies like immunotherapy and chemotherapy, affecting how well a treatment works. The gut microbiome influences the immune response's activity, function, and development. Generally, certain gut bacteria impact the antitumor actions during cancer by creating particular metabolites or triggering T-cell responses. Yet, certain bacterial species have been found to promote cellular proliferation and metastasis in cancer, and comprehending these interactions in the context of cancer may help identify possible treatment targets. Notwithstanding the improvements in the field, additional research is still required to comprehend the underlying processes, examine the effects on existing therapies, and pinpoint certain bacteria and immune cells that can cause this interaction.
Collapse
Affiliation(s)
- Yachana Mishra
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Abhigyan Ranjan
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Vijay Mishra
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Aditi Chattaraj
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | - Alaa A A Aljabali
- Department of Pharmaceutical Sciences, Yarmouk University, Irbid, Jordan
| | - Mohamed El-Tanani
- College of Pharmacy, Ras Alkhama Medical and Health Sciences University, United Arab Emirates
| | - Altijana Hromić-Jahjefendić
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Hrasnicka cesta 15, Sarajevo 71000, Bosnia and Herzegovina
| | - Vladimir N Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Murtaza M Tambuwala
- Lincoln Medical School, University of Lincoln, Brayford Pool, Lincoln LN6 7TS, England, United Kingdom.
| |
Collapse
|
14
|
Sahle M, Wachendörfer M, Palkowitz AL, Nasehi R, Aveic S, Fischer H. A Fibrin-Based Human Multicellular Gingival 3D Model Provides Biomimicry and Enables Long-Term In Vitro Studies. Macromol Biosci 2024; 24:e2300162. [PMID: 37716014 DOI: 10.1002/mabi.202300162] [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: 04/18/2023] [Revised: 08/09/2023] [Indexed: 09/18/2023]
Abstract
Collagen-type I gels are widely used for the fabrication of 3D in vitro gingival models. Unfortunately, their long-term stability is low, which limits the variety of in vitro applications. To overcome this problem and achieve better hydrolytic stability of 3D gingival models, fibrin-based hydrogel blends with increased long-term stability in vitro are investigated. Two different fibrin-based hydrogels are tested: fibrin 2.5% (w/v) and fibrin 1% (w/v)/gelatin 5% (w/v). Appropriate numbers of primary human gingival fibroblasts (HGFs) and OKG4/bmi1/TERT (OKG) keratinocytes are optimized to achieve a homogeneous distribution of cells under the assumed 3D conditions. Both hydrogels support the viability of HGFs and the stability of the hydrogel over 28 days. In vitro cultivation at the air-liquid interface triggers keratinization of the epithelium and increases its thickness, allowing the formation of multiple tissue-like layers. The presence of HGFs in the hydrogel further enhances epithelial differentiation. In conclusion, a fibrin-based 3D gingival model mimics the histology of native gingiva in vitro and ensures its long-term stability in comparison with the previously reported collagen paralogs. These results open new perspectives for extending the period within which specific biological or pathological conditions of artificial gingival tissue can be evaluated.
Collapse
Affiliation(s)
- Maike Sahle
- Department of Dental Materials and Biomaterials Research, RWTH Aachen University Hospital, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Mattis Wachendörfer
- Department of Dental Materials and Biomaterials Research, RWTH Aachen University Hospital, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Alena L Palkowitz
- Department of Dental Materials and Biomaterials Research, RWTH Aachen University Hospital, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Ramin Nasehi
- Department of Dental Materials and Biomaterials Research, RWTH Aachen University Hospital, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Sanja Aveic
- Department of Dental Materials and Biomaterials Research, RWTH Aachen University Hospital, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Horst Fischer
- Department of Dental Materials and Biomaterials Research, RWTH Aachen University Hospital, Pauwelsstrasse 30, 52074, Aachen, Germany
| |
Collapse
|
15
|
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: 17] [Impact Index Per Article: 8.5] [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
|
16
|
Liu Q, Guo T, Dang W, Song Z, Wen Y, Luo H, Wang A. Correlation between salivary cytokine profiles and white spot lesions in adolescent patients receiving clear aligner orthodontic treatment. BMC Oral Health 2023; 23:857. [PMID: 37957648 PMCID: PMC10641999 DOI: 10.1186/s12903-023-03561-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Accepted: 10/20/2023] [Indexed: 11/15/2023] Open
Abstract
BACKGROUND To explore the relationship between changes in salivary cytokine levels and the occurrence of white spot lesions in adolescents receiving clear aligner orthodontic treatment and investigate the predictive value of various factors for lesion occurrence. METHODS We retrospectively analyzed sixthy eight adolescent in the permanent dentition period, who received clear aligner orthodontics in our hospital were randomly divided into two groups according to the occurrence or aggravation of white spot lesions after treatment. The general condition of the oral cavity was analyzed, saliva was collected, and inflammation-related cytokines with varying transcription levels between groups were screened by transcriptome analysis. The expression levels of inflammatory cytokines in the saliva of the patients in the two groups were measured, and the risk factors for white spot lesions were screened by correlation analysis and binary logistic regression analysis. The value of the independent and combined application of risk factors for predicting the occurrence of white spot lesions in adolescent patients after invisible orthodontic treatment was analyzed by receiver operating characteristic (ROC) curve analysis. RESULTS Transcriptome and GO and KEGG pathway analyses showed that there were differences in the transcription levels of inflammatory cytokines such as CXCL1, CXCL2, CXCL8, CCL3, CCL4, IL-1β and IL-2 between groups. The levels of CXCL8, CCL3, CCL4, IL-1β and IL-2 in the saliva of patients with white spot lesions were significantly higher in patients after invisible orthodontic treatment (P < 0.05). Correlation analysis and binary logistic regression analysis showed that elevated levels of CXCL8, IL-1β and IL-2 were independent risk factors for the occurrence of white spot lesions (P < 0.05). CXCL8 had the highest independent predictive value for the occurrence of white spot lesions (AUC = 0.773, P < 0.05), and the combination of IL-1β and IL-2 was also of high value in predicting the occurrence of white spot lesions. CONCLUSION After invisible orthodontic treatment, the oral microenvironment, including inflammatory cytokine levels, in adolescent patients changes; in particular, the levels of inflammatory cytokines such as CXCLs and ILs change. CXCL8 expression is significantly associated with the occurrence of white spot lesions and is an important potential target for the prevention and treatment of white spot lesions in the future.
Collapse
Affiliation(s)
- Qian Liu
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Clinical Research Center for Oral Diseases, Department of Orthodontics, School of Stomatology, Air Force Medical University, Xi'an, China
| | - Tao Guo
- Department of Orthodontics, TaiKang Shanghai Bybo Dental Hospital, Shanghai, China
| | - Wei Dang
- Shaanxi Provincial Key Laboratory of Craniomaxillofacial Precision Medicine Research, Department of Prosthodontics, Stomatological Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Zhixin Song
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Clinical Research Center for Oral Diseases, Department of Orthodontics, School of Stomatology, Air Force Medical University, Xi'an, China
| | - Yi Wen
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Clinical Research Center for Oral Diseases, Department of Orthodontics, School of Stomatology, Air Force Medical University, Xi'an, China
| | - Houzhuo Luo
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Clinical Research Center for Oral Diseases, Department of Orthodontics, School of Stomatology, Air Force Medical University, Xi'an, China.
| | - Axian Wang
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi Clinical Research Center for Oral Diseases, Department of Orthodontics, School of Stomatology, Air Force Medical University, Xi'an, China.
| |
Collapse
|
17
|
AlFatlawi Z, Huang M, Chau DYS, D'Aiuto F. Three dimensional (3D) gingival models in periodontal research: a systematic review. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2023; 34:58. [PMID: 37938480 PMCID: PMC10632299 DOI: 10.1007/s10856-023-06761-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 10/19/2023] [Indexed: 11/09/2023]
Abstract
The aim of this study is to systematically appraise the evidence on available full thickness 3D gingival and mucosal models (3D culture in scaffold base system) and their application in periodontal and peri-implant research. This study involved a systematic review of twenty-two studies obtained from searching from five electronic databases: MEDLINE-OVID, EMBASE, EBSCOhost, Web of Science Core Collection and LILACS, as well as a hand search of eligible articles up to September 2022. A total of 2338 studies were initially identified, after removal of duplicates (573), abstracts/title selection (1765), and full text screening (95), twenty-two studies were included, thirty-seven models were identified. Several cellular markers were reported by the studies included. The expression of keratinocytes differentiation markers (K4, K5, K10, K13, K14, K16, K17, K18, K19, involucrin, laminin5), proliferation marker (Ki67, CD90), and vimentin, Type I, II and IV collagen produced by fibroblasts were investigated in thirty models. No quantitative analyses were performed, and results of the review confirmed a substantial level of heterogeneity across experiments. In conclusion, there is currently insufficient evidence to conclude that the available 3D gingival and mucosal models can entirely recapitulate the human gingival tissue/mucosa and provide a useful research tool for periodontal and peri-implant research. This review also highlighted the lack of a standardized protocol to construct and characterize 3D gingival models. A new protocol is proposed for the characterization of in vitro gingival models for future research.
Collapse
Affiliation(s)
- Z AlFatlawi
- Periodontology Unit, UCL Eastman Dental Institute, 21 University Street, London, WC1E 6DE, UK
- Division of Biomaterials and Tissue Engineering, UCL Eastman Dental Institute, Royal Free Campus, Rowland Hill Street, London, NW3 2PF, UK
| | - M Huang
- Periodontology Unit, UCL Eastman Dental Institute, 21 University Street, London, WC1E 6DE, UK
| | - D Y S Chau
- Division of Biomaterials and Tissue Engineering, UCL Eastman Dental Institute, Royal Free Campus, Rowland Hill Street, London, NW3 2PF, UK.
| | - F D'Aiuto
- Periodontology Unit, UCL Eastman Dental Institute, 21 University Street, London, WC1E 6DE, UK
| |
Collapse
|
18
|
Rikken G, Meesters LD, Jansen PAM, Rodijk-Olthuis D, van Vlijmen-Willems IMJJ, Niehues H, Smits JPH, Oláh P, Homey B, Schalkwijk J, Zeeuwen PLJM, van den Bogaard EH. Novel methodologies for host-microbe interactions and microbiome-targeted therapeutics in 3D organotypic skin models. MICROBIOME 2023; 11:227. [PMID: 37849006 PMCID: PMC10580606 DOI: 10.1186/s40168-023-01668-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 09/08/2023] [Indexed: 10/19/2023]
Abstract
BACKGROUND Following descriptive studies on skin microbiota in health and disease, mechanistic studies on the interplay between skin and microbes are on the rise, for which experimental models are in great demand. Here, we present a novel methodology for microbial colonization of organotypic skin and analysis thereof. RESULTS An inoculation device ensured a standardized application area on the stratum corneum and a homogenous distribution of bacteria, while preventing infection of the basolateral culture medium even during prolonged culture periods for up to 2 weeks at a specific culture temperature and humidity. Hereby, host-microbe interactions and antibiotic interventions could be studied, revealing diverse host responses to various skin-related bacteria and pathogens. CONCLUSIONS Our methodology is easily transferable to a wide variety of organotypic skin or mucosal models and different microbes at every cell culture facility at low costs. We envision that this study will kick-start skin microbiome studies using human organotypic skin cultures, providing a powerful alternative to experimental animal models in pre-clinical research. Video Abstract.
Collapse
Affiliation(s)
- Gijs Rikken
- Department of Dermatology, Radboud University Medical Center (Radboudumc), Nijmegen, The Netherlands
| | - Luca D Meesters
- Department of Dermatology, Radboud University Medical Center (Radboudumc), Nijmegen, The Netherlands
| | - Patrick A M Jansen
- Department of Dermatology, Radboud University Medical Center (Radboudumc), Nijmegen, The Netherlands
| | - Diana Rodijk-Olthuis
- Department of Dermatology, Radboud University Medical Center (Radboudumc), Nijmegen, The Netherlands
| | | | - Hanna Niehues
- Department of Dermatology, Radboud University Medical Center (Radboudumc), Nijmegen, The Netherlands
| | - Jos P H Smits
- Department of Dermatology, Radboud University Medical Center (Radboudumc), Nijmegen, The Netherlands
- Department of Dermatology, University Hospital Düsseldorf, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Peter Oláh
- Department of Dermatology, University Hospital Düsseldorf, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Bernhard Homey
- Department of Dermatology, University Hospital Düsseldorf, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Joost Schalkwijk
- Department of Dermatology, Radboud University Medical Center (Radboudumc), Nijmegen, The Netherlands
| | - Patrick L J M Zeeuwen
- Department of Dermatology, Radboud University Medical Center (Radboudumc), Nijmegen, The Netherlands
| | - Ellen H van den Bogaard
- Department of Dermatology, Radboud University Medical Center (Radboudumc), Nijmegen, The Netherlands.
| |
Collapse
|
19
|
Adelfio M, Bonzanni M, Callen GE, Paster BJ, Hasturk H, Ghezzi CE. A physiologically relevant culture platform for long-term studies of in vitro gingival tissue. Acta Biomater 2023; 167:321-334. [PMID: 37331612 PMCID: PMC10528240 DOI: 10.1016/j.actbio.2023.06.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 06/06/2023] [Accepted: 06/08/2023] [Indexed: 06/20/2023]
Abstract
There is a clinical need to understand the etiologies of periodontitis, considering the growing socio-economic impact of the disease. Despite recent advances in oral tissue engineering, experimental approaches have failed to develop a physiologically relevant gingival model that combines tissue organization with salivary flow dynamics and stimulation of the shedding and non-shedding oral surfaces. Herein, we develop a dynamic gingival tissue model composed of a silk scaffold, replicating the cyto-architecture and oxygen profile of the human gingiva, along with a saliva-mimicking medium that reflected the ionic composition, viscosity, and non-Newtonian behavior of human saliva. The construct was cultured in a custom designed bioreactor, in which force profiles on the gingival epithelium were modulated through analysis of inlet position, velocity and vorticity to replicate the physiological shear stress of salivary flow. The gingival bioreactor supported the long-term in vivo features of the gingiva and improved the integrity of the epithelial barrier, critical against the invasion of pathogenic bacteria. Furthermore, the challenge of the gingival tissue with P. gingivalis lipopolysaccharide, as an in vitro surrogate for microbial interactions, indicated a greater stability of the dynamic model in maintaining tissue homeostasis and, thus, its applicability in long-term studies. The model will be integrated into future studies with the human subgingival microbiome to investigate host-pathogen and host-commensal interactions. STATEMENT OF SIGNIFICANCE: The major societal impact of human microbiome had reverberated up to the establishment of the Common Fund's Human Microbiome Project, that has the intent of studying the role of microbial communities in human health and diseases, including periodontitis, atopic dermatitis, or asthma and inflammatory bowel disease. In addition, these chronic diseases are emergent drivers of global socioeconomic status. Not only common oral diseases have been shown to be directly correlated with several systemic conditions, but they are differentially impacting some racial/ethnic and socioeconomic groups. To address this growing social disparity, the development of in vitro gingival model would provide a time and cost-effective experimental platform, able to mimic the spectrum of periodontal disease presentation, for the identification of predictive biomarkers for early-stage diagnosis.
Collapse
Affiliation(s)
- M Adelfio
- Department of Biomedical Engineering, University of Massachusetts Lowell, 1 University Avenue, Lowell, MA 01854, USA
| | - M Bonzanni
- Department of Neuroscience, School of Medicine, Tufts University, 136 Harrison Avenue, Boston, MA 02111, USA
| | - G E Callen
- Department of Biomedical Engineering, University of Massachusetts Lowell, 1 University Avenue, Lowell, MA 01854, USA
| | - B J Paster
- The Forsyth Institute, 245 First St, Cambridge, MA 02142, USA
| | - H Hasturk
- The Forsyth Institute, 245 First St, Cambridge, MA 02142, USA
| | - C E Ghezzi
- Department of Biomedical Engineering, University of Massachusetts Lowell, 1 University Avenue, Lowell, MA 01854, USA.
| |
Collapse
|
20
|
Tang K, Luo ML, Zhou W, Niu LN, Chen JH, Wang F. The integration of peri-implant soft tissues around zirconia abutments: Challenges and strategies. Bioact Mater 2023; 27:348-361. [PMID: 37180640 PMCID: PMC10172871 DOI: 10.1016/j.bioactmat.2023.04.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 03/23/2023] [Accepted: 04/09/2023] [Indexed: 05/16/2023] Open
Abstract
Stable soft tissue integration around the implant abutment attenuates pathogen penetration, protects underlying bone tissue, prevents peri-implantitis and is essential in maintaining long-term implant stability. The desire for "metal free" and "aesthetic restoration" has favored zirconia over titanium abutments, especially for implant restorations in the anterior region and for patients with thin gingival biotype. Soft tissue attachment to the zirconia abutment surface remains a challenge. A comprehensive review of advances in zirconia surface treatment (micro-design) and structural design (macro-design) affecting soft tissue attachment is presented and strategies and research directions are discussed. Soft tissue models for abutment research are described. Guidelines for development of zirconia abutment surfaces that promote soft tissue integration and evidence-based references to inform clinical choice of abutment structure and postoperative maintenance are presented.
Collapse
Affiliation(s)
- Kai Tang
- National Clinical Research Center for Oral Diseases & State Key Laboratory of Military Stomatology &Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Meng-Lin Luo
- Institute of Stomatology & Oral Maxilla Facial Key Laboratory, The First Medical Center, Chinese PLA General Hospital & Department of Stomatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Wei Zhou
- National Clinical Research Center for Oral Diseases & State Key Laboratory of Military Stomatology &Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Li-Na Niu
- National Clinical Research Center for Oral Diseases & State Key Laboratory of Military Stomatology &Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
| | - Ji-Hua Chen
- National Clinical Research Center for Oral Diseases & State Key Laboratory of Military Stomatology &Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
- Corresponding author.
| | - Fu Wang
- National Clinical Research Center for Oral Diseases & State Key Laboratory of Military Stomatology &Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, 710032, China
- Corresponding author.
| |
Collapse
|
21
|
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
|
22
|
Bruno JS, Al-Qadami GH, Laheij AMGA, Bossi P, Fregnani ER, Wardill HR. From Pathogenesis to Intervention: The Importance of the Microbiome in Oral Mucositis. Int J Mol Sci 2023; 24:ijms24098274. [PMID: 37175980 PMCID: PMC10179181 DOI: 10.3390/ijms24098274] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/11/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023] Open
Abstract
Oral mucositis (OM) is a common and impactful toxicity of standard cancer therapy, affecting up to 80% of patients. Its aetiology centres on the initial destruction of epithelial cells and the increase in inflammatory signals. These changes in the oral mucosa create a hostile environment for resident microbes, with oral infections co-occurring with OM, especially at sites of ulceration. Increasing evidence suggests that oral microbiome changes occur beyond opportunistic infection, with a growing appreciation for the potential role of the microbiome in OM development and severity. This review collects the latest articles indexed in the PubMed electronic database which analyse the bacterial shift through 16S rRNA gene sequencing methodology in cancer patients under treatment with oral mucositis. The aims are to assess whether changes in the oral and gut microbiome causally contribute to oral mucositis or if they are simply a consequence of the mucosal injury. Further, we explore the emerging role of a patient's microbial fingerprint in OM development and prediction. The maintenance of resident bacteria via microbial target therapy is under constant improvement and should be considered in the OM treatment.
Collapse
Affiliation(s)
- Julia S Bruno
- Instituto de Ensino e Pesquisa, Hospital Sírio-Libanês, São Paulo 01308-060, Brazil
| | - Ghanyah H Al-Qadami
- School of Biomedicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide 5005, Australia
| | - Alexa M G A Laheij
- Department of Oral Medicine, Academic Centre for Dentistry (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, 1081 LA Amsterdam, The Netherlands
- Department of Preventive Dentistry, Academic Centre for Dentistry (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, 1081 LA Amsterdam, The Netherlands
- Department of Oral Maxillofacial Surgery, Amsterdam UMC, University of Amsterdam, 1081 LA Amsterdam, The Netherlands
| | - Paolo Bossi
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, 25121 Brescia, Italy
| | - Eduardo R Fregnani
- Instituto de Ensino e Pesquisa, Hospital Sírio-Libanês, São Paulo 01308-060, Brazil
| | - Hannah R Wardill
- School of Biomedicine, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide 5005, Australia
- The Supportive Oncology Research Group, Precision Cancer Medicine Theme, The South Australian Health and Medical Research Institute, Adelaide 5000, Australia
| |
Collapse
|
23
|
Adelfio M, Martin‐Moldes Z, Erndt‐Marino J, Tozzi L, Duncan MJ, Hasturk H, Kaplan DL, Ghezzi CE. Three-Dimensional Humanized Model of the Periodontal Gingival Pocket to Study Oral Microbiome. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2205473. [PMID: 36825685 PMCID: PMC10131835 DOI: 10.1002/advs.202205473] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 02/03/2023] [Indexed: 06/18/2023]
Abstract
The oral cavity contains distinct microenvironments that serve as oral barriers, such as the non-shedding surface of the teeth (e.g., enamel), the epithelial mucosa and gingival tissue (attached gingiva) where microbial communities coexist. The interactions and balances between these communities are responsible for oral tissue homeostasis or dysbiosis, that ultimately dictate health or disease. Disruption of this equilibrium can lead to chronic inflammation and permanent tissue damage in the case of chronic periodontitis. There are currently no experimental tissue models able to mimic the structural, physical, and metabolic conditions present in the human oral gingival tissue to support the long-term investigation of host-pathogens imbalances. Herein, the authors report an in vitro 3D anatomical gingival tissue model, fabricated from silk biopolymer by casting a replica mold of an adult human mandibular gingiva to recreate a tooth-gum unit. The model is based on human primary cultures that recapitulate physiological tissue organization, as well as a native oxygen gradient within the gingival pocket to support human subgingival plaque microbiome with a physiologically relevant level of microbial diversity up to 24 h. The modulation of inflammatory markers in the presence of oral microbiome indicates the humanized functional response of this model and establishes a new set of tools to investigate host-pathogen imbalances in gingivitis and periodontal diseases.
Collapse
Affiliation(s)
- Miryam Adelfio
- Department of Biomedical EngineeringUniversity of Massachusetts LowellLowellMA01854USA
| | | | | | - Lorenzo Tozzi
- Department of Biomedical EngineeringTufts UniversityMedfordMA02155USA
| | | | - Hatice Hasturk
- Center for Clinical and Translational ResearchThe Forsyth InstituteCambridgeMA02142USA
| | - David L. Kaplan
- Department of Biomedical EngineeringTufts UniversityMedfordMA02155USA
| | - Chiara E. Ghezzi
- Department of Biomedical EngineeringUniversity of Massachusetts LowellLowellMA01854USA
- Department of Biomedical EngineeringTufts UniversityMedfordMA02155USA
| |
Collapse
|
24
|
Gard AL, Luu RJ, Maloney R, Cooper MH, Cain BP, Azizgolshani H, Isenberg BC, Borenstein JT, Ong J, Charest JL, Vedula EM. A high-throughput, 28-day, microfluidic model of gingival tissue inflammation and recovery. Commun Biol 2023; 6:92. [PMID: 36690695 PMCID: PMC9870913 DOI: 10.1038/s42003-023-04434-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 01/05/2023] [Indexed: 01/24/2023] Open
Abstract
Nearly half of American adults suffer from gum disease, including mild inflammation of gingival tissue, known as gingivitis. Currently, advances in therapeutic treatments are hampered by a lack of mechanistic understanding of disease progression in physiologically relevant vascularized tissues. To address this, we present a high-throughput microfluidic organ-on-chip model of human gingival tissue containing keratinocytes, fibroblast and endothelial cells. We show the triculture model exhibits physiological tissue structure, mucosal barrier formation, and protein biomarker expression and secretion over several weeks. Through inflammatory cytokine administration, we demonstrate the induction of inflammation measured by changes in barrier function and cytokine secretion. These states of inflammation are induced at various time points within a stable culture window, providing a robust platform for evaluation of therapeutic agents. These data reveal that the administration of specific small molecule inhibitors mitigates the inflammatory response and enables tissue recovery, providing an opportunity for identification of new therapeutic targets for gum disease with the potential to facilitate relevant preclinical drug efficacy and toxicity testing.
Collapse
Affiliation(s)
| | | | - Ryan Maloney
- Bioengineering Division, Draper, Cambridge, MA, USA
| | | | - Brian P Cain
- Bioengineering Division, Draper, Cambridge, MA, USA
| | | | | | | | - Jane Ong
- Colgate-Palmolive Company, Piscataway, NJ, USA
| | | | - Else M Vedula
- Bioengineering Division, Draper, Cambridge, MA, USA.
| |
Collapse
|
25
|
de Jongh CA, de Vries TJ, Bikker FJ, Gibbs S, Krom BP. Mechanisms of Porphyromonas gingivalis to translocate over the oral mucosa and other tissue barriers. J Oral Microbiol 2023; 15:2205291. [PMID: 37124549 PMCID: PMC10134951 DOI: 10.1080/20002297.2023.2205291] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2023] Open
Abstract
Introduction The oral pathogen Porphyromonas gingivalis is not only associated with periodontitis but also with systemic diseases elsewhere in the body. The mechanisms by which P. gingivalis travels from the oral cavity to other organs in the body are largely unknown. This review describes the four putative mechanisms supported by experimental evidence, which enable translocation of P. gingivalis over the oral mucosa, endothelial barriers and subsequent dissemination into the bloodstream. Mechanisms The first mechanism: proteolytic enzymes secreted by P. gingivalis degrade adhesion molecules between tissue cells, and the extracellular matrix. This weakens the structural integrity of the mucosa and allows P. gingivalis to penetrate the tissue. The second is transcytosis: bacteria actively enter tissue cells and transfer to the next layer or the extracellular space. By travelling from cell to cell, P. gingivalis reaches deeper structures. Thirdly, professional phagocytes take up P. gingivalis and travel to the bloodstream where P. gingivalis is released. Lastly, P. gingivalis can adhere to the hyphae forming Candida albicans. These hyphae can penetrate the mucosal tissue, which may allow P. gingivalis to reach deeper structures. Conclusion More research could elucidate targets to inhibit P. gingivalis dissemination and prevent the onset of various systemic diseases.
Collapse
Affiliation(s)
- Caroline A. de Jongh
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Department of Periodontology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Department of Oral Biochemistry, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Teun J. de Vries
- Department of Periodontology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Floris J. Bikker
- Department of Oral Biochemistry, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - Susan Gibbs
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), 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
- CONTACT Bastiaan P. Krom Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
26
|
Zhang Y, Shang L, Roffel S, Krom BP, Gibbs S, Deng D. Stable reconstructed human gingiva–microbe interaction model: Differential response to commensals and pathogens. Front Cell Infect Microbiol 2022; 12:991128. [PMID: 36339338 PMCID: PMC9631029 DOI: 10.3389/fcimb.2022.991128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 10/05/2022] [Indexed: 11/13/2022] Open
Abstract
Background To investigate human oral health and disease, models are required which represent the interactions between the oral mucosa and microbiome. Our aim was to develop an organotypic model which maintains viability of both host and microbes for an extended period of time. Methods Reconstructed Human Gingiva (RHG) were cultured air-lifted with or without penicillin-streptomycin (PS) and topically exposed to Streptococcus gordonii (commensal) or Aggregatibacter actinomycetemcomitans (pathogen) for 72 hours in agar. RHG histology, viability and cytokines (ELISA), and bacterial viability (colony forming units) and location (FISH) were assessed. Results The low concentration of topically applied agar did not influence RHG viability. Topically applied bacteria in agar remained localized and viable for 72 hours and did not spill over to infect RHG culture medium. PS in RHG culture medium killed topically applied bacteria. Co-culture with living bacteria did not influence RHG viability (Ki67 expression, MTT assay) or histology (epithelium differentiation, Keratin10 expression). RHG exposed to S. gordonii (with or without PS) did not influence low level of IL-6, IL-8, CCL2, CCL5, CCL20 or CXCL1 secretion. However, all cytokines increased (except CCL2) when RHG were co-cultured with A. actinomycetemcomitans. The effect was significantly more in the presence of living, rather than dead, A. actinomycetemcomitans. Both bacteria resulted in increased expression of RHG antimicrobial peptides (AMPs) Elafin and HBD-2, with S. gordonii exposure resulting in the most Elafin secretion. Conclusion This technical advance enables living human oral host–microbe interactions to be investigated during a 72-hour period and shows differences in innate immunology triggered by S. gordonii and A. actinomycetemcomitans.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Department of Orthodontic, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, China
| | - Lin Shang
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Sanne Roffel
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Bastiaan P. Krom
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Susan Gibbs
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Department of Molecular Cell Biology and Immunology, Amsterdam Infection and Immunity Institute, Amsterdam University Medical Centre, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Dongmei Deng
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- *Correspondence: Dongmei Deng,
| |
Collapse
|
27
|
Lin SK, Wu YF, Chang WJ, Feng SW, Huang HM. The Treatment Efficiency and Microbiota Analysis of Sapindus mukorossi Seed Oil on the Ligature-Induced Periodontitis Rat Model. Int J Mol Sci 2022; 23:8560. [PMID: 35955695 PMCID: PMC9369273 DOI: 10.3390/ijms23158560] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 07/28/2022] [Accepted: 07/30/2022] [Indexed: 02/05/2023] Open
Abstract
Periodontitis is a common oral disease mainly caused by bacterial infection and inflammation of the gingiva. In the prevention or treatment of periodontitis, anti-bacterial agents are used to inhibit pathogen growth, despite increasing levels of bacterial resistance. Sapindus mukorossi Gaertn (SM) seed oil has proven anti-bacterial and anti-inflammation properties. However, the possibility of using this plant to prevent or treat periodontitis has not been reported previously. The aim of this study was to evaluate the effects of SM oil on experimental periodontitis in rats by using micro-CT and microbiota analysis. The distance between cementoenamel junction (CEJ) and alveolar bone crest (ABC) on the sagittal micro-CT slide showed that total bone loss (TBL) was significantly lower in CEJ-ABC distances between SM oil and SM oil-free groups on Day 14. Histology data also showed less alveolar bone resorption, a result consistent result with micro-CT imaging. The microbiota analyzed at phylum and class levels were compared between the SM oil and SM oil-free groups on Day 7 and Day 14. At the phylum level, Proteobacteria, Firmicutes, Bacteroidetes, and Actinobacteria were the dominant bacterium. Firmicutes in box plot analysis was significantly less in the SM oil group than in the SM oil-free group on Day 7. At the class level, Bacteroidia, Gammaproteobacteria, Bacilli, Clostridia, and Erysipelotrichia were the dominant bacteria. The bacteria composition proportion of Bacilli, Clostridiay, and Erysipelotrichia could be seen in the SM oil group significantly less than in t SM oil-free group on Day 7. Overall, the present results show that topical application of SM oil can reduce bone resorption and change bacteria composition in the ligature-induced periodontitis model. According to these results, it is reasonable to suggest SM oil as a potential material for preventing oral disease.
Collapse
Affiliation(s)
- Shih-Kai Lin
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Yi-Fan Wu
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Wei-Jen Chang
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Department of Dentistry, Shuang Ho Hospital, Taipei Medical University, New Taipei City 235041, Taiwan
| | - Sheng-Wei Feng
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Haw-Ming Huang
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
28
|
Johnstone KF, Herzberg MC. Antimicrobial peptides: Defending the mucosal epithelial barrier. FRONTIERS IN ORAL HEALTH 2022; 3:958480. [PMID: 35979535 PMCID: PMC9376388 DOI: 10.3389/froh.2022.958480] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 06/30/2022] [Indexed: 11/13/2022] Open
Abstract
The recent epidemic caused by aerosolized SARS-CoV-2 virus illustrates the importance and vulnerability of the mucosal epithelial barrier against infection. Antimicrobial proteins and peptides (AMPs) are key to the epithelial barrier, providing immunity against microbes. In primitive life forms, AMPs protect the integument and the gut against pathogenic microbes. AMPs have also evolved in humans and other mammals to enhance newer, complex innate and adaptive immunity to favor the persistence of commensals over pathogenic microbes. The canonical AMPs are helictical peptides that form lethal pores in microbial membranes. In higher life forms, this type of AMP is exemplified by the defensin family of AMPs. In epithelial tissues, defensins, and calprotectin (complex of S100A8 and S100A9) have evolved to work cooperatively. The mechanisms of action differ. Unlike defensins, calprotectin sequesters essential trace metals from microbes, which inhibits growth. This review focuses on defensins and calprotectin as AMPs that appear to work cooperatively to fortify the epithelial barrier against infection. The antimicrobial spectrum is broad with overlap between the two AMPs. In mice, experimental models highlight the contribution of both AMPs to candidiasis as a fungal infection and periodontitis resulting from bacterial dysbiosis. These AMPs appear to contribute to innate immunity in humans, protecting the commensal microflora and restricting the emergence of pathobionts and pathogens. A striking example in human innate immunity is that elevated serum calprotectin protects against neonatal sepsis. Calprotectin is also remarkable because of functional differences when localized in epithelial and neutrophil cytoplasm or released into the extracellular environment. In the cytoplasm, calprotectin appears to protect against invasive pathogens. Extracellularly, calprotectin can engage pathogen-recognition receptors to activate innate immune and proinflammatory mechanisms. In inflamed epithelial and other tissue spaces, calprotectin, DNA, and histones are released from degranulated neutrophils to form insoluble antimicrobial barriers termed neutrophil extracellular traps. Hence, calprotectin and other AMPs use several strategies to provide microbial control and stimulate innate immunity.
Collapse
Affiliation(s)
| | - Mark C. Herzberg
- Department of Diagnostic and Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
29
|
Takkem A, Zakaraia S, Silan A, Alghazawi M, Sahyouni W, AL-Manadili A. The Apoptotic and Antiproliferative Effects of Capsaicin in the Developmental Stages of Oral Squamous Cell Carcinoma Induced in Hamsters. Cureus 2022; 14:e26073. [PMID: 35865422 PMCID: PMC9293258 DOI: 10.7759/cureus.26073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/18/2022] [Indexed: 11/06/2022] Open
Abstract
Background and aim Several epidemiological and experimental studies have approve that the vegetarian diet has an anticancer effect. Capsaicin is the active botanical ingredient found in red chili peppers. While the data strongly argue for the significant anticancer benefits of capsaicin, nevertheless, much information is required to shed light on the anticancer molecular mechanisms to improve knowledge and suggest potential therapeutic mechanisms for the use of capsaicin against cancer. This study aimed to investigate the effect of capsaicin on the rate of cell division and apoptosis in the development of oral squamous cell carcinoma induced in the buccal pouch of hamsters. Materials and methods The sample consisted of two groups; the first group consisted of 20 hamsters with the application of carcinogenic 7,12-dimethylbenz(a)anthracene (DMBA) in the buccal pouch (the control group) and the second group (the study group) also consisted of 20 hamsters with the application of DMBA in alternatively with capsaicin. Tissue biopsies were taken from experimental animals after sacrificing. The samples were immunostained for the detection of Ki-67 and Bcl-2 proteins. Results Immunohistochemical staining by monoclonal antibody to Ki-67 and Bcl-2 in the study group showed lower expression at all stages of oral cancer development compared with their expression in the control group. After performing the one-way (ANOVA) test, we found statistically significant differences by comparing the expression degree of Ki-67 and Bcl-2 proteins in both study groups, where the p-value was less than 0.05. Conclusion We conclude from the data of our study that capsaicin has an anti-cancer role in oral squamous cell carcinoma if applied in the digestive tract of experimental animals by inhibiting the proliferation of cancer cells and activating apoptosis in them.
Collapse
|
30
|
Takenaka S, Sotozono M, Ohkura N, Noiri Y. Evidence on the Use of Mouthwash for the Control of Supragingival Biofilm and Its Potential Adverse Effects. Antibiotics (Basel) 2022; 11:727. [PMID: 35740134 PMCID: PMC9219991 DOI: 10.3390/antibiotics11060727] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/19/2022] [Accepted: 05/26/2022] [Indexed: 12/14/2022] Open
Abstract
Antimicrobial mouthwash improves supragingival biofilm control when used in conjunction with mechanical removal as part of an oral hygiene routine. Mouthwash is intended to suppress bacterial adhesion during biofilm formation processes and is not aimed at mature biofilms. The most common evidence-based effects of mouthwash on the subgingival biofilm include the inhibition of biofilm accumulation and its anti-gingivitis property, followed by its cariostatic activities. There has been no significant change in the strength of the evidence over the last decade. A strategy for biofilm control that relies on the elimination of bacteria may cause a variety of side effects. The exposure of mature oral biofilms to mouthwash is associated with several possible adverse reactions, such as the emergence of resistant strains, the effects of the residual structure, enhanced pathogenicity following retarded penetration, and ecological changes to the microbiota. These concerns require further elucidation. This review aims to reconfirm the intended effects of mouthwash on oral biofilm control by summarizing systematic reviews from the last decade and to discuss the limitations of mouthwash and potential adverse reactions to its use. In the future, the strategy for oral biofilm control may shift to reducing the biofilm by detaching it or modulating its quality, rather than eliminating it, to preserve the benefits of the normal resident oral microflora.
Collapse
Affiliation(s)
- Shoji Takenaka
- Division of Cariology, Operative Dentistry and Endodontics, Faculty of Dentistry & Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8514, Japan; (M.S.); (N.O.); (Y.N.)
| | | | | | | |
Collapse
|
31
|
Implications of microbe-mediated crosstalk in the gut: Impact on metabolic diseases. Biochim Biophys Acta Mol Cell Biol Lipids 2022; 1867:159180. [PMID: 35568374 DOI: 10.1016/j.bbalip.2022.159180] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 04/21/2022] [Accepted: 05/02/2022] [Indexed: 01/06/2023]
Abstract
Metabolic diseases continue to afflict most of the U.S. population. Advancements in gut microbiota research have led to the discovery of various functional roles of microorganisms that influence the development of obesity and co-morbidities including type 2 diabetes, non-alcoholic fatty liver disease and cardiovascular disease. Many mechanisms behind these host-microbe interactions stem from processes involving the intestinal epithelium including lipid metabolism. Thus, the purpose of this review is to discuss gut microbe-mediated changes in intestinal physiology and lipid metabolism that contribute to obesity, type 2 diabetes, non-alcoholic fatty liver disease and cardiovascular disease. Within each disease state, the causal role of bacteria in both driving disease development and protecting against metabolic disease will be discussed.
Collapse
|
32
|
Abstract
Microbial communities are eubiotic ecosystems that interact dynamically and synergistically with the human body. Imbalances in these interactions may cause dysbiosis by enhancing the occurrence of inflammatory conditions, such as periodontal or inflammatory bowel diseases. However, the mechanisms that lie behind eubiosis-dysbiosis transitions are still unclear and constantly being redefined. While the societal impact of these diseases is steadily increasing, the lack of a clear understanding behind the onset of the inflammatory conditions prevents the proper clinical strategies from being formulated. Although preclinical and clinical models and short-term planar in vitro cultures represent superb research tools, they are still lacking human relevance and long-term use. Bioreactors and organs-on-a-chip have attracted interest because of their ability to recreate and sustain the physical, structural, and mechanical features of the native environment, as well as to support long-term coculture of mammalian cells and the microbiome through modulation of pH and oxygen gradients. Existing devices, however, are still under development to sustain the microbiome-host coculture over long periods of time. In this scenario, to understand disease triggers and develop therapeutics, research efforts should command the development of three-dimensional constructs that would allow the investigation of processes underlying the microbial community assembly and how microorganisms influence host traits in both acute and chronic conditions.
Collapse
Affiliation(s)
- Miryam Adelfio
- University of Massachusetts-Lowell, Department of Biomedical Engineering, One University Avenue, Lowell, Massachusetts 01854, United States
| | - Chiara Elia Ghezzi
- University of Massachusetts-Lowell, Department of Biomedical Engineering, One University Avenue, Lowell, Massachusetts 01854, United States
| |
Collapse
|
33
|
Tubero Euzebio Alves V, Al-Attar A, Alimova Y, Maynard MH, Kirakodu S, Martinez-Porras A, Hawk GS, Ebersole JL, Stamm S, Gonzalez OA. Streptococcus gordonii-Induced miRNAs Regulate CCL20 Responses in Human Oral Epithelial Cells. Infect Immun 2022; 90:e0058621. [PMID: 35099275 PMCID: PMC8929334 DOI: 10.1128/iai.00586-21] [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: 11/19/2021] [Accepted: 12/10/2021] [Indexed: 11/20/2022] Open
Abstract
The mechanisms through which oral commensal bacteria mitigates uncontrolled inflammatory responses of the oral mucosa remain unknown. Here, we show that representative oral bacterial species normally associated with oral health [S. gordonii (Sg), V. parvula (Vp), A. naeslundii (An), C. sputigena (Cs), and N. mucosa (Nm)] enhanced differential chemokine responses in oral epithelial cells (OECs), with some bacteria (An, Vp, and Nm) inducing higher chemokine levels (CXCL1, CXCL8) than others (Sg, Cs). Although all bacterial species (except Cs) increased CCL20 mRNA levels consistent with protein elevations in cell lysates, only An, Vp, and Nm induced higher CCL20 secretion, similar to the effect of the oral pathogen F. nucleatum (Fn). In contrast, most CCL20 remained associated with OECs exposed to Sg and negligible amounts released into the cell supernatants. Consistently, Sg attenuated An-induced CCL20. MiR-4516 and miR-663a were identified as Sg-specifically induced miRNAs modulating validated targets of chemokine-associated pathways. Cell transfection with miR-4516 and miR-663a decreased An- and Fn-induced CCL20. MiRNA upregulation and attenuation of An-induced CCL20 by Sg were reversed by catalase. Up-regulation of both miRNAs was specifically enhanced by oral streptococci H2O2-producers. These findings suggest that CCL20 levels produced by OECs in response to bacterial challenge are regulated by Sg-induced miR-4516 and miR-663a in a mechanism that involves hydrogen peroxide. This type of molecular mechanism could partly explain the central role of specific oral streptococcal species in balancing inflammatory and antimicrobial responses given the critical role of CCL20 in innate (antimicrobial) and adaptive immunity (modulates Th17 responses).
Collapse
Affiliation(s)
| | - Ahmad Al-Attar
- Center for Oral Health Research, College of Dentistry, University of Kentucky, Lexington, Kentucky, USA
| | - Yelena Alimova
- Center for Oral Health Research, College of Dentistry, University of Kentucky, Lexington, Kentucky, USA
| | - Marshall H. Maynard
- Center for Oral Health Research, College of Dentistry, University of Kentucky, Lexington, Kentucky, USA
| | - Sreenatha Kirakodu
- Center for Oral Health Research, College of Dentistry, University of Kentucky, Lexington, Kentucky, USA
| | - Andrés Martinez-Porras
- Center for Oral Health Research, College of Dentistry, University of Kentucky, Lexington, Kentucky, USA
| | - Gregory S. Hawk
- Department of Statistics, College of Arts and Sciences, University of Kentucky, Lexington, Kentucky, USA
| | - Jeffrey L. Ebersole
- Department of Biomedical Sciences, School of Dental Medicine, University of Nevada Las Vegas, USA
| | - Stefan Stamm
- Department of Biochemistry, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
| | - Octavio A. Gonzalez
- Center for Oral Health Research, College of Dentistry, University of Kentucky, Lexington, Kentucky, USA
- Division of Periodontology, College of Dentistry, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
34
|
McDew-White M, Lee E, Alvarez X, Sestak K, Ling BJ, Byrareddy SN, Okeoma CM, Mohan M. Cannabinoid control of gingival immune activation in chronically SIV-infected rhesus macaques involves modulation of the indoleamine-2,3-dioxygenase-1 pathway and salivary microbiome. EBioMedicine 2022; 75:103769. [PMID: 34954656 PMCID: PMC8715300 DOI: 10.1016/j.ebiom.2021.103769] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 11/28/2021] [Accepted: 12/07/2021] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND HIV/SIV-associated periodontal disease (gingivitis/periodontitis) (PD) represents a major comorbidity affecting people living with HIV (PLWH) on combination anti-retroviral therapy (cART). PD is characterized by chronic inflammation and dysbiosis. Nevertheless, the molecular mechanisms and use of feasible therapeutic strategies to reduce/reverse inflammation and dysbiosis remain understudied and unaddressed. METHODS Employing a systems biology approach, we report molecular, metabolome and microbiome changes underlying PD and its modulation by phytocannabinoids [delta-9-tetrahydrocannabinol (Δ9-THC)] in uninfected and SIV-infected rhesus macaques (RMs) untreated (VEH-untreated/SIV) or treated with vehicle (VEH/SIV) or Δ9-THC (THC/SIV). FINDINGS VEH- untreated/SIV but not THC/SIV RMs showed significant enrichment of genes linked to anti-viral defense, interferon-β, NFκB, RIG-1, and JAK-STAT signaling. We focused on the anti-microbial DUOX1 and immune activation marker IDO1 that were reciprocally regulated in the gingiva of VEH-untreated/SIV RMs. Both proteins localized to the gingival epithelium and CD163+ macrophages, and showed differential expression in the gingiva of THC/SIV and VEH/SIV RMs. Additionally, inflammation-associated miR-21, miR-142-3p, miR-223, and miR-125a-5p showed significantly higher expression in the gingiva of VEH/SIV RMs. In human primary gingival epithelial cells, miR-125a-5p post-transcriptionally downregulated DUOX1 and THC inhibited IDO1 protein expression through a cannabinoid receptor-2 mediated mechanism. Interestingly, THC/SIV RMs showed relatively reduced plasma levels of kynurenine, kynurenate, and the neurotoxic quinolinate compared to VEH/SIV RMs at 5 months post SIV infection (MPI). Most importantly, THC blocked HIV/SIV-induced depletion of Firmicutes and Bacteroidetes, and reduced Gammaproteobacteria abundance in saliva. Reduced IDO1 protein expression was associated with significantly (p<0.05) higher abundance of Prevotella, Lactobacillus (L. salivarius, L. buchneri, L. fermentum, L. paracasei, L. rhamnosus, L. johnsonii) and Bifidobacteria and reduced abundance of the pathogenic Porphyromonas cangingivalis and Porphyromonas macacae at 5MPI. INTERPRETATION The data provides deeper insights into the molecular mechanisms underlying HIV/SIV-induced PD and more importantly, the anti-inflammatory and anti-dysbiotic properties of THC in the oral cavity. Overall, these translational findings suggest that phytocannabinoids may help reduce gingival/systemic inflammation, salivary dysbiosis and potentially metabolic disease/syndrome in PLWH on cART and those with no access to cART or do not suppress the virus under cART. FUNDING Research reported in this publication was supported by the National Institutes of Health Award Numbers R01DA052845 (MM and SNB), R01DA050169 (MM and CO), R01DA042524 and R56DE026930 (MM), and P51OD011104 and P51OD011133. The content is solely the responsibility of the authors and does not necessarily represent the official views of the NIH.
Collapse
Affiliation(s)
- Marina McDew-White
- Texas Biomedical Research Institute, Southwest National Primate Research Center, 8715 West Military Road, San Antonio, TX 78227, United States
| | - Eunhee Lee
- Texas Biomedical Research Institute, Southwest National Primate Research Center, 8715 West Military Road, San Antonio, TX 78227, United States
| | - Xavier Alvarez
- Texas Biomedical Research Institute, Southwest National Primate Research Center, 8715 West Military Road, San Antonio, TX 78227, United States
| | - Karol Sestak
- PreCliniTria, LLC., Mandeville, LA 70471, United States; Tulane National Primate Research Center, Covington LA 70433, United States
| | - Binhua J Ling
- Texas Biomedical Research Institute, Southwest National Primate Research Center, 8715 West Military Road, San Antonio, TX 78227, United States
| | - Siddappa N Byrareddy
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Chioma M Okeoma
- Department of Pharmacology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794-8651, United States
| | - Mahesh Mohan
- Texas Biomedical Research Institute, Southwest National Primate Research Center, 8715 West Military Road, San Antonio, TX 78227, United States.
| |
Collapse
|
35
|
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: 0] [Impact Index Per Article: 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
|
36
|
Wu X, Qian L, Liu K, Wu J, Shan Z. Gastrointestinal microbiome and gluten in celiac disease. Ann Med 2021; 53:1797-1805. [PMID: 34647492 PMCID: PMC8519548 DOI: 10.1080/07853890.2021.1990392] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 09/30/2021] [Indexed: 01/11/2023] Open
Abstract
Coeliac disease (CD), also known as gluten sensitive enteropathy, is an autoimmune intestinal disease induced by gluten in genetically susceptible individuals. Gluten is a common ingredient in daily diet and is one of the main environmental factors to induce coeliac disease. Adhering to gluten free diet (GFD) is an effective method for treating CD. Microbiota plays an extremely important role in maintaining human health, and diet is the main factor to regulate the composition and function of gut microbiota. Recent studies have shown that gluten metabolism is closely related to gastrointestinal tract (GIT) microbiota. With the increasing prevalence of coeliac disease, there is a need for alternative treatments to GFD. In this review, biological medication of gluten, relationship between gluten and gut microflora, effect of GFD on GIT microflora, and effect of probiotics on CD were reviewed. By analysing the research progress on relationship between gluten and gastrointestinal microbiome in coeliac disease, this review tried to explore the prospective and potential mechanism of microecological agents in treating coeliac disease.
Collapse
Affiliation(s)
- Xingxing Wu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Lin Qian
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Kexin Liu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Jing Wu
- Institute of Chinese Medicine, Nanjing Drum Tower Hospital, Nanjing University, Drum Tower Clinical Medicine College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhaowei Shan
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
37
|
Saliva-derived microcosm biofilms grown on different oral surfaces in vitro. NPJ Biofilms Microbiomes 2021; 7:74. [PMID: 34504090 PMCID: PMC8429667 DOI: 10.1038/s41522-021-00246-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 08/20/2021] [Indexed: 12/24/2022] Open
Abstract
The microbial composition of a specific oral niche could be influenced by initial bacterial adherence, nutrient and physiological property of the local surface. To investigate the influence of nutrient and surface properties on microbial composition, saliva-derived biofilms were grown in agar on three substrata: Reconstructed Human Gingiva (RHG), a hydroxyapatite (HAP) surface, and a titanium (TI) surface. Agar was mixed with either Brain Heart Infusion (BHI) or Thompson (TP) medium. After 1, 3, or 5 days, biofilm viability (by colony forming units) and microbiome profiles (by 16 S rDNA amplicon sequencing) were determined. On RHG, biofilm viability and composition were similar between BHI and TP. However, on the abiotic substrata, biofilm properties greatly depended on the type of medium and substratum. In BHI, the viability of HAP-biofilm first decreased and then increased, whereas that of TI-biofilm decreased in time until a 6-log reduction. In TP, either no or a 2-log reduction in viability was observed for HAP- or TI-biofilms respectively. Furthermore, different bacterial genera (or higher level) were differentially abundant in the biofilms on 3 substrata: Haemophilus and Porphyromonas for RHG; Bacilli for HAP and Prevotella for TI. In conclusion, RHG, the biotic substratum, is able to support a highly viable and diverse microbiome. In contrast, the viability and diversity of the biofilms on the abiotic substrata were influenced by the substrata type, pH of the environment and the richness of the growth media. These results suggest that the host (oral mucosa) plays a vital role in the oral ecology.
Collapse
|
38
|
The Bigger Picture: Why Oral Mucosa Heals Better Than Skin. Biomolecules 2021; 11:biom11081165. [PMID: 34439831 PMCID: PMC8394648 DOI: 10.3390/biom11081165] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/29/2021] [Accepted: 08/02/2021] [Indexed: 02/06/2023] Open
Abstract
Wound healing is an essential process to restore tissue integrity after trauma. Large skin wounds such as burns often heal with hypertrophic scarring and contractures, resulting in disfigurements and reduced joint mobility. Such adverse healing outcomes are less common in the oral mucosa, which generally heals faster compared to skin. Several studies have identified differences between oral and skin wound healing. Most of these studies however focus only on a single stage of wound healing or a single cell type. The aim of this review is to provide an extensive overview of wound healing in skin versus oral mucosa during all stages of wound healing and including all cell types and molecules involved in the process and also taking into account environmental specific factors such as exposure to saliva and the microbiome. Next to intrinsic properties of resident cells and differential expression of cytokines and growth factors, multiple external factors have been identified that contribute to oral wound healing. It can be concluded that faster wound closure, the presence of saliva, a more rapid immune response, and increased extracellular matrix remodeling all contribute to the superior wound healing and reduced scar formation in oral mucosa, compared to skin.
Collapse
|
39
|
Lagosz-Cwik KB, Wielento A, Lipska W, Kantorowicz M, Darczuk D, Kaczmarzyk T, Gibbs S, Potempa J, Grabiec AM. hTERT-immortalized gingival fibroblasts respond to cytokines but fail to mimic primary cell responses to Porphyromonas gingivalis. Sci Rep 2021; 11:10770. [PMID: 34031466 PMCID: PMC8144196 DOI: 10.1038/s41598-021-90037-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 05/05/2021] [Indexed: 01/30/2023] Open
Abstract
In periodontitis, gingival fibroblasts (GFs) interact with and respond to oral pathogens, significantly contributing to perpetuation of chronic inflammation and tissue destruction. The aim of this study was to determine the usefulness of the recently released hTERT-immortalized GF (TIGF) cell line for studies of host–pathogen interactions. We show that TIGFs are unable to upregulate expression and production of interleukin (IL)-6, IL-8 and prostaglandin E2 upon infection with Porphyromonas gingivalis despite being susceptible to adhesion and invasion by this oral pathogen. In contrast, induction of inflammatory mediators in TNFα- or IL-1β-stimulated TIGFs is comparable to that observed in primary GFs. The inability of TIGFs to respond directly to P. gingivalis is caused by a specific defect in Toll-like receptor-2 (TLR2) expression, which is likely driven by TLR2 promoter hypermethylation. Consistently, TIGFs fail to upregulate inflammatory genes in response to the TLR2 agonists Pam2CSK4 and Pam3CSK4. These results identify important limitations of using TIGFs to study GF interaction with oral pathogens, though these cells may be useful for studies of TLR2-independent processes. Our observations also emphasize the importance of direct comparisons between immortalized and primary cells prior to using cell lines as models in studies of any biological processes.
Collapse
Affiliation(s)
- Katarzyna B Lagosz-Cwik
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
| | - Aleksandra Wielento
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland
| | - Weronika Lipska
- Department of Periodontology and Clinical Oral Pathology, Faculty of Medicine, Jagiellonian University Medical College, Kraków, Poland
| | - Malgorzata Kantorowicz
- Department of Periodontology and Clinical Oral Pathology, Faculty of Medicine, Jagiellonian University Medical College, Kraków, Poland
| | - Dagmara Darczuk
- Department of Periodontology and Clinical Oral Pathology, Faculty of Medicine, Jagiellonian University Medical College, Kraków, Poland
| | - Tomasz Kaczmarzyk
- Department of Periodontology and Clinical Oral Pathology, Faculty of Medicine, Jagiellonian University Medical College, Kraków, Poland.,Department of Oral Surgery, Faculty of Medicine, Jagiellonian University Medical College, Kraków, Poland
| | - Susan Gibbs
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.,Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Jan Potempa
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland. .,Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, KY, USA.
| | - Aleksander M Grabiec
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland.
| |
Collapse
|
40
|
Relationship of the Esophageal Microbiome and Tissue Gene Expression and Links to the Oral Microbiome: A Randomized Clinical Trial. Clin Transl Gastroenterol 2020; 11:e00235. [PMID: 33512805 PMCID: PMC7721221 DOI: 10.14309/ctg.0000000000000235] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
INTRODUCTION Although the microbiome is altered in various esophageal diseases, there is no direct evidence for a link between the oral or esophageal microbiome and underlying esophageal tissue. Here, we aimed to address these gaps through use of an antimicrobial mouth rinse to modify the esophageal microbiome and tissue gene expression. METHODS In this randomized controlled trial, patients scheduled to undergo endoscopy for clinical indications used chlorhexidine mouth rinse or no treatment for 2 weeks before endoscopy. Oral swabs and saliva were collected at baseline and at follow-up, and the esophagus was sampled on the day of endoscopy. The microbiome was analyzed by 16S rRNA gene sequencing, and esophageal tissue gene expression was ascertained by RNA-Seq. RESULTS Twenty subjects were enrolled and included in the analyses. Within individuals, the oral and esophageal microbiome composition was significantly correlated. Chlorhexidine treatment associated with significant alterations to the relative abundance of several esophageal bacterial taxa, and to expression of genes in the esophagus including reductions in periostin, claudin-18, chemokines CXCL1 and CXCL13, and several members of the tumor necrosis factor receptor superfamily. A taxon in genus Haemophilus in the esophagus also associated with significant changes in tissue gene expression. DISCUSSION The oral and esophageal microbiomes are closely related within individuals, and esophageal microbiome alterations correlate with tissue gene expression changes. The esophageal microbiome may act as an important cofactor that influences pathogenesis and outcomes of diseases such as eosinophilic esophagitis, gastroesophageal reflux, and Barrett's esophagus.
Collapse
|
41
|
Jurdziński KT, Potempa J, Grabiec AM. Epigenetic regulation of inflammation in periodontitis: cellular mechanisms and therapeutic potential. Clin Epigenetics 2020; 12:186. [PMID: 33256844 PMCID: PMC7706209 DOI: 10.1186/s13148-020-00982-7] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 11/17/2020] [Indexed: 02/06/2023] Open
Abstract
Epigenetic mechanisms, namely DNA and histone modifications, are critical regulators of immunity and inflammation which have emerged as potential targets for immunomodulating therapies. The prevalence and significant morbidity of periodontitis, in combination with accumulating evidence that genetic, environmental and lifestyle factors cannot fully explain the susceptibility of individuals to disease development, have driven interest in epigenetic regulation as an important factor in periodontitis pathogenesis. Aberrant promoter methylation profiles of genes involved in inflammatory activation, including TLR2, PTGS2, IFNG, IL6, IL8, and TNF, have been observed in the gingival tissue, peripheral blood or buccal mucosa from patients with periodontitis, correlating with changes in expression and disease severity. The expression of enzymes that regulate histone acetylation, in particular histone deacetylases (HDACs), is also dysregulated in periodontitis-affected gingival tissue. Infection of gingival epithelial cells, gingival fibroblasts and periodontal ligament cells with the oral pathogens Porphyromonas gingivalis or Treponema denticola induces alterations in expression and activity of chromatin-modifying enzymes, as well as site-specific and global changes in DNA methylation profiles and in histone acetylation and methylation marks. These epigenetic changes are associated with excessive production of inflammatory cytokines, chemokines, and matrix-degrading enzymes that can be suppressed by small molecule inhibitors of HDACs (HDACi) or DNA methyltransferases. HDACi and inhibitors of bromodomain-containing BET proteins ameliorate inflammation, osteoclastogenesis, and alveolar bone resorption in animal models of periodontitis, suggesting their clinical potential as host modulation therapeutic agents. However, broader application of epigenomic methods will be required to create a comprehensive map of epigenetic changes in periodontitis. The integration of functional studies with global analyses of the epigenetic landscape will provide critical information on the therapeutic and diagnostic potential of epigenetics in periodontal disease.
Collapse
Affiliation(s)
- Krzysztof T Jurdziński
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland
| | - Jan Potempa
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland.,Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, KY, USA
| | - Aleksander M Grabiec
- Department of Microbiology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Kraków, Poland.
| |
Collapse
|
42
|
Shang L, Deng D, Roffel S, Gibbs S. Differential influence of Streptococcus mitis on host response to metals in reconstructed human skin and oral mucosa. Contact Dermatitis 2020; 83:347-360. [PMID: 32677222 PMCID: PMC7693211 DOI: 10.1111/cod.13668] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 07/10/2020] [Accepted: 07/15/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND Skin and oral mucosa are continuously exposed to potential metal sensitizers while hosting abundant microbes, which may influence the host response to sensitizers. This host response may also be influenced by the route of exposure that is skin or oral mucosa, due to their different immune properties. OBJECTIVE Determine how commensal Streptococcus mitis influences the host response to nickel sulfate (sensitizer) and titanium(IV) bis(ammonium lactato)dihydroxide (questionable sensitizer) in reconstructed human skin (RHS) and gingiva (RHG). METHODS RHS/RHG was exposed to nickel or titanium, in the presence or absence of S. mitis for 24 hours. Histology, cytokine secretion, and Toll-like receptors (TLRs) expression were assessed. RESULTS S. mitis increased interleukin (IL)-6, CXCL8, CCL2, CCL5, and CCL20 secretion in RHS but not in RHG; co-application with nickel further increased cytokine secretion. In contrast, titanium suppressed S. mitis-induced cytokine secretion in RHS and had no influence on RHG. S. mitis and metals differentially regulated TLR1 and TLR4 in RHS, and predominantly TLR4 in RHG. CONCLUSION Co-exposure of S. mitis and nickel resulted in a more potent innate immune response in RHS than in RHG, whereas titanium remained inert. These results indicate the important influence of commensal microbes and the route of exposure on the host's response to metals.
Collapse
Affiliation(s)
- Lin Shang
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam (ACTA)University of Amsterdam and Vrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Dongmei Deng
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam (ACTA)University of Amsterdam and Vrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Sanne Roffel
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA)University of Amsterdam and Vrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Susan Gibbs
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA)University of Amsterdam and Vrije Universiteit AmsterdamAmsterdamThe Netherlands
- Department of Molecular Cell Biology and ImmunologyAmsterdam UMC, Vrije Universiteit AmsterdamAmsterdamThe Netherlands
| |
Collapse
|
43
|
Alfonso García SL, Parada-Sanchez MT, Arboleda Toro D. The phenotype of gingival fibroblasts and their potential use in advanced therapies. Eur J Cell Biol 2020; 99:151123. [PMID: 33070040 DOI: 10.1016/j.ejcb.2020.151123] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 08/13/2020] [Accepted: 08/15/2020] [Indexed: 02/06/2023] Open
Abstract
Advanced therapies in medicine use stem cells, gene editing, and tissues to treat a wide range of conditions. One of their goals is to stimulate endogenous repair of tissues and organs by manipulating stem cells and their niche, as well as to optimize the intrinsic characteristics and plasticity of differentiated cells in adult tissues. In this context, fibroblasts emerge as an alternative source to stem cells because they share phenotypic and regenerative characteristics. Specifically, fibroblasts of the oral mucosae have been shown to have improved regenerative capacity compared to other fibroblast populations. Additionally, their easy access by means of minimally invasive procedures without generating aesthetic problems, with easy and rapid in vitro expansion and with great capacity to respond to extrinsic factors, make oral fibroblasts an attractive and interesting resource for regenerative medicine. This review summarizes current concepts regarding the phenotypic and functional aspects of human Gingival Fibroblasts and their niche, differentiating them from other fibroblast populations of oral-lining mucosa and skin fibroblasts. Furthermore, some applications are presented in regenerative medicine, emphasizing on the biological potential of human Gingival Fibroblasts.
Collapse
Affiliation(s)
- Sandra Liliana Alfonso García
- Department of Integrated Basic Studies, Faculty of Dentistry, Universidad de Antioquia, Medellín, 050010, Colombia; Department of Oral Health, Faculty of Dentistry, Universidad Nacional de Colombia, Bogotá, 111311, Colombia.
| | | | - David Arboleda Toro
- Department of Integrated Basic Studies, Faculty of Dentistry, Universidad de Antioquia, Medellín, 050010, Colombia
| |
Collapse
|
44
|
Kennedy MS, Chang EB. The microbiome: Composition and locations. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2020; 176:1-42. [PMID: 33814111 DOI: 10.1016/bs.pmbts.2020.08.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The human body is home to a diverse and functionally important assemblage of symbiotic microbes that varies predictably over different spatial scales, both within and across body sites. The composition of these spatially distinct microbial consortia can be impacted by a variety of stochastic and deterministic forces, including dispersal from different source communities, and selection by regionally-specific host processes for the enrichment of physiologically significant taxa. In this chapter, we review the composition, function, and assembly of the healthy human gastrointestinal, skin, vaginal, and respiratory microbiomes, with special emphasis on the regional distribution of microbes throughout the gastrointestinal tract.
Collapse
Affiliation(s)
- Megan S Kennedy
- Medical Scientist Training Program, Pritzker School of Medicine, The University of Chicago, Chicago, IL, United States; Department of Ecology & Evolution, The University of Chicago, Chicago, IL, United States
| | - Eugene B Chang
- Department of Medicine, Knapp Center for Biomedical Discovery, The University of Chicago, Chicago, IL, United States.
| |
Collapse
|
45
|
Gao H, Yang Y, Cao Z, Ran J, Zhang C, Huang Y, Yang M, Zhao S, An Q, Pan H. Characteristics of the Jejunal Microbiota in 35-Day-Old Saba and Landrace Piglets. Pol J Microbiol 2020; 69:367-378. [PMID: 33574866 PMCID: PMC7810115 DOI: 10.33073/pjm-2020-041] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 08/17/2020] [Accepted: 08/19/2020] [Indexed: 12/18/2022] Open
Abstract
The balanced microbiological system is a significant hallmark of piglet health. One of the crucial factors affecting intestinal microbiota is the host’s genetics. This study explored the difference in the diversity of jejunal microbiota between Saba (SB) and Landrace (LA) piglets. Nine Saba and nine Landrace piglets were fed with sow’s milk until day 35. Jejunal contents were harvested for 16S rRNA sequencing. The birth weight, body weight, and average daily gain of Saba piglets were lower than those of Landrace piglets (p < 0.01). Firmicutes were the main phylum in Saba and Landrace piglets, and the Saba piglets had a higher (p < 0.05) abundance of Bacteroidetes compared with Landrace piglets. The two most abundant genera were Lactobacilli and Clostridium XI in the jejunum of Landrace and Saba piglets. Compared with Landrace piglets, the Saba piglets had significantly lower (p < 0.05) abundance of Veillonella, Streptococcus, and Saccharibacteria genera incertae sedis. The functional prediction showed that “d-glutamine and d-glutamate metabolism” and “one carbon pool by folate” pathways were enriched in Saba piglets, while “limonene and pinene degradation”, “tryptophan metabolism”, and “sulfur relay system” pathways were enriched in Landrace piglets. In summary, the growth performance was higher for Landrace piglets compared with Saba piglets due to their genetic characteristics. The rich diversity and fewer infection-associated taxa were observed in Saba piglets, partially accounting for their higher adaptability to environmental perturbations than Landrace piglets. Furthermore, different pig breeds may regulate their health through different metabolic pathways.
Collapse
Affiliation(s)
- Huan Gao
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed Science, Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Yuting Yang
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed Science, Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Zhenhui Cao
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed Science, Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Jinming Ran
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed Science, Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Chunyong Zhang
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed Science, Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Ying Huang
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed Science, Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Minghua Yang
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed Science, Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Sumei Zhao
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed Science, Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Qingcong An
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed Science, Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Hongbin Pan
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed Science, Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
| |
Collapse
|
46
|
Rodrigues Neves CT, Spiekstra SW, de Graaf NPJ, Rustemeyer T, Feilzer AJ, Kleverlaan CJ, Gibbs S. Titanium salts tested in reconstructed human skin with integrated MUTZ-3-derived Langerhans cells show an irritant rather than a sensitizing potential. Contact Dermatitis 2020; 83:337-346. [PMID: 32677096 PMCID: PMC7689826 DOI: 10.1111/cod.13666] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 07/10/2020] [Accepted: 07/15/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND The nature of clinically related adverse reactions to titanium is still unknown. OBJECTIVE To determine whether titanium salts have irritant or sensitizing potential in a reconstructed human skin (RHS) model with integrated Langerhans cells (LCs). METHODS RHS-LCs (ie, reconstructed epidermis) containing primary differentiated keratinocytes and CFSE+ CD1a+ -LCs generated from the MUTZ-3 cell line on a primary fibroblast-populated collagen hydrogel (dermis) were topically exposed to titanium(IV) bis(ammonium lactato)dihydroxide (TiALH). LC migration and plasticity were determined. RESULTS TiALH resulted in CFSE+ CD1a+ -LC migration out of the epidermis. Neutralizing antibodies to CCL5 and CXCL12 showed that LC migration was CCL5 and not CXCL12 mediated. LCs accumulating within the dermis after TiALH exposure were CFSE+ Lang+ CD68+ which is characteristic of a phenotypic switch of MUTZ-LC to a macrophage-like cell. Furthermore, TiALH did not result in increased interleukin (IL)-1β or CCR7 messenger RNA (mRNA) in the dermis, but did result in increased IL-10 mRNA. In addition, monocultures of MUTZ-LCs failed to increase LC maturation biomarkers CD83, CD86, and CXCL-8 when exposed to noncytotoxic concentrations of four different titanium salts. CONCLUSION These results classify titanium salts as irritants rather than sensitizers and indicate that titanium implant-related complaints could be due to localized irritant-mediated inflammation arising from leachable agents rather than a titanium metal allergy.
Collapse
Affiliation(s)
- Charlotte T Rodrigues Neves
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Centre, Amsterdam Infection and Immunity Institute, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Sander W Spiekstra
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Centre, Amsterdam Infection and Immunity Institute, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Niels P J de Graaf
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Centre, Amsterdam Infection and Immunity Institute, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.,Department of Oral Cell Biology, Academic Centre for Dentistry (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Thomas Rustemeyer
- Department of Dermatology, Amsterdam University Medical Centre (location AMC), Amsterdam, The Netherlands
| | - Albert J Feilzer
- Department of Dental Materials Science, Academic Centre for Dentistry (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Cees J Kleverlaan
- Department of Oral Cell Biology, Academic Centre for Dentistry (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Susan Gibbs
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Centre, Amsterdam Infection and Immunity Institute, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.,Department of Oral Cell Biology, Academic Centre for Dentistry (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
47
|
Barker E, AlQobaly L, Shaikh Z, Franklin K, Moharamzadeh K. Implant Soft-Tissue Attachment Using 3D Oral Mucosal Models-A Pilot Study. Dent J (Basel) 2020; 8:E72. [PMID: 32645887 PMCID: PMC7558259 DOI: 10.3390/dj8030072] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 06/23/2020] [Accepted: 07/03/2020] [Indexed: 01/25/2023] Open
Abstract
PURPOSE The aim of this study was to investigate soft-tissue attachment to different metal, ceramic, and polymer implant surfaces using an inflamed, three-dimensional (3D), tissue-engineered, human oral mucosal model, as well as multiple-endpoint qualitative and quantitative biological approaches. METHODS Normal human oral fibroblasts, OKF6/TERT-2 keratinocytes and THP-1 monocytes were cultured, and full-thickness, 3D oral mucosal models were engineered inside tissue culture inserts. Sand-blasted and acid-etched (SLA) and machined (M) titanium-zirconium alloy (TiZr; commercially known as Roxolid; Institut Straumann AG, Switzerland), ceramic (ZrO2), and polyether ether ketone (PEEK) rods (Ø 4 mm × 8 mm) were inserted into the center of tissue-engineered oral mucosa following a Ø 4mm punch biopsy. Inflammation was simulated with addition of the lipopolysaccharide (LPS) of Escherichia coli (E. coli) and tumor necrosis factor (TNF)-alpha to the culture medium. Implant soft-tissue attachment was assessed using histology, an implant pull-test with PrestoBlue assay, and scanning electron microscopy (SEM). RESULTS Inflamed, full-thickness, 3D human oral mucosal models with inserted implants were successfully engineered and histologically characterized. The implant pull-test with PrestoBlue assay showed higher viability of the tissue that remained attached to the TiZr-SLA surface compared to the other test groups. This difference was statistically significant (p < 0.05). SEM analysis showed evidence of epithelial cell attachment on different implant surfaces. CONCLUSIONS The inflamed, 3D, oral mucosal model has the potential to be used as a suitable in vitro test system for visualization and quantification of implant soft-tissue attachment. The results of our study indicate greater soft tissue attachment to TiZr-SLA compared to TiZr-M, ceramic, and PEEK surfaces.
Collapse
Affiliation(s)
| | | | | | | | - Keyvan Moharamzadeh
- School of Clinical Dentistry, University of Sheffield, Western Bank, Sheffield S10 2TN, UK; (E.B.); (L.A.); (Z.S.); (K.F.)
| |
Collapse
|
48
|
Physiological Advantage of Phenotypic Heterogeneity in a Quorum-Sensing Population. J Indian Inst Sci 2020. [DOI: 10.1007/s41745-020-00175-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
49
|
Mikolai C, Kommerein N, Ingendoh‐Tsakmakidis A, Winkel A, Falk CS, Stiesch M. Early host–microbe interaction in a peri‐implant oral mucosa‐biofilm model. Cell Microbiol 2020; 22:e13209. [DOI: 10.1111/cmi.13209] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 02/11/2020] [Accepted: 04/15/2020] [Indexed: 12/11/2022]
Affiliation(s)
- Carina Mikolai
- Department of Prosthetic Dentistry and Biomedical Materials ScienceHannover Medical School Hannover Germany
| | - Nadine Kommerein
- Department of Prosthetic Dentistry and Biomedical Materials ScienceHannover Medical School Hannover Germany
| | | | - Andreas Winkel
- Department of Prosthetic Dentistry and Biomedical Materials ScienceHannover Medical School Hannover Germany
| | - Christine S. Falk
- Institute of Transplant ImmunologyHannover Medical School Hannover Germany
| | - Meike Stiesch
- Department of Prosthetic Dentistry and Biomedical Materials ScienceHannover Medical School Hannover Germany
| |
Collapse
|
50
|
Tabatabaei F, Moharamzadeh K, Tayebi L. Three-Dimensional In Vitro Oral Mucosa Models of Fungal and Bacterial Infections. TISSUE ENGINEERING PART B-REVIEWS 2020; 26:443-460. [PMID: 32131719 DOI: 10.1089/ten.teb.2020.0016] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Oral mucosa is the target tissue for many microorganisms involved in periodontitis and other infectious diseases affecting the oral cavity. Three-dimensional (3D) in vitro and ex vivo oral mucosa equivalents have been used for oral disease modeling and investigation of the mechanisms of oral bacterial and fungal infections. This review was conducted to analyze different studies using 3D oral mucosa models for the evaluation of the interactions of different microorganisms with oral mucosa. In this study, based on our inclusion criteria, 43 articles were selected and analyzed. Different types of 3D oral mucosa models of bacterial and fungal infections were discussed in terms of the biological system used, culture conditions, method of infection, and the biological endpoints assessed in each study. The critical analysis revealed some contradictory reports in this field of research in the literature. Challenges in recovering bacteria from oral mucosa models were further discussed, suggesting possible future directions in microbiomics, including the use of oral mucosa-on-a-chip. The potential use of these 3D tissue models for the evaluation of the effects of antiseptic agents on bacteria and oral mucosa was also addressed. This review concluded that there were many aspects that would require optimization and standardization with regard to using oral mucosal models for infection by microorganisms. Using new technologies-such as microfluidics and bioreactors-could help to reproduce some of the physiologically relevant conditions and further simulate the clinical situation. Impact statement Tissue-engineered or commercial models of the oral mucosa are very useful for the study of diseases that involve the interaction of microorganisms and oral epithelium. In this review, challenges in recovering bacteria from oral mucosa models, the potential use of these three-dimensional tissue models for the evaluation of the effects of antiseptic agents, and future directions in microbiomics are discussed.
Collapse
Affiliation(s)
- Fahimeh Tabatabaei
- School of Dentistry, Marquette University, Milwaukee, Wisconsin.,Department of Dental Biomaterials, School of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Keyvan Moharamzadeh
- School of Clinical Dentistry, University of Sheffield, Sheffield, United Kingdom
| | - Lobat Tayebi
- School of Dentistry, Marquette University, Milwaukee, Wisconsin
| |
Collapse
|