1
|
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
|
2
|
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: 0] [Impact Index Per Article: 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
|
3
|
Plaza C, Capallere C, Meyrignac C, Arcioni M, Imbert I. Development of 3D gingival in vitro models using primary gingival cells. In Vitro Cell Dev Biol Anim 2024:10.1007/s11626-024-00923-1. [PMID: 38888654 DOI: 10.1007/s11626-024-00923-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 04/29/2024] [Indexed: 06/20/2024]
Abstract
Since March 2013, animal testing for toxicity evaluation of cosmetic ingredients is banned in Europe. This directive applies to all personal care ingredients including oral ingredients. Gingival in vitro 3D models are commercially available. However, it is essential to develop "in house model" to modulate several parameters to study oral diseases, determine the toxicity of ingredients, test biocompatibility, and evaluate different formulations of cosmetic ingredients. Our expertise in tissue engineering allowed us to reconstruct human oral tissues from normal human gingival cells (fibroblasts and keratinocytes). Indeed, isolation from surgical leftover was performed to culture these gingival cells. These cells keep their endogenous capacity to proliferate allowing reconstruction of equivalent tissue close to in vivo tissue. Reconstruction of gingival epithelium, chorion equivalent, and the combination of these two tissues (full thickness) using primary gingival cells displayed all characteristics of an in vivo gingival model.
Collapse
Affiliation(s)
- Christelle Plaza
- Ashland Specialties France, 655 Route du Pin Montard, 06904, Sophia Antipolis, France.
| | - Christophe Capallere
- Ashland Specialties France, 655 Route du Pin Montard, 06904, Sophia Antipolis, France
| | - Celine Meyrignac
- Ashland Specialties France, 655 Route du Pin Montard, 06904, Sophia Antipolis, France
| | - Marianne Arcioni
- Ashland Specialties France, 655 Route du Pin Montard, 06904, Sophia Antipolis, France
| | - Isabelle Imbert
- Ashland Specialties France, 655 Route du Pin Montard, 06904, Sophia Antipolis, France
| |
Collapse
|
4
|
Mazzinelli E, Favuzzi I, Arcovito A, Castagnola R, Fratocchi G, Mordente A, Nocca G. Oral Mucosa Models to Evaluate Drug Permeability. Pharmaceutics 2023; 15:pharmaceutics15051559. [PMID: 37242801 DOI: 10.3390/pharmaceutics15051559] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 05/02/2023] [Accepted: 05/18/2023] [Indexed: 05/28/2023] Open
Abstract
Due to its numerous advantages, such as excellent drug accessibility, rapid absorption, and bypass of first-pass metabolism, the route of drug administration that involves crossing the oral mucosa is highly favored. As a result, there is significant interest in investigating the permeability of drugs through this region. The purpose of this review is to describe the various ex vivo and in vitro models used to study the permeability of conveyed and non-conveyed drugs through the oral mucosa, with a focus on the most effective models. Currently, there is a growing need for standardized models of this mucosa that can be used for developing new drug delivery systems. Oral Mucosa Equivalents (OMEs) may provide a promising future perspective as they are capable of overcoming limitations present in many existing models.
Collapse
Affiliation(s)
- Elena Mazzinelli
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Roma, Italy
| | - Ilaria Favuzzi
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Roma, Italy
| | - Alessandro Arcovito
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Roma, Italy
- Fondazione Policlinico Universitario "A. Gemelli", IRCCS, Largo Agostino Gemelli 8, 00168 Roma, Italy
| | - Raffaella Castagnola
- UOC Odontoiatria Generale e Ortodonzia, Dipartimento Scienze dell'Invecchiamento, Neurologiche, Ortopediche e della Testa Collo, Fondazione Policlinico Universitario "A. Gemelli", IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy
- Dipartimento di Testa-Collo e Organi di Senso, Università Cattolica del Sacro Cuore, Largo Agostino Gemelli 8, 00168 Rome, Italy
| | - Giorgia Fratocchi
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Roma, Italy
| | - Alvaro Mordente
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Roma, Italy
- Fondazione Policlinico Universitario "A. Gemelli", IRCCS, Largo Agostino Gemelli 8, 00168 Roma, Italy
| | - Giuseppina Nocca
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Roma, Italy
- Fondazione Policlinico Universitario "A. Gemelli", IRCCS, Largo Agostino Gemelli 8, 00168 Roma, Italy
| |
Collapse
|
5
|
Riaz A, Gidvall S, Prgomet Z, Hernandez AR, Ruzgas T, Nilsson EJ, Davies J, Valetti S. Three-Dimensional Oral Mucosal Equivalents as Models for Transmucosal Drug Permeation Studies. Pharmaceutics 2023; 15:pharmaceutics15051513. [PMID: 37242755 DOI: 10.3390/pharmaceutics15051513] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 05/12/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023] Open
Abstract
Oral transmucosal administration, where drugs are absorbed directly through the non-keratinized, lining mucosa of the mouth, represents a solution to drug delivery with several advantages. Oral mucosal equivalents (OME) developed as 3D in vitro models are of great interest since they express the correct cell differentiation and tissue architecture, simulating the in vivo conditions better than monolayer cultures or animal tissues. The aim of this work was to develop OME to be used as a membrane for drug permeation studies. We developed both full-thickness (i.e., connective plus epithelial tissue) and split-thickness (i.e., only epithelial tissue) OME using non-tumor-derived human keratinocytes OKF6 TERT-2 obtained from the floor of the mouth. All the OME developed here presented similar transepithelial electrical resistance (TEER) values, comparable to the commercial EpiOral™. Using eletriptan hydrobromide as a model drug, we found that the full-thickness OME had similar drug flux to EpiOral™ (28.8 vs. 29.6 µg/cm2/h), suggesting that the model had the same permeation barrier properties. Furthermore, full-thickness OME showed an increase in ceramide content together with a decrease in phospholipids in comparison to the monolayer culture, indicating that lipid differentiation occurred due to the tissue-engineering protocols. The split-thickness mucosal model resulted in 4-5 cell layers with basal cells still undergoing mitosis. The optimum period at the air-liquid interface for this model was twenty-one days; after longer times, signs of apoptosis appeared. Following the 3R principles, we found that the addition of Ca2+, retinoic acid, linoleic acid, epidermal growth factor and bovine pituitary extract was important but not sufficient to fully replace the fetal bovine serum. Finally, the OME models presented here offer a longer shelf-life than the pre-existing models, which paves the way for the further investigation of broader pharmaceutical applications (i.e., long-term drug exposure, effect on the keratinocytes' differentiation and inflammatory conditions, etc.).
Collapse
Affiliation(s)
- Azra Riaz
- Biomedical Science, Faculty of Health and Society, Malmö University, 205 06 Malmö, Sweden
- Biofilms-Research Center for Biointerfaces (BRCB), Malmö University, 205 06 Malmö, Sweden
| | - Sanna Gidvall
- Biomedical Science, Faculty of Health and Society, Malmö University, 205 06 Malmö, Sweden
- Biofilms-Research Center for Biointerfaces (BRCB), Malmö University, 205 06 Malmö, Sweden
| | - Zdenka Prgomet
- Section for Oral Biology and Pathology, Faculty of Odontology, Malmö University, 205 06 Malmö, Sweden
| | - Aura Rocio Hernandez
- Biomedical Science, Faculty of Health and Society, Malmö University, 205 06 Malmö, Sweden
- Biofilms-Research Center for Biointerfaces (BRCB), Malmö University, 205 06 Malmö, Sweden
| | - Tautgirdas Ruzgas
- Biomedical Science, Faculty of Health and Society, Malmö University, 205 06 Malmö, Sweden
- Biofilms-Research Center for Biointerfaces (BRCB), Malmö University, 205 06 Malmö, Sweden
| | - Emelie J Nilsson
- Biomedical Science, Faculty of Health and Society, Malmö University, 205 06 Malmö, Sweden
- Biofilms-Research Center for Biointerfaces (BRCB), Malmö University, 205 06 Malmö, Sweden
| | - Julia Davies
- Biofilms-Research Center for Biointerfaces (BRCB), Malmö University, 205 06 Malmö, Sweden
- Section for Oral Biology and Pathology, Faculty of Odontology, Malmö University, 205 06 Malmö, Sweden
| | - Sabrina Valetti
- Biomedical Science, Faculty of Health and Society, Malmö University, 205 06 Malmö, Sweden
- Biofilms-Research Center for Biointerfaces (BRCB), Malmö University, 205 06 Malmö, Sweden
| |
Collapse
|
6
|
Gould SJ, Foey AD, Salih VM. An organotypic oral mucosal infection model to study host-pathogen interactions. J Tissue Eng 2023; 14:20417314231197310. [PMID: 37873034 PMCID: PMC10590543 DOI: 10.1177/20417314231197310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 08/10/2023] [Indexed: 10/25/2023] Open
Abstract
Early in vitro oral mucosal infection models (OMMs) failed to consider the suitability of the model environment to represent the host immune response. Denture stomatitis (DS) is mediated by Candida albicans, but the role of Staphylococcus aureus remains uncertain. A collagen hydrogel-based OMM containing HaCaT and HGF cell types was developed, characterised and employed to study of tissue invasion and pro-inflammatory cytokine production in response to pathogens. Models formed a robust epithelium. Despite their inflammatory baseline, 24-h infection with C. albicans, and/or S. aureus led to tissue invasion, and significantly upregulated IL-6 and IL-8 production by OMMs when compared to the unstimulated control. No significant difference in IL-6 or IL-8 production by OMMs was observed between single and dual infections. These attributes indicate that this newly developed OMM is suitable for the study of DS and could be implemented for the wider study of oral infection.
Collapse
Affiliation(s)
- Samantha J Gould
- Department of Clinical and Biomedical Sciences, University of Exeter, Exeter, Devon, UK
| | - Andrew D Foey
- School of Biomedical Health Sciences, University of Plymouth, Plymouth, Devon, UK
| | - Vehid M Salih
- Peninsula Dental School, University of Plymouth, Plymouth, Devon, UK
| |
Collapse
|
7
|
Buccal films: A review of therapeutic opportunities, formulations & relevant evaluation approaches. J Control Release 2022; 352:1071-1092. [PMID: 36351519 DOI: 10.1016/j.jconrel.2022.10.058] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 10/26/2022] [Accepted: 10/28/2022] [Indexed: 11/19/2022]
Abstract
The potential of the mucoadhesive film technology is hard to ignore, owing to perceived superior patient acceptability versus buccal tablets, and significant therapeutic opportunities compared to conventional oral drug delivery systems, especially for those who suffer from dysphagia. In spite of this, current translation from published literature into the commercial marketplace is virtually non-existent, with no authorised mucoadhesive buccal films available in the UK and very few available in the USA. This review seeks to provide an overview of the mucoadhesive buccal film technology and identify key areas upon which to focus scientific efforts to facilitate the wider adoption of this patient-centric dosage form. Several indications and opportunities for development were identified, while discussing the patient-related factors influencing the use of these dosage forms. In addition, an overview of the technologies behind the manufacturing of these films was provided, highlighting manufacturing methods like solvent casting, hot melt extrusion, inkjet printing and three-dimensional printing. Over thirty mucoadhesive polymers were identified as being used in film formulations, with details surrounding their mucoadhesive capabilities as well as their inclusion alongside other key formulation constituents provided. Lastly, the importance of physiologically relevant in vitro evaluation methodologies was emphasised, which seek to improve in vivo correlations, potentially leading to better translation of mucoadhesive buccal films from the literature into the commercial marketplace.
Collapse
|
8
|
Edmans JG, Ollington B, Colley HE, Santocildes-Romero ME, Siim Madsen L, Hatton PV, Spain SG, Murdoch C. Electrospun patch delivery of anti-TNFα F(ab) for the treatment of inflammatory oral mucosal disease. J Control Release 2022; 350:146-157. [PMID: 35973471 DOI: 10.1016/j.jconrel.2022.08.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 08/01/2022] [Accepted: 08/10/2022] [Indexed: 10/15/2022]
Abstract
Chronic ulcerative oral mucosal inflammatory diseases, including oral lichen planus and recurrent aphthous stomatitis, are painful and highly prevalent, yet lack effective clinical management. In recent years, systemic biologic therapies, including monoclonal antibodies that block the activity of cytokines, have been increasingly used to treat a range of immune-mediated inflammatory conditions such as rheumatoid arthritis and psoriasis. The ability to deliver similar therapeutic agents locally to the oral epithelium could radically alter treatment options for oral mucosal inflammatory diseases, where pro-inflammatory cytokines, in particular tumour-necrosis factor-α (TNFα), are major drivers of pathogenesis. To address this, an electrospun dual-layer mucoadhesive patch comprising medical-grade polymers was investigated for the delivery of F(ab) biologics to the oral mucosa. A fluorescent-labelled F(ab) was incorporated into mucoadhesive membranes using electrospinning with 97% v/v ethanol as a solvent. The F(ab) was detected within the fibres in aggregates when visualised by confocal microscopy. Biotinylated F(ab) was rapidly eluted from the patch (97 ± 5% released within 3 h) without loss of antigen-binding activity. Patches applied to oral epithelium models successfully delivered the F(ab), with fluorescent F(ab) observed within the tissue and 5.1 ± 1.5% cumulative transepithelial permeation reached after 9 h. Neutralising anti-TNFα F(ab) fragments were generated from whole IgG by papain cleavage, as confirmed by SDS-PAGE, then incorporated into patches. F(ab)-containing patches had TNFα neutralising activity, as shown by the suppression of TNFα-mediated CXCL8 release from oral keratinocytes cultured as monolayers. Patches were applied to lipopolysaccharide-stimulated immune-competent oral mucosal ulcer equivalents that contained primary macrophages. Anti-TNFα patch treatment led to reduced levels of active TNFα along with a reduction in the levels of disease-implicated T-cell chemokines (CCL3, CCL5, and CXCL10) to baseline concentrations. This is the first report of an effective device for the delivery of antibody-based biologics to the oral mucosa, enabling the future development of new therapeutic strategies to treat painful conditions.
Collapse
Affiliation(s)
- Jake G Edmans
- School of Clinical Dentistry, 19 Claremont Crescent, University of Sheffield, Sheffield S10 2TA, UK; Department of Chemistry, Brook Hill, University of Sheffield, Sheffield S3 7HF, UK
| | - Bethany Ollington
- School of Clinical Dentistry, 19 Claremont Crescent, University of Sheffield, Sheffield S10 2TA, UK
| | - Helen E Colley
- School of Clinical Dentistry, 19 Claremont Crescent, University of Sheffield, Sheffield S10 2TA, UK.
| | | | - Lars Siim Madsen
- AFYX Therapeutics, Lergravsej 57, 2. tv, 2300 Copenhagen, Denmark
| | - Paul V Hatton
- School of Clinical Dentistry, 19 Claremont Crescent, University of Sheffield, Sheffield S10 2TA, UK
| | - Sebastian G Spain
- Department of Chemistry, Brook Hill, University of Sheffield, Sheffield S3 7HF, UK
| | - Craig Murdoch
- School of Clinical Dentistry, 19 Claremont Crescent, University of Sheffield, Sheffield S10 2TA, UK
| |
Collapse
|
9
|
Pimentel BNADS, Marin-Dett FH, Assis M, Barbugli PA, Longo E, Vergani CE. Antifungal Activity and Biocompatibility of α-AgVO 3, α-Ag 2WO 4, and β-Ag 2MoO 4 Using a Three-Dimensional Coculture Model of the Oral Mucosa. Front Bioeng Biotechnol 2022; 10:826123. [PMID: 35237581 PMCID: PMC8883331 DOI: 10.3389/fbioe.2022.826123] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/05/2022] [Indexed: 11/18/2022] Open
Abstract
Fungal infections have become a major concern in the medical community, especially those caused by Candida spp. Within this species, Candida albicans stands out for being an opportunistic commensal fungus that can cause superficial and invasive infections. Current antifungal therapy involves the local and/or systemic use of drugs such as azoles, polyenes, and echinocandins. These antifungals are based on highly specific target sites, and the development of resistance may occur with changes in the enzymatic pathways that serve as the drug targets. Thus, the development of new antifungal drugs is highly recommended to prevent drug resistance. The present investigation evaluated the antifungal activity of silver-containing microcrystals such as silver vanadate (α-AgVO3), silver tungstate (α-Ag2WO4), and silver molybdate (β-Ag2MoO4). In addition to having antimicrobial activity, such compounds should not cause damage to underlying tissues. Thus, to better assess the biocompatibility of new compounds, a new three-dimensional (3D) coculture model involving three cell lines was developed. The validation of the model was based on fluorescent markers and confocal laser microscopy. The biocompatibility of silver-containing microcrystals was evaluated by MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay. 3D coculture was infected with C. albicans biofilm and challenged with α-AgVO3, α-Ag2WO4, and β-Ag2MoO4. The action of microcrystals on C. albicans biofilm was evaluated by colony-forming units (CFU/ml) and LIVE/DEAD staining. In addition, production of proinflammatory cytokines interleukin 6 (IL-6), IL-8, IL-1β, and tumor necrosis factor α (TNF-α) was measured by cytometric bead array kit using flow cytometry. The 3D coculture model described here proved to be adequate to assess both the biocompatibility of the new materials and the infectious processes. Regarding the biocompatibility of the microcrystals, only α-AgVO3 (15.62 µg/ml) showed a decrease in cell viability. The antibiofilm activity of α-Ag2WO4 was similar to that of the standard drug (fluconazole). Although α-Ag2WO4 was able to induce the production of IL-6, IL-8, and IL-1β, no differences in cytokine production were observed between noninfected and infected models treated with this microcrystal. β-Ag2MoO4 inhibits the production of TNF-α in the infected model; however, it showed no antibiofilm activity. Based on the biocompatibility and antifungal findings, α-Ag2WO4 is a promising material for treating C. albicans infection.
Collapse
Affiliation(s)
- Bruna Natália Alves da Silva Pimentel
- Laboratory of Applied Microbiology, Department of Dental Materials and Prosthodontics, School of Dentistry, São Paulo State University (UNESP), Araraquara, Brazil
| | - Freddy Humberto Marin-Dett
- Department of Clinical Analysis, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, Brazil
| | - Marcelo Assis
- CDMF, LIEC, Chemistry Department, Federal University of São Carlos (UFSCar), São Carlos, Brazil
| | - Paula Aboud Barbugli
- Laboratory of Applied Microbiology, Department of Dental Materials and Prosthodontics, School of Dentistry, São Paulo State University (UNESP), Araraquara, Brazil.,Department of Clinical Analysis, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, Brazil
| | - Elson Longo
- CDMF, LIEC, Chemistry Department, Federal University of São Carlos (UFSCar), São Carlos, Brazil
| | - Carlos Eduardo Vergani
- Laboratory of Applied Microbiology, Department of Dental Materials and Prosthodontics, School of Dentistry, São Paulo State University (UNESP), Araraquara, Brazil
| |
Collapse
|
10
|
Stasko N, Cockrell AS, Kocher JF, Henson I, Emerson D, Wang Y, Smith JR, Henderson NH, Wood H, Bradrick SS, Jones T, Santander J, McNeil JG. A randomized, controlled, feasibility study of RD-X19 in subjects with mild-to-moderate COVID-19 in the outpatient setting. Clin Transl Sci 2022; 15:1291-1303. [PMID: 35137532 PMCID: PMC9099126 DOI: 10.1111/cts.13249] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 01/25/2022] [Accepted: 01/26/2022] [Indexed: 11/28/2022] Open
Abstract
The RD-X19 is an investigational, handheld medical device precisely engineered to emit blue light through the oral cavity to target the oropharynx and surrounding tissues. At doses shown to be non-cytotoxic in an in vitro 3-dimensional human epithelial tissue model, the monochromatic visible light delivered by RD-X19 results in light-initiated expression of immune stimulating cytokines IL-1α and IL-1β, with corresponding inhibition of SARS-CoV-2 replication. A single exposure of 425 nm blue light at 60 J/cm2 led to >99% reductions against all SARS-CoV-2 strains tested in vitro, including the more transmissible (Alpha) and immune evasive (Beta) variants. These preclinical findings along with other studies led to a randomized, double-blind, sham-controlled early feasibility study using the investigational device as a treatment for outpatients with mild to moderate COVID-19. The study enrolled 31 subjects with a positive SARS-CoV-2 antigen test and at least two moderate COVID-19 signs and symptoms at baseline. Subjects were randomized 2:1 (RD-X19: sham) and treated twice daily for four days. Efficacy outcome measures included assessments of SARS-CoV-2 saliva viral load and clinical assessments of COVID-19. There were no local application site reactions and no device-related adverse events. At the end of study (Day 8), the mean change in log10 viral load was -3.29 for RD-X19 and -1.81 for sham, demonstrating a treatment benefit of -1.48 logs [95% confidence internal (CI), -2.88 to -0.071, nominal p=0.040]. Among the clinical outcome measures, differences between RD-X19 and sham were also observed, with a 57-hour reduction of median time to sustained resolution of COVID-19 signs and symptoms (log rank test, nominal p=0.044).
Collapse
Affiliation(s)
| | | | | | | | | | - Ye Wang
- Symbio, LLC, Port Jefferson, New York, USA
| | | | | | | | | | - Terry Jones
- J&S Studies, Inc., College Station, Texas, USA
| | | | | |
Collapse
|
11
|
Sakulpaptong W, Clairmonte IA, Blackstone BN, Leblebicioglu B, Powell HM. 3D engineered human gingiva fabricated with electrospun collagen scaffolds provides a platform for in vitro analysis of gingival seal to abutment materials. PLoS One 2022; 17:e0263083. [PMID: 35113915 PMCID: PMC8812907 DOI: 10.1371/journal.pone.0263083] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 01/11/2022] [Indexed: 11/28/2022] Open
Abstract
In order to advance models of human oral mucosa towards routine use, these models must faithfully mimic the native tissue structure while also being scalable and cost efficient. The goal of this study was to develop a low-cost, keratinized human gingival model with high fidelity to human attached gingiva and demonstrate its utility for studying the implant-tissue interface. Primary human gingival fibroblasts (HGF) and keratinocytes (HGK) were isolated from clinically healthy gingival biopsies. Four matrices, electrospun collagen (ES), decellularized dermis (DD), type I collagen gels (Gel) and released type I collagen gels (Gel-R)) were tested to engineer lamina propria and gingiva. HGF viability was similar in all matrices except for Gel-R, which was significantly decreased. Cell penetration was largely limited to the top layers of all matrices. Histomorphometrically, engineered human gingiva was found to have similar appearance to the native normal human gingiva except absence of rete pegs. Immunohistochemical staining for cell phenotype, differentiation and extracellular matrix composition and organization within 3D engineered gingiva made with electrospun collagen was mostly in agreement with normal gingival tissue staining. Additionally, five types of dental material posts (5-mm diameter x 3-mm height) with different surface characteristics were used [machined titanium, SLA (sandblasted-acid etched) titanium, TiN-coated (titanium nitride-coated) titanium, ceramic, and PEEK (Polyetheretherketone) to investigate peri-implant soft tissue attachment studied by histology and SEM. Engineered epithelial and stromal tissue migration to the implant-gingival tissue interface was observed in machined, SLA, ceramic, and PEEK groups, while TiN was lacking attachment. Taken together, the results suggest that electrospun collagen scaffolds provide a scalable, reproducible and cost-effective lamina propria and 3D engineered gingiva that can be used to explore biomaterial-soft tissue interface.
Collapse
Affiliation(s)
- Wichurat Sakulpaptong
- Division of Periodontology, College of Dentistry, The Ohio State University, Columbus, OH, United States of America
- Faculty of Dentistry, Department of Oral Medicine and Periodontology, Mahidol University, Bangkok, Thailand
| | - Isabelle A. Clairmonte
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH, United States of America
| | - Britani N. Blackstone
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH, United States of America
| | - Binnaz Leblebicioglu
- Division of Periodontology, College of Dentistry, The Ohio State University, Columbus, OH, United States of America
| | - Heather M. Powell
- Department of Materials Science and Engineering, The Ohio State University, Columbus, OH, United States of America
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, OH, United States of America
- Research Department, Shriners Children’s Ohio, Dayton, Ohio, United States of America
- * E-mail:
| |
Collapse
|
12
|
Helena Macedo M, Baião A, Pinto S, Barros AS, Almeida H, Almeida A, das Neves J, Sarmento B. Mucus-producing 3D cell culture models. Adv Drug Deliv Rev 2021; 178:113993. [PMID: 34619286 DOI: 10.1016/j.addr.2021.113993] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 08/23/2021] [Accepted: 09/29/2021] [Indexed: 10/20/2022]
Abstract
In vitro cell-based models have been used for a long time since they are normally easily obtained and have an advantageous cost-benefit. Besides, they can serve a variety of ends, from studying drug absorption and metabolism to disease modeling. However, some in vitro models are too simplistic, not accurately representing the living tissues. It has been shown, mainly in the last years, that fully mimicking a tissue composition and architecture can be paramount for cellular behavior and, consequently, for the outcomes of the studies using such models. Because of this, 3D in vitro cell models have been gaining much attention, since they are able to better replicate the in vivo environment. In this review we focus on 3D models that contain mucus-producing cells, as mucus can play a pivotal role in drug absorption. Being frequently overlooked, this viscous fluid can have an impact on drug delivery. Thus, the aim of this review is to understand to which extent can mucus affect mucosal drug delivery and to provide a state-of-the-art report on the existing 3D cell-based mucus models.
Collapse
|
13
|
Morgan D, Berggren KL, Spiess CD, Smith HM, Tejwani A, Weir SJ, Lominska CE, Thomas SM, Gan GN. Mitogen-activated protein kinase-activated protein kinase-2 (MK2) and its role in cell survival, inflammatory signaling, and migration in promoting cancer. Mol Carcinog 2021; 61:173-199. [PMID: 34559922 DOI: 10.1002/mc.23348] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 08/25/2021] [Accepted: 08/27/2021] [Indexed: 12/19/2022]
Abstract
Cancer and the immune system share an intimate relationship. Chronic inflammation increases the risk of cancer occurrence and can also drive inflammatory mediators into the tumor microenvironment enhancing tumor growth and survival. The p38 MAPK pathway is activated both acutely and chronically by stress, inflammatory chemokines, chronic inflammatory conditions, and cancer. These properties have led to extensive efforts to find effective drugs targeting p38, which have been unsuccessful. The immediate downstream serine/threonine kinase and substrate of p38 MAPK, mitogen-activated-protein-kinase-activated-protein-kinase-2 (MK2) protects cells against stressors by regulating the DNA damage response, transcription, protein and messenger RNA stability, and motility. The phosphorylation of downstream substrates by MK2 increases inflammatory cytokine production, drives an immune response, and contributes to wound healing. By binding directly to p38 MAPK, MK2 is responsible for the export of p38 MAPK from the nucleus which gives MK2 properties that make it unique among the large number of p38 MAPK substrates. Many of the substrates of both p38 MAPK and MK2 are separated between the cytosol and nucleus and interfering with MK2 and altering this intracellular translocation has implications for the actions of both p38 MAPK and MK2. The inhibition of MK2 has shown promise in combination with both chemotherapy and radiotherapy as a method for controlling cancer growth and metastasis in a variety of cancers. Whereas the current data are encouraging the field requires the development of selective and well tolerated drugs to target MK2 and a better understanding of its effects for effective clinical use.
Collapse
Affiliation(s)
- Deri Morgan
- Department of Radiation Oncology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Kiersten L Berggren
- Department of Internal Medicine, Division of Medical Oncology, Section of Radiation Oncology, UNM School of Medicine, The University of New Mexico, Albuquerque, New Mexico, USA
| | - Colby D Spiess
- Department of Radiation Oncology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Hannah M Smith
- Department of Radiation Oncology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Ajay Tejwani
- Department of Radiation Oncology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Scott J Weir
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Christopher E Lominska
- Department of Radiation Oncology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Sufi M Thomas
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas, USA.,Department of Otolaryngology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Gregory N Gan
- Department of Radiation Oncology, University of Kansas Medical Center, Kansas City, Kansas, USA.,Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
14
|
Ollington B, Colley HE, Murdoch C. Immunoresponsive Tissue-Engineered Oral Mucosal Equivalents Containing Macrophages. Tissue Eng Part C Methods 2021; 27:462-471. [PMID: 34210153 PMCID: PMC8403184 DOI: 10.1089/ten.tec.2021.0124] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Macrophages play a key role in orchestrating the host immune response toward invading organisms or non-self molecules in the oral mucosa. Three-dimensional (3D) oral mucosal equivalents (OME) containing oral fibroblasts and keratinocytes are used extensively to mimic the human oral mucosa where they have been employed to examine innate immune responses to both bacterial and fungal pathogens as well as to biomaterials. Although the presence of immune cells is critical in generating an immune response, very few studies have incorporated leukocytes into OME, and to date, none have contained primary human macrophages. In this study, we report the generation of an immunocompetent OME to investigate immune responses toward bacterial challenge. Primary human monocyte-derived macrophages (MDM) were as responsive to bacterial lipopolysaccharide (LPS) challenge when cultured within a 3D hydrogel in terms of proinflammatory cytokine (IL-6, CXCL8, and TNF-α) gene expression and protein secretion compared with culture as two-dimensional monolayers. MDM were incorporated into a type 1 collagen hydrogel along with oral fibroblasts and the apical surface seeded with oral keratinocytes to generate an MDM-containing OME. Full-thickness MDM-OME displayed a stratified squamous epithelium and a fibroblast-populated connective tissue containing CD68-positive MDM that could be readily isolated to a single-cell population for further analysis by collagenase treatment followed by flow cytometry. When stimulated with LPS, MDM-OME responded with increased proinflammatory cytokine secretion, most notably for TNF-α that increased 12-fold when compared with OME alone. Moreover, this proinflammatory response was inhibited by pretreatment with dexamethasone, showing that MDM-OME are also amenable to drug treatment. Dual-labeled immunofluorescence confocal microscopy revealed that MDM were the sole source of TNF-α production within MDM-OME. These data show functional activity of MDM-OME and illustrate their usefulness for investigations aimed at monitoring the immune response of the oral mucosa to pathogens, biomaterials, and for tissue toxicity and anti-inflammatory drug delivery studies.
Collapse
Affiliation(s)
- Bethany Ollington
- School of Clinical Dentistry, University of Sheffield, Sheffield, United Kingdom
| | - Helen E Colley
- School of Clinical Dentistry, University of Sheffield, Sheffield, United Kingdom
| | - Craig Murdoch
- School of Clinical Dentistry, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
15
|
Cytokine-Mediated Inflammation in the Oral Cavity and Its Effect on Lipid Nanocarriers. NANOMATERIALS 2021; 11:nano11051330. [PMID: 34070004 PMCID: PMC8157841 DOI: 10.3390/nano11051330] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/06/2021] [Accepted: 05/12/2021] [Indexed: 01/17/2023]
Abstract
Topical drug administration to the oral mucosa proves to be a promising treatment alternative for inflammatory diseases. However, disease-related changes in the cell barrier must be considered when developing such delivery systems. This study aimed at investigating the changes in the lining mucosa caused by inflammation and evaluating the consequences on drug delivery systems such as nanostructured lipid carriers (NLC). For this, TR146 cells were treated with inflammatory cytokines and bacterial components. Cell viability and integrity, reactive oxygen species (ROS), and interleukin (IL)-8 release were used as endpoints to assess inflammation. Translocation of phosphatidylserine, cytoskeletal arrangement, opening of desmosomes, and cell proliferation were examined. Transport studies with NLC were performed considering active and passive pathways. The results showed that IL-1ß and tumor necrosis factor α induced inflammation by increasing IL-8 and ROS production (22-fold and 2-fold). Morphologically, loss of cell–cell connections and formation of stress fibers and hyperplasia were observed. The charge of the cell membrane shifted from neutral to negative, which increased the absorption of NLC due to the repulsive interactions between the hydrophobic negative particles and the cell membrane on the one hand, and interactions with lipophilic membrane proteins such as caveolin on the other.
Collapse
|
16
|
d'Enfert C, Kaune AK, Alaban LR, Chakraborty S, Cole N, Delavy M, Kosmala D, Marsaux B, Fróis-Martins R, Morelli M, Rosati D, Valentine M, Xie Z, Emritloll Y, Warn PA, Bequet F, Bougnoux ME, Bornes S, Gresnigt MS, Hube B, Jacobsen ID, Legrand M, Leibundgut-Landmann S, Manichanh C, Munro CA, Netea MG, Queiroz K, Roget K, Thomas V, Thoral C, Van den Abbeele P, Walker AW, Brown AJP. The impact of the Fungus-Host-Microbiota interplay upon Candida albicans infections: current knowledge and new perspectives. FEMS Microbiol Rev 2021; 45:fuaa060. [PMID: 33232448 PMCID: PMC8100220 DOI: 10.1093/femsre/fuaa060] [Citation(s) in RCA: 142] [Impact Index Per Article: 47.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 11/18/2020] [Indexed: 12/11/2022] Open
Abstract
Candida albicans is a major fungal pathogen of humans. It exists as a commensal in the oral cavity, gut or genital tract of most individuals, constrained by the local microbiota, epithelial barriers and immune defences. Their perturbation can lead to fungal outgrowth and the development of mucosal infections such as oropharyngeal or vulvovaginal candidiasis, and patients with compromised immunity are susceptible to life-threatening systemic infections. The importance of the interplay between fungus, host and microbiota in driving the transition from C. albicans commensalism to pathogenicity is widely appreciated. However, the complexity of these interactions, and the significant impact of fungal, host and microbiota variability upon disease severity and outcome, are less well understood. Therefore, we summarise the features of the fungus that promote infection, and how genetic variation between clinical isolates influences pathogenicity. We discuss antifungal immunity, how this differs between mucosae, and how individual variation influences a person's susceptibility to infection. Also, we describe factors that influence the composition of gut, oral and vaginal microbiotas, and how these affect fungal colonisation and antifungal immunity. We argue that a detailed understanding of these variables, which underlie fungal-host-microbiota interactions, will present opportunities for directed antifungal therapies that benefit vulnerable patients.
Collapse
Affiliation(s)
- Christophe d'Enfert
- Unité Biologie et Pathogénicité Fongiques, Département de Mycologie, Institut Pasteur, USC 2019 INRA, 25, rue du Docteur Roux, 75015 Paris, France
| | - Ann-Kristin Kaune
- Aberdeen Fungal Group, Institute of Medical Sciences, University of Aberdeen, Ashgrove Road West, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Leovigildo-Rey Alaban
- BIOASTER Microbiology Technology Institute, 40 avenue Tony Garnier, 69007 Lyon, France
- Université de Paris, Sorbonne Paris Cité, 25, rue du Docteur Roux, 75015 Paris, France
| | - Sayoni Chakraborty
- Microbial Immunology Research Group, Emmy Noether Junior Research Group Adaptive Pathogenicity Strategies, and the Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Beutenbergstraße 11a, 07745 Jena, Germany
- Institute of Microbiology, Friedrich Schiller University, Neugasse 25, 07743 Jena, Germany
| | - Nathaniel Cole
- Gut Microbiology Group, Rowett Institute, University of Aberdeen, Ashgrove Road West, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Margot Delavy
- Unité Biologie et Pathogénicité Fongiques, Département de Mycologie, Institut Pasteur, USC 2019 INRA, 25, rue du Docteur Roux, 75015 Paris, France
- Université de Paris, Sorbonne Paris Cité, 25, rue du Docteur Roux, 75015 Paris, France
| | - Daria Kosmala
- Unité Biologie et Pathogénicité Fongiques, Département de Mycologie, Institut Pasteur, USC 2019 INRA, 25, rue du Docteur Roux, 75015 Paris, France
- Université de Paris, Sorbonne Paris Cité, 25, rue du Docteur Roux, 75015 Paris, France
| | - Benoît Marsaux
- ProDigest BV, Technologiepark 94, B-9052 Gent, Belgium
- Center for Microbial Ecology and Technology (CMET), Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, Coupure Links, 9000 Ghent, Belgium
| | - Ricardo Fróis-Martins
- Immunology Section, Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 266a, Zurich 8057, Switzerland
- Institute of Experimental Immunology, University of Zurich, Winterthurerstrasse 190, Zürich 8057, Switzerland
| | - Moran Morelli
- Mimetas, Biopartner Building 2, J.H. Oortweg 19, 2333 CH Leiden, The Netherlands
| | - Diletta Rosati
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Geert Grooteplein 28, 6525 GA Nijmegen, The Netherlands
| | - Marisa Valentine
- Microbial Immunology Research Group, Emmy Noether Junior Research Group Adaptive Pathogenicity Strategies, and the Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Beutenbergstraße 11a, 07745 Jena, Germany
| | - Zixuan Xie
- Gut Microbiome Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119–129, 08035 Barcelona, Spain
| | - Yoan Emritloll
- Unité Biologie et Pathogénicité Fongiques, Département de Mycologie, Institut Pasteur, USC 2019 INRA, 25, rue du Docteur Roux, 75015 Paris, France
| | - Peter A Warn
- Magic Bullet Consulting, Biddlecombe House, Ugbrook, Chudleigh Devon, TQ130AD, UK
| | - Frédéric Bequet
- BIOASTER Microbiology Technology Institute, 40 avenue Tony Garnier, 69007 Lyon, France
| | - Marie-Elisabeth Bougnoux
- Unité Biologie et Pathogénicité Fongiques, Département de Mycologie, Institut Pasteur, USC 2019 INRA, 25, rue du Docteur Roux, 75015 Paris, France
| | - Stephanie Bornes
- Université Clermont Auvergne, INRAE, VetAgro Sup, UMRF0545, 20 Côte de Reyne, 15000 Aurillac, France
| | - Mark S Gresnigt
- Microbial Immunology Research Group, Emmy Noether Junior Research Group Adaptive Pathogenicity Strategies, and the Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Beutenbergstraße 11a, 07745 Jena, Germany
| | - Bernhard Hube
- Microbial Immunology Research Group, Emmy Noether Junior Research Group Adaptive Pathogenicity Strategies, and the Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Beutenbergstraße 11a, 07745 Jena, Germany
| | - Ilse D Jacobsen
- Microbial Immunology Research Group, Emmy Noether Junior Research Group Adaptive Pathogenicity Strategies, and the Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Beutenbergstraße 11a, 07745 Jena, Germany
| | - Mélanie Legrand
- Unité Biologie et Pathogénicité Fongiques, Département de Mycologie, Institut Pasteur, USC 2019 INRA, 25, rue du Docteur Roux, 75015 Paris, France
| | - Salomé Leibundgut-Landmann
- Immunology Section, Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 266a, Zurich 8057, Switzerland
- Institute of Experimental Immunology, University of Zurich, Winterthurerstrasse 190, Zürich 8057, Switzerland
| | - Chaysavanh Manichanh
- Gut Microbiome Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119–129, 08035 Barcelona, Spain
| | - Carol A Munro
- Aberdeen Fungal Group, Institute of Medical Sciences, University of Aberdeen, Ashgrove Road West, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Geert Grooteplein 28, 6525 GA Nijmegen, The Netherlands
| | - Karla Queiroz
- Mimetas, Biopartner Building 2, J.H. Oortweg 19, 2333 CH Leiden, The Netherlands
| | - Karine Roget
- NEXBIOME Therapeutics, 22 allée Alan Turing, 63000 Clermont-Ferrand, France
| | - Vincent Thomas
- BIOASTER Microbiology Technology Institute, 40 avenue Tony Garnier, 69007 Lyon, France
| | - Claudia Thoral
- NEXBIOME Therapeutics, 22 allée Alan Turing, 63000 Clermont-Ferrand, France
| | | | - Alan W Walker
- Gut Microbiology Group, Rowett Institute, University of Aberdeen, Ashgrove Road West, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Alistair J P Brown
- MRC Centre for Medical Mycology, Department of Biosciences, University of Exeter, Geoffrey Pope Building, Stocker Road, Exeter EX4 4QD, UK
| |
Collapse
|
17
|
Gomez-Casado C, Sanchez-Solares J, Izquierdo E, Díaz-Perales A, Barber D, Escribese MM. Oral Mucosa as a Potential Site for Diagnosis and Treatment of Allergic and Autoimmune Diseases. Foods 2021; 10:970. [PMID: 33925074 PMCID: PMC8146604 DOI: 10.3390/foods10050970] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 04/23/2021] [Accepted: 04/26/2021] [Indexed: 12/15/2022] Open
Abstract
Most prevalent food allergies during early childhood are caused by foods with a high allergenic protein content, such as milk, egg, nuts, or fish. In older subjects, some respiratory allergies progressively lead to food-induced allergic reactions, which can be severe, such as urticaria or asthma. Oral mucosa remodeling has been recently proven to be a feature of severe allergic phenotypes and autoimmune diseases. This remodeling process includes epithelial barrier disruption and the release of inflammatory signals. Although little is known about the immune processes taking place in the oral mucosa, there are a few reports describing the oral mucosa-associated immune system. In this review, we will provide an overview of the recent knowledge about the role of the oral mucosa in food-induced allergic reactions, as well as in severe respiratory allergies or food-induced autoimmune diseases, such as celiac disease.
Collapse
Affiliation(s)
- Cristina Gomez-Casado
- Institute of Applied Molecular Medicine, Department of Basic Medical Sciences, Faculty of Medicine, San Pablo CEU University, 28003 Madrid, Spain; (J.S.-S.); (E.I.); (D.B.); (M.M.E.)
| | - Javier Sanchez-Solares
- Institute of Applied Molecular Medicine, Department of Basic Medical Sciences, Faculty of Medicine, San Pablo CEU University, 28003 Madrid, Spain; (J.S.-S.); (E.I.); (D.B.); (M.M.E.)
| | - Elena Izquierdo
- Institute of Applied Molecular Medicine, Department of Basic Medical Sciences, Faculty of Medicine, San Pablo CEU University, 28003 Madrid, Spain; (J.S.-S.); (E.I.); (D.B.); (M.M.E.)
| | - Araceli Díaz-Perales
- Center of Plant Biotechnology and Genomics, Technical University of Madrid, 28040 Madrid, Spain;
| | - Domingo Barber
- Institute of Applied Molecular Medicine, Department of Basic Medical Sciences, Faculty of Medicine, San Pablo CEU University, 28003 Madrid, Spain; (J.S.-S.); (E.I.); (D.B.); (M.M.E.)
| | - María M. Escribese
- Institute of Applied Molecular Medicine, Department of Basic Medical Sciences, Faculty of Medicine, San Pablo CEU University, 28003 Madrid, Spain; (J.S.-S.); (E.I.); (D.B.); (M.M.E.)
| |
Collapse
|
18
|
Said Z, Murdoch C, Hansen J, Siim Madsen L, Colley HE. Corticosteroid delivery using oral mucosa equivalents for the treatment of inflammatory mucosal diseases. Eur J Oral Sci 2021; 129:e12761. [PMID: 33645844 PMCID: PMC8048628 DOI: 10.1111/eos.12761] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 11/24/2020] [Accepted: 11/25/2020] [Indexed: 12/14/2022]
Abstract
Oral lichen planus (OLP) is an immune‐mediated disease of the oral mucosa with idiopathic aetiology. It is frequently treated with topical corticosteroids (applied as gels, mouthwashes, or sprays); however, the mucosal exposure times of topical corticosteroids are short because of removal by the constant flow of saliva and mechanical forces. In this study we used cell monolayers, as well as oral mucosal equivalents (OMEs) containing activated T‐cells, to examine corticosteroid potency and delivery of clobetasol‐17‐propionate from a novel electrospun mucoadhesive patch. The OMEs displayed tight junctions, desmosomes, hemidesmosomes, and an efficient permeability barrier. Following application of corticosteroids to cells cultured as monolayers, the degree of cytotoxicity measured correlated to the level of potency recognized for each corticosteroid; by contrast, OMEs were largely unaffected by corticosteroid treatment. Permeation of clobetasol‐17‐propionate into and through the OMEs was time‐ and dose‐dependent, regardless of whether this corticosteroid was delivered in liquid form or from a mucoadhesive patch, and both liquid‐ and patch‐delivered clobetasol‐17‐propionate significantly reduced the secretion of interleukin‐2 by activated T‐cells. This study confirms that OMEs are more suitable models than cell monolayers for evaluating toxicity and drug delivery. After topical exposure, clobetasol‐17‐propionate accumulated in OMEs at a higher level than betamethasone‐17‐valerate and hydrocortisone‐17‐valerate, and exerted its immunosuppressive actions following application via the patch delivery system, highlighting the efficacy of this mode of drug delivery to treat OLP.
Collapse
Affiliation(s)
- Zulfahmi Said
- School of Clinical Dentistry, University of Sheffield, Sheffield, UK.,Faculty of Dentistry, Universiti Sains Islam Malaysia, Kuala Lumpur, Malaysia
| | - Craig Murdoch
- School of Clinical Dentistry, University of Sheffield, Sheffield, UK
| | | | | | - Helen E Colley
- School of Clinical Dentistry, University of Sheffield, Sheffield, UK
| |
Collapse
|
19
|
Wang S, Zuo A, Guo J. Types and evaluation of in vitro penetration models for buccal mucosal delivery. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2020.102122] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
20
|
Klausner M, Handa Y, Aizawa S. In vitro three-dimensional organotypic culture models of the oral mucosa. In Vitro Cell Dev Biol Anim 2021; 57:148-159. [PMID: 33447968 PMCID: PMC7808300 DOI: 10.1007/s11626-020-00539-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 12/14/2020] [Indexed: 11/26/2022]
Abstract
Three-dimensional, organotypic models of the oral mucosa have been developed to study a wide variety of phenomena occurring in the oral cavity. Although a number of models have been developed in academic research labs, only a few models have been commercialized. Models from academic groups offer a broader range of phenotypes while the commercial models are more focused on the oral and gingival mucosa. The commercialized models are manufactured under highly controlled conditions and meet the requirements of quality standards, which leads to high levels of reproducibility. These in vitro models have been used to evaluate the irritancy of oral care products such as toothpastes, mouthwashes, and mucoadhesives. The effects of cigarette smoke on oral cavity tissues have been studied and compared to those of e-cigarettes. Oral tissue models have facilitated investigation of the mechanisms of oral mucositis and oral candidiasis and have been used to examine transbuccal drug delivery rates and the absorption of nanoparticles. Infection studies have investigated the effects of HIV-1 along with the effects of commensal and pathogenic bacteria. More recently, a differentiated oral tissue model has been shown to express the ACE2 receptor, which is known to be important for the receptor-mediated entry of the SARS-CoV-2 coronavirus into human cells and tissues. Hence, oral mucosal models may find application in determining whether viral infection of the oral mucosa is possible and whether such infection has implications vis-a-vis the current COVID-19 pandemic. As is apparent, these models are used in a broad variety of applications and often offer advantages versus animal models in terms of reproducibility, avoiding species extrapolation, and the ethical concerns related to human and animal experimentation. The goals of this paper are to review commercially available models of the human buccal and gingival mucosa and highlight their use to gain a better understanding of a broad range of phenomena affecting tissues in the oral cavity.
Collapse
Affiliation(s)
| | - Yuki Handa
- Kurabo Industries Ltd, Neyagawa, Osaka, Japan
| | | |
Collapse
|
21
|
Mountcastle SE, Cox SC, Sammons RL, Jabbari S, Shelton RM, Kuehne SA. A review of co-culture models to study the oral microenvironment and disease. J Oral Microbiol 2020; 12:1773122. [PMID: 32922679 PMCID: PMC7448840 DOI: 10.1080/20002297.2020.1773122] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 04/25/2020] [Accepted: 05/19/2020] [Indexed: 12/19/2022] Open
Abstract
Co-cultures allow for the study of cell-cell interactions between different eukaryotic species or with bacteria. Such an approach has enabled researchers to more closely mimic complex tissue structures. This review is focused on co-culture systems modelling the oral cavity, which have been used to evaluate this unique cellular environment and understand disease progression. Over time, these systems have developed significantly from simple 2D eukaryotic cultures and planktonic bacteria to more complex 3D tissue engineered structures and biofilms. Careful selection and design of the co-culture along with critical parameters, such as seeding density and choice of analysis method, have resulted in several advances. This review provides a comparison of existing co-culture systems for the oral environment, with emphasis on progression of 3D models and the opportunity to harness techniques from other fields to improve current methods. While filling a gap in navigating this literature, this review ultimately supports the development of this vital technique in the field of oral biology.
Collapse
Affiliation(s)
- Sophie E Mountcastle
- EPSRC Centre for Doctoral Training in Physical Sciences for Health, University of Birmingham, Birmingham, UK
- School of Dentistry, University of Birmingham, Birmingham, UK
| | - Sophie C Cox
- School of Chemical Engineering, University of Birmingham, Birmingham, UK
| | | | - Sara Jabbari
- School of Mathematics, University of Birmingham, Birmingham, UK
- Institute of Microbiology and Infection, University of Birmingham, Birmingham, UK
| | | | - Sarah A Kuehne
- School of Dentistry, University of Birmingham, Birmingham, UK
- Institute of Microbiology and Infection, University of Birmingham, Birmingham, UK
| |
Collapse
|
22
|
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: 3.0] [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
|
23
|
Novel and revisited approaches in nanoparticle systems for buccal drug delivery. J Control Release 2020; 320:125-141. [DOI: 10.1016/j.jconrel.2020.01.006] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 01/02/2020] [Accepted: 01/04/2020] [Indexed: 12/15/2022]
|
24
|
Bierbaumer L, Schwarze UY, Gruber R, Neuhaus W. Cell culture models of oral mucosal barriers: A review with a focus on applications, culture conditions and barrier properties. Tissue Barriers 2018; 6:1479568. [PMID: 30252599 PMCID: PMC6389128 DOI: 10.1080/21688370.2018.1479568] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Understanding the function of oral mucosal epithelial barriers is essential for a plethora of research fields such as tumor biology, inflammation and infection diseases, microbiomics, pharmacology, drug delivery, dental and biomarker research. The barrier properties are comprised by a physical, a transport and a metabolic barrier, and all these barrier components play pivotal roles in the communication between saliva and blood. The sum of all epithelia of the oral cavity and salivary glands is defined as the blood-saliva barrier. The functionality of the barrier is regulated by its microenvironment and often altered during diseases. A huge array of cell culture models have been developed to mimic specific parts of the blood-saliva barrier, but no ultimate standard in vitro models have been established. This review provides a comprehensive overview about developed in vitro models of oral mucosal barriers, their applications, various cultivation protocols and corresponding barrier properties.
Collapse
Affiliation(s)
- Lisa Bierbaumer
- a Competence Unit Molecular Diagnostics, Center Health and Bioresources, Austrian Institute of Technology (AIT) GmbH , Vienna , Austria
| | - Uwe Yacine Schwarze
- b Department of Oral Biology , School of Dentistry, Medical University of Vienna , Vienna , Austria.,c Austrian Cluster for Tissue Regeneration , Vienna , Austria
| | - Reinhard Gruber
- b Department of Oral Biology , School of Dentistry, Medical University of Vienna , Vienna , Austria.,c Austrian Cluster for Tissue Regeneration , Vienna , Austria.,d Department of Periodontology , School of Dental Medicine, University of Bern , Bern , Switzerland
| | - Winfried Neuhaus
- a Competence Unit Molecular Diagnostics, Center Health and Bioresources, Austrian Institute of Technology (AIT) GmbH , Vienna , Austria
| |
Collapse
|
25
|
Buchs R, Lehner B, Meuwly P, Schnyder B. Host-Pathogen Interaction Reconstituted in Three-Dimensional Cocultures of Mucosa and Candida albicans. Tissue Eng Part C Methods 2018; 24:412-417. [PMID: 29901432 DOI: 10.1089/ten.tec.2018.0042] [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: 11/12/2022] Open
Abstract
Candida albicans frequently causes recurrent intimal infectious disease (ID). This demands the treatment of multiple phases of the infection. The objective of this study was to uncover the host-pathogen interaction using two-dimensional (2D) epithelium cell-barrier and three-dimensional (3D) subepithelium tissue cells of human mucosa. The 2D cell cultures assessed C. albicans adhesion. Addition of the antifungal drug Fluconazol did not inhibit the adhesion, despite its pathogen growth inhibition (minimal inhibitory concentration value 0.08 μg/mL). A 3D tissue was engineered in multitranswells by placing human fibroblast cultures on a thick porous scaffold. This contained the yeast placed in the top compartment and prevented passive penetration. After 28 h, the pathogen transmigrated the barrier and was collected in the bottom compartment. A change in pathogen morphology was observed where hypha formed and grew to be 231 μm long after 28 h. The hypha was thus long enough to cross the 200 μm thick 3D tissue. The 3D infection was inhibited by addition of Fluconazol (0.08 μg/mL), confirming that penetration is dependent on pathogen growth. In conclusion, ID was reconstituted step-by-step on 2D epithelium surface and in 3D connective tissue of human mucosa. Fluconazol growth-inhibition of the pathogen C. albicans was confirmed in the 3D tissue. We thus propose that this ID in vitro test is suitable for the identification and characterization of new treatments against C. albicans.
Collapse
Affiliation(s)
- Romina Buchs
- 1 School of Engineering, Institute of Life Technologies, University of Applied Sciences , HES-SO Valais/Wallis, Sion, Switzerland
| | - Bruno Lehner
- 1 School of Engineering, Institute of Life Technologies, University of Applied Sciences , HES-SO Valais/Wallis, Sion, Switzerland
| | | | - Bruno Schnyder
- 1 School of Engineering, Institute of Life Technologies, University of Applied Sciences , HES-SO Valais/Wallis, Sion, Switzerland
| |
Collapse
|
26
|
Hassanzadeh P, Atyabi F, Dinarvand R. Tissue engineering: Still facing a long way ahead. J Control Release 2018; 279:181-197. [DOI: 10.1016/j.jconrel.2018.04.024] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 04/09/2018] [Accepted: 04/11/2018] [Indexed: 02/07/2023]
|
27
|
Walladbegi J, Smith SA, Grayson AK, Murdoch C, Jontell M, Colley HE. Cooling of the oral mucosa to prevent adverse effects of chemotherapeutic agents: An in vitro study. J Oral Pathol Med 2018; 47:477-483. [PMID: 29469972 DOI: 10.1111/jop.12696] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/16/2018] [Indexed: 12/19/2022]
Abstract
BACKGROUND The cytotoxic effect of chemotherapeutic agents to the oral mucosa, as a side effect of cancer treatment, is a major problem. Cooling the oral mucosa using ice chips in conjunction with chemotherapy is known to reduce the severity of oral mucositis. However, although the use of ice chips is of clinical value, this method of cooling has inherent problems including discomfort for the patient, non-uniformity and fluctuations in cooling temperature throughout the oral cavity. Furthermore, despite being used clinically, it is not known what reduction in temperature is required to prevent oral mucositis. The aim of this study was therefore to determine in vitro if the cytotoxic effect of 5-fluorouracil (5-FU) on the oral mucosa could be reduced by lowering the temperature during chemotherapeutic treatment. METHODS Tissue-engineered oral mucosal (TEOM) models were incubated at 20, 25, 30 or 35°C for 30 minutes followed by exposure to a clinically relevant concentration of 5-FU (162 μg/mL) for 2 hours and compared with untreated models (35°C). Cell viability and inflammatory cytokine production (IL-6 and TNF-α) were measured using PrestoBlue® and ELISA, respectively. RESULTS TEOM models incubated at 20°C showed an increased cell viability and had a reduced IL-6 and TNF-α production compared to models treated with 5-FU incubated at 35°C. CONCLUSION This study demonstrates a reduced cytotoxic effect to the TEOM by reducing the temperature of the tissue during chemotherapy treatment and suggests that decreasing the temperature to 20°C could have clinical advantages.
Collapse
Affiliation(s)
- Java Walladbegi
- Department of Oral Medicine & Pathology, Institute of Odontology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Sarah A Smith
- School of Clinical Dentistry, University of Sheffield, Sheffield, UK
| | - Amy K Grayson
- School of Clinical Dentistry, University of Sheffield, Sheffield, UK
| | - Craig Murdoch
- School of Clinical Dentistry, University of Sheffield, Sheffield, UK
| | - Mats Jontell
- Department of Oral Medicine & Pathology, Institute of Odontology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Helen E Colley
- School of Clinical Dentistry, University of Sheffield, Sheffield, UK
| |
Collapse
|
28
|
Morse DJ, Wilson MJ, Wei X, Lewis MAO, Bradshaw DJ, Murdoch C, Williams DW. Denture-associated biofilm infection in three-dimensional oral mucosal tissue models. J Med Microbiol 2018; 67:364-375. [PMID: 29458673 PMCID: PMC5882079 DOI: 10.1099/jmm.0.000677] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Purpose In vitro analyses of virulence, pathogenicity and associated host cell responses are important components in the study of biofilm infections. The Candida-related infection, denture-associated oral candidosis, affects up to 60 % of denture wearers and manifests as inflammation of palatal tissues contacting the denture-fitting surface. Commercially available three-dimensional tissue models can be used to study infection, but their use is limited for many academic research institutions, primarily because of the substantial purchase costs. The aim of this study was to develop and evaluate the use of in vitro tissue models to assess infections by biofilms on acrylic surfaces through tissue damage and Candida albicans virulence gene expression. Methodology In vitro models were compared against commercially available tissue equivalents (keratinocyte-only, SkinEthic; full-thickness, MatTek Corporation). An in vitro keratinocyte-only tissue was produced using a cancer-derived cell line, TR146, and a full-thickness model incorporating primary fibroblasts and immortalised normal oral keratinocytes was also generated. The in vitro full-thickness tissues incorporated keratinocytes and fibroblasts, and have potential for future further development and analysis. Results Following polymicrobial infection with biofilms on acrylic surfaces, both in-house developed models were shown to provide equivalent results to the SkinEthic and MatTek models in terms of tissue damage: a significant (P<0.05) increase in LDH activity for mixed species biofilms compared to uninfected control, and no significant difference (P>0.05) in the expression of most C. albicans virulence genes when comparing tissue models of the same type. Conclusion Our results confirm the feasibility and suitability of using these alternative in vitro tissue models for such analyses.
Collapse
Affiliation(s)
- Daniel J Morse
- Oral and Biomedical Sciences, School of Dentistry, Cardiff University, Cardiff, UK
| | - Melanie J Wilson
- Oral and Biomedical Sciences, School of Dentistry, Cardiff University, Cardiff, UK
| | - Xiaoqing Wei
- Oral and Biomedical Sciences, School of Dentistry, Cardiff University, Cardiff, UK
| | - Michael A O Lewis
- Oral and Biomedical Sciences, School of Dentistry, Cardiff University, Cardiff, UK
| | | | - Craig Murdoch
- School of Clinical Dentistry, University of Sheffield, Sheffield, UK
| | - David W Williams
- Oral and Biomedical Sciences, School of Dentistry, Cardiff University, Cardiff, UK
| |
Collapse
|
29
|
Application of standard cell cultures and 3D in vitro tissue models as an effective tool in drug design and development. Pharmacol Rep 2017. [DOI: 10.1016/j.pharep.2017.03.014] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
30
|
Pritchard M, Jack A, Powell L, Sadh H, Rye P, Hill K, Thomas D. Alginate oligosaccharides modify hyphal infiltration ofCandida albicansin anin vitromodel of invasive human candidosis. J Appl Microbiol 2017. [DOI: 10.1111/jam.13516] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- M.F. Pritchard
- Advanced Therapies Group; Cardiff University School of Dentistry; College of Biomedical and Life Sciences; Cardiff UK
| | - A.A. Jack
- Advanced Therapies Group; Cardiff University School of Dentistry; College of Biomedical and Life Sciences; Cardiff UK
| | - L.C. Powell
- Advanced Therapies Group; Cardiff University School of Dentistry; College of Biomedical and Life Sciences; Cardiff UK
| | - H. Sadh
- Advanced Therapies Group; Cardiff University School of Dentistry; College of Biomedical and Life Sciences; Cardiff UK
| | | | - K.E. Hill
- Advanced Therapies Group; Cardiff University School of Dentistry; College of Biomedical and Life Sciences; Cardiff UK
| | - D.W. Thomas
- Advanced Therapies Group; Cardiff University School of Dentistry; College of Biomedical and Life Sciences; Cardiff UK
| |
Collapse
|
31
|
Song G, Banov D, Bassani AS, Valdez BC. Evaluation of the Safety, Cell Migration, and Mucoadhesive Properties of a Mucoadhesive Polymer Blend in Human Oral Mucosa. AAPS PharmSciTech 2017; 18:1617-1623. [PMID: 27645471 DOI: 10.1208/s12249-016-0630-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 09/06/2016] [Indexed: 11/30/2022] Open
Abstract
The efficacy of active pharmaceutical ingredients (API) in compounded medications for oral mucosa greatly depends on the composition of the base. Here, we assessed the safety, facilitation of cell migration, and mucoadhesive properties of a newly developed mucoadhesive polymer blend (MPB) which contains pullulan, tamarindus indica polysaccharide, and sodium hyaluronate. No cell death was observed when human oral keratinocyte (HOK) and fibroblast (HOrF) cells were exposed to 1% MPB for 24 h. Epithelial cells in a 3D buccal tissue model (EpiOral) were unaffected when exposed to 50% MPB for 20 h whereas 1% Triton X-100 killed 93% cells after 4.5 h. The expressions of cytokines IL1α and IL1β and cell proliferation markers PCNA, CYCLIN A, and CYCLIN D1 in EpiOral tissue did not increase suggesting that MPB is neither an irritant nor a mitogen. Markers of apoptosis such as cleavage of CASPASES 8/9, upregulation of pro-apoptosis NOXA protein, and downregulation of anti-apoptosis XIAP protein were observed in Triton X-100-treated cells but not in cells exposed to MPB. The migration of HOK and HOrF cells was stimulated by MPB, and the expression of E-CADHERIN in the EpiOral tissues was unaffected. Moreover, MPB showed stronger mucoadhesion on the human EpiOral tissue model compared with a reference product. We conclude that MPB can safely deliver API within the oral mucosa, facilitate cell migration, and may increase drug efficacy through its strong mucoadhesive property.
Collapse
|
32
|
Jennings LR, Colley HE, Ong J, Panagakos F, Masters JG, Trivedi HM, Murdoch C, Whawell S. Development and Characterization of In Vitro Human Oral Mucosal Equivalents Derived from Immortalized Oral Keratinocytes. Tissue Eng Part C Methods 2016; 22:1108-1117. [PMID: 27846777 DOI: 10.1089/ten.tec.2016.0310] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Tissue-engineered oral mucosal equivalents (OME) are being increasingly used to measure toxicity, drug delivery, and to model oral diseases. Current OME mainly comprise normal oral keratinocytes (NOK) cultured on top of a normal oral fibroblasts-containing matrix. However, the commercial supply of NOK is limited, restricting widespread use of these mucosal models. In addition, NOK suffer from poor longevity and donor-to-donor variability. Therefore, we constructed, characterized, and tested the functionality of OME based on commercial TERT2-immortalized oral keratinocytes (FNB6) to produce a more readily available alternative to NOK-based OME. FNB6 OME cultured at an air-to-liquid interface for 14 days exhibited expression of differentiation markers cytokeratin 13 in the suprabasal layers and cytokeratin 14 in basal layer of the epithelium. Proliferating cells were restricted to the basal epithelium, and there was immuno-positive expression of E-cadherin confirming the presence of established cell-to-cell contacts. The histology and expression of these structural markers paralleled those observed in the normal oral mucosa and NOK-based models. On stimulation with TNFα and IL-1, FNB6 OME displayed a similar global gene expression profile to NOK-based OME, with increased expression of many common pro-inflammatory molecules such as chemokines (CXCL8), cytokines (IL-6), and adhesion molecules (ICAM-1) when analyzed by gene array and quantitative PCR. Similarly, pathway analysis showed that both FNB6 and NOK models initiated similar intracellular signaling on stimulation. Gene expression in FNB6 OME was more consistent than NOK-based OME that suffered from donor variation in response to stimuli. Mucosal equivalents based on immortalized FNB6 cells are accessible, reproducible and will provide an alternative animal experimental system for studying mucosal drug delivery systems, host-pathogen interactions, and drug-induced toxicity.
Collapse
Affiliation(s)
- Luke R Jennings
- 1 School of Clinical Dentistry, University of Sheffield , Sheffield, United Kingdom
| | - Helen E Colley
- 1 School of Clinical Dentistry, University of Sheffield , Sheffield, United Kingdom
| | - Jane Ong
- 2 Colgate-Palmolive Company , Piscataway, New Jersey
| | | | | | | | - Craig Murdoch
- 1 School of Clinical Dentistry, University of Sheffield , Sheffield, United Kingdom
| | - Simon Whawell
- 1 School of Clinical Dentistry, University of Sheffield , Sheffield, United Kingdom
| |
Collapse
|
33
|
Cheng R, Li D, Shi X, Gao Q, Wei C, Li X, Li Y, Zhou H. Reduced CX3CL1 Secretion Contributes to the Susceptibility of Oral Leukoplakia-Associated Fibroblasts to Candida albicans. Front Cell Infect Microbiol 2016; 6:150. [PMID: 27891323 PMCID: PMC5104956 DOI: 10.3389/fcimb.2016.00150] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 10/28/2016] [Indexed: 02/05/2023] Open
Abstract
Candida leukoplakia (OLK) is a kind of oral leukoplakia combined with chronic candidal infection, which plays an important role in the malignant transformation of OLK. However, little is known about the etiology, including susceptibility of leukoplakia to candidal adhesion, invasion and infection. Some antimicrobial peptides secreted by oral epithelial cells or fibroblasts potentially have antifungal activities against Candida albicans (C. albicans). In this study, we established three co-culture models to simulate different C. albicans-fibroblasts interactions during progression of candida leukoplakia. The susceptibility of oral leukoplakia-associated fibroblasts (LKAFs) to C. albicans and its underlying mechanism were determined. Samples of 14 LKAFs and 10 normal fibroblasts (NFs) were collected. The co-culture models showed that LKAFs had promoted the adhesion, invasion, and survival of C. albicans compared with NFs. CX3CL1, a chemokine with antifungal activity, was less abundant in LKAFs than NFs. Overexpression of CX3CL1 via transfection in LKAFs could partly restore the resistance to C. albicans. We also showed that inhibition of ERK could suppress CX3CL1 secretion. While phosphor-ERK was inhibited in LKAFs compared with NFs. Besides, the mRNA expression of a shedding enzyme for CX3CL1, disintegrin and metalloproteinase domain (ADAM) 17 was decreased in LKAFs than NFs. In conclusion, LKAFs produced and secreted less CX3CL1 by inhibiting the ERK signaling pathway, thereby contributing to impaired cell resistance to C. albicans.
Collapse
Affiliation(s)
- Ran Cheng
- State Key Laboratory of Oral Diseases, Department of Preventive Dentistry, West China Hospital of Stomatology, Sichuan University Chengdu, China
| | - Duo Li
- State Key Laboratory of Oral Diseases, Department of Oral Medicine, West China Hospital of Stomatology, Sichuan UniversityChengdu, China; Department of Oral Medicine, School of Stomatology, Dalian Medical UniversityDalian, China
| | - Xueke Shi
- State Key Laboratory of Oral Diseases, Department of Oral Medicine, West China Hospital of Stomatology, Sichuan University Chengdu, China
| | - Qinghong Gao
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University Chengdu, China
| | - Changlei Wei
- State Key Laboratory of Oral Diseases, Department of Oral Medicine, West China Hospital of Stomatology, Sichuan University Chengdu, China
| | - Xiaoyu Li
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University Chengdu, China
| | - Yan Li
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University Chengdu, China
| | - Hongmei Zhou
- State Key Laboratory of Oral Diseases, Department of Oral Medicine, West China Hospital of Stomatology, Sichuan University Chengdu, China
| |
Collapse
|
34
|
Gursoy UK, Gursoy M, Könönen E, Sintim HO, Uitto VJ, Syrjänen S. Construction and characterization of a multilayered gingival keratinocyte culture model: the TURK-U model. Cytotechnology 2016; 68:2345-2354. [PMID: 27752840 DOI: 10.1007/s10616-016-0029-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 09/17/2016] [Indexed: 12/01/2022] Open
Abstract
In construction of epithelial cells as multilayers, the cells are grown submerged to confluence on fibroblast-embedded collagen gels and, then, lifted to air to promote their stratification. We recently demonstrated that gingival epithelial cells form uniform monolayers on semi-permeable nitrocellulose membranes, supported with a semi-solid growth medium, which allows the cells to grow at an air-liquid-solid interface from the beginning of the culturing protocol. In this study, the aim was to further develop our previous model to form a multilayered gingival epithelial culture model. Two different epithelial cell lines (HaCaT from skin and HMK from gingiva) were used in all experiments. Both cell lines were grown first as monolayers for 3 days. After that, keratinocytes were trypsinized, counted and seeded on a sterile semi-permeable nitrocellulose membrane placed on the top of a semi-solid growth medium, forming an air-liquid-solid interface for the cells to grow. At days 1, 4, and 7, epithelial cells were fixed, embedded in paraffin, and sectioned for routine Hematoxylin-Eosin staining and immunohistochemistry for cytokeratin (Ck). At day 1, HMK cells grew as monolayers, while HaCaT cells stratified forming an epithelium with two to three layers. At day 4, a stratified epithelium in the HMK model had four to five layers and its proliferation continued up to day 7. HaCaT cells formed a dense and weakly proliferating epithelium with three to four layers of stratification at day 4 but the proliferation disappeared at day 7. At all days, both models were strongly positive for Ck5, Ck7, and Ck 19, and weakly positive for Ck10. Gingival epithelial cells stratify successfully on semi-permeable nitrocellulose membranes, supported with a semi-solid growth medium. This technique allows researchers to construct uniform gingival epithelial cell multilayers at an air-liquid-solid interface, without using collagen gels, resulting in a more reproducible method.
Collapse
Affiliation(s)
- Ulvi K Gursoy
- Department of Periodontology, Institute of Dentistry, University of Turku, Lemminkäisenkatu 2, 20520, Turku, Finland.
| | - Mervi Gursoy
- Department of Periodontology, Institute of Dentistry, University of Turku, Lemminkäisenkatu 2, 20520, Turku, Finland
| | - Eija Könönen
- Department of Periodontology, Institute of Dentistry, University of Turku, Lemminkäisenkatu 2, 20520, Turku, Finland.,Oral Health Care, Welfare Division, Turku, Finland
| | - Herman O Sintim
- Department of Chemistry and Purdue Institute for Drug Discovery and Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, IN, USA
| | - Veli-Jukka Uitto
- Department of Oral and Maxillofacial Diseases, University of Helsinki, Helsinki, Finland
| | - Stina Syrjänen
- Department of Oral Pathology, Institute of Dentistry, University of Turku, Turku, Finland
| |
Collapse
|
35
|
Zanetti F, Sewer A, Mathis C, Iskandar AR, Kostadinova R, Schlage WK, Leroy P, Majeed S, Guedj E, Trivedi K, Martin F, Elamin A, Merg C, Ivanov NV, Frentzel S, Peitsch MC, Hoeng J. Systems Toxicology Assessment of the Biological Impact of a Candidate Modified Risk Tobacco Product on Human Organotypic Oral Epithelial Cultures. Chem Res Toxicol 2016; 29:1252-69. [PMID: 27404394 DOI: 10.1021/acs.chemrestox.6b00174] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Cigarette smoke (CS) has been reported to increase predisposition to oral cancer and is also recognized as a risk factor for many conditions including periodontal diseases, gingivitis, and other benign mucosal disorders. Smoking cessation remains the most effective approach for minimizing the risk of smoking-related diseases. However, reduction of harmful constituents by heating rather than combusting tobacco, without modifying the amount of nicotine, is a promising new paradigm in harm reduction. In this study, we compared effects of exposure to aerosol derived from a candidate modified risk tobacco product, the tobacco heating system (THS) 2.2, with those of CS generated from the 3R4F reference cigarette. Human organotypic oral epithelial tissue cultures (EpiOral, MatTek Corporation) were exposed for 28 min to 3R4F CS or THS2.2 aerosol, both diluted with air to comparable nicotine concentrations (0.32 or 0.51 mg nicotine/L aerosol/CS for 3R4F and 0.31 or 0.46 mg/L for THS2.2). We also tested one higher concentration (1.09 mg/L) of THS2.2. A systems toxicology approach was employed combining cellular assays (i.e., cytotoxicity and cytochrome P450 activity assays), comprehensive molecular investigations of the buccal epithelial transcriptome (mRNA and miRNA) by means of computational network biology, measurements of secreted proinflammatory markers, and histopathological analysis. We observed that the impact of 3R4F CS was greater than THS2.2 aerosol in terms of cytotoxicity, morphological tissue alterations, and secretion of inflammatory mediators. Analysis of the transcriptomic changes in the exposed oral cultures revealed significant perturbations in various network models such as apoptosis, necroptosis, senescence, xenobiotic metabolism, oxidative stress, and nuclear factor (erythroid-derived 2)-like 2 (NFE2L2) signaling. The stress responses following THS2.2 aerosol exposure were markedly decreased, and the exposed cultures recovered more completely compared with those exposed to 3R4F CS.
Collapse
Affiliation(s)
- Filippo Zanetti
- Philip Morris International Research and Development , Quai Jeanrenaud 5, 2000 Neuchâtel, Switzerland
| | - Alain Sewer
- Philip Morris International Research and Development , Quai Jeanrenaud 5, 2000 Neuchâtel, Switzerland
| | - Carole Mathis
- Philip Morris International Research and Development , Quai Jeanrenaud 5, 2000 Neuchâtel, Switzerland
| | - Anita R Iskandar
- Philip Morris International Research and Development , Quai Jeanrenaud 5, 2000 Neuchâtel, Switzerland
| | - Radina Kostadinova
- Philip Morris International Research and Development , Quai Jeanrenaud 5, 2000 Neuchâtel, Switzerland
| | - Walter K Schlage
- Biology Consultant , Max-Baermann-Str. 21, 51429 Bergisch Gladbach, Germany
| | - Patrice Leroy
- Philip Morris International Research and Development , Quai Jeanrenaud 5, 2000 Neuchâtel, Switzerland
| | - Shoaib Majeed
- Philip Morris International Research and Development , Quai Jeanrenaud 5, 2000 Neuchâtel, Switzerland
| | - Emmanuel Guedj
- Philip Morris International Research and Development , Quai Jeanrenaud 5, 2000 Neuchâtel, Switzerland
| | - Keyur Trivedi
- Philip Morris International Research and Development , Quai Jeanrenaud 5, 2000 Neuchâtel, Switzerland
| | - Florian Martin
- Philip Morris International Research and Development , Quai Jeanrenaud 5, 2000 Neuchâtel, Switzerland
| | - Ashraf Elamin
- Philip Morris International Research and Development , Quai Jeanrenaud 5, 2000 Neuchâtel, Switzerland
| | - Céline Merg
- Philip Morris International Research and Development , Quai Jeanrenaud 5, 2000 Neuchâtel, Switzerland
| | - Nikolai V Ivanov
- Philip Morris International Research and Development , Quai Jeanrenaud 5, 2000 Neuchâtel, Switzerland
| | - Stefan Frentzel
- Philip Morris International Research and Development , Quai Jeanrenaud 5, 2000 Neuchâtel, Switzerland
| | - Manuel C Peitsch
- Philip Morris International Research and Development , Quai Jeanrenaud 5, 2000 Neuchâtel, Switzerland
| | - Julia Hoeng
- Philip Morris International Research and Development , Quai Jeanrenaud 5, 2000 Neuchâtel, Switzerland
| |
Collapse
|
36
|
Delben JA, Zago CE, Tyhovych N, Duarte S, Vergani CE. Effect of Atmospheric-Pressure Cold Plasma on Pathogenic Oral Biofilms and In Vitro Reconstituted Oral Epithelium. PLoS One 2016; 11:e0155427. [PMID: 27224027 PMCID: PMC4880209 DOI: 10.1371/journal.pone.0155427] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 04/28/2016] [Indexed: 01/13/2023] Open
Abstract
Considering the ability of atmospheric-pressure cold plasma (ACP) to disrupt the biofilm matrix and rupture cell structure, it can be an efficient tool against virulent oral biofilms. However, it is fundamental that ACP does not cause damage to oral tissue. So, this study evaluated (1) the antimicrobial effect of ACP on single- and dual-species biofilms of Candida albicans and Staphylococcus aureus as well as (2) the biological safety of ACP on in vitro reconstituted oral epithelium. Standardized cell suspensions of each microorganism were prepared for biofilm culture on acrylic resin discs at 37°C for 48 hours. The biofilms were submitted to ACP treatment at 10 mm of plasma tip-to-sample distance during 60 seconds. Positive controls were penicillin G and fluconazole for S. aureus and C. albicans, respectively. The biofilms were analyzed through counting of viable colonies, confocal laser scanning microscopy, scanning electron microscopy and fluorescence microscopy for detection of reactive oxygen species. The in vitro reconstituted oral epithelium was submitted to similar ACP treatment and analyzed through histology, cytotoxocity test (LDH release), viability test (MTT assay) and imunnohistochemistry (Ki67 expression). All plasma-treated biofilms presented significant log10 CFU/mL reduction, alteration in microorganism/biofilm morphology, and reduced viability in comparison to negative and positive controls. In addition, fluorescence microscopy revealed presence of reactive oxygen species in all plasma-treated biofilms. Low cytotoxicity and high viability were observed in oral epithelium of negative control and plasma group. Histology showed neither sign of necrosis nor significant alteration in plasma-treated epithelium. Ki67-positive cells revealed maintenance of cell proliferation in plasma-treated epithelium. Atmospheric-pressure cold plasma is a promissing approach to eliminate single- and dual-species biofilms of C. albicans and S. aureus without having toxic effects in oral epithelium.
Collapse
Affiliation(s)
- Juliana Aparecida Delben
- Department of Dental Materials and Prosthodontics, Araraquara Dental School, Sao Paulo State University, Araraquara, Sao Paulo, Brazil
| | - Chaiene Evelin Zago
- Department of Dental Materials and Prosthodontics, Araraquara Dental School, Sao Paulo State University, Araraquara, Sao Paulo, Brazil
| | - Natalia Tyhovych
- Department of Basic Science and Craniofacial Biology, College of Dentistry, New York University, New York, New York, United States of America
| | - Simone Duarte
- Department of Basic Science and Craniofacial Biology, College of Dentistry, New York University, New York, New York, United States of America
| | - Carlos Eduardo Vergani
- Department of Dental Materials and Prosthodontics, Araraquara Dental School, Sao Paulo State University, Araraquara, Sao Paulo, Brazil
- * E-mail:
| |
Collapse
|
37
|
Cavalcanti YW, Morse DJ, da Silva WJ, Del-Bel-Cury AA, Wei X, Wilson M, Milward P, Lewis M, Bradshaw D, Williams DW. Virulence and pathogenicity of Candida albicans is enhanced in biofilms containing oral bacteria. BIOFOULING 2015; 31:27-38. [PMID: 25574582 DOI: 10.1080/08927014.2014.996143] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
This study examined the influence of bacteria on the virulence and pathogenicity of candidal biofilms. Mature biofilms (Candida albicans-only, bacteria-only, C. albicans with bacteria) were generated on acrylic and either analysed directly, or used to infect a reconstituted human oral epithelium (RHOE). Analyses included Candida hyphae enumeration and assessment of Candida virulence gene expression. Lactate dehydrogenase (LDH) activity and Candida tissue invasion following biofilm infection of the RHOE were also measured. Candida hyphae were more prevalent (p < 0.05) in acrylic biofilms also containing bacteria, with genes encoding secreted aspartyl-proteinases (SAP4/SAP6) and hyphal-wall protein (HWP1) up-regulated (p < 0.05). Candida adhesin genes (ALS3/EPA1), SAP6 and HWP1 were up-regulated in mixed-species biofilm infections of RHOE. Multi-species infections exhibited higher hyphal proportions (p < 0.05), up-regulation of IL-18, higher LDH activity and tissue invasion. As the presence of bacteria in acrylic biofilms promoted Candida virulence, consideration should be given to the bacterial component when managing denture biofilm associated candidoses.
Collapse
|
38
|
Kinikoglu B, Damour O, Hasirci V. Tissue engineering of oral mucosa: a shared concept with skin. J Artif Organs 2014; 18:8-19. [PMID: 25326194 DOI: 10.1007/s10047-014-0798-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Accepted: 10/07/2014] [Indexed: 12/17/2022]
Abstract
Tissue-engineered oral mucosa, in the form of epithelial cell sheets or full-thickness oral mucosa equivalents, is a potential solution for many patients with congenital defects or with tissue loss due to diseases or tumor excision following a craniofacial cancer diagnosis. In the laboratory, it further serves as an in vitro model, alternative to in vivo testing of oral care products, and provides insight into the behavior of the oral mucosal cells in healthy and pathological tissues. This review covers the old and new generation scaffold types and materials used in oral mucosa engineering; discusses similarities and differences between oral mucosa and skin, the methods developed to reconstruct oral mucosal defects; and ends with future perspectives on oral mucosa engineering.
Collapse
Affiliation(s)
- Beste Kinikoglu
- Department of Medical Biology, School of Medicine, Acibadem University, 34742, Istanbul, Turkey,
| | | | | |
Collapse
|
39
|
Alves CT, Wei XQ, Silva S, Azeredo J, Henriques M, Williams DW. Candida albicans promotes invasion and colonisation of Candida glabrata in a reconstituted human vaginal epithelium. J Infect 2014; 69:396-407. [DOI: 10.1016/j.jinf.2014.06.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Revised: 05/29/2014] [Accepted: 06/02/2014] [Indexed: 01/12/2023]
|
40
|
Morales JO, Huang S, Williams RO, McConville JT. Films loaded with insulin-coated nanoparticles (ICNP) as potential platforms for peptide buccal delivery. Colloids Surf B Biointerfaces 2014; 122:38-45. [DOI: 10.1016/j.colsurfb.2014.05.025] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Revised: 05/06/2014] [Accepted: 05/16/2014] [Indexed: 01/16/2023]
|
41
|
Lefebvre DE, Venema K, Gombau L, Valerio LG, Raju J, Bondy GS, Bouwmeester H, Singh RP, Clippinger AJ, Collnot EM, Mehta R, Stone V. Utility of models of the gastrointestinal tract for assessment of the digestion and absorption of engineered nanomaterials released from food matrices. Nanotoxicology 2014; 9:523-42. [PMID: 25119418 DOI: 10.3109/17435390.2014.948091] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Engineered metal/mineral, lipid and biochemical macromolecule nanomaterials (NMs) have potential applications in food. Methodologies for the assessment of NM digestion and bioavailability in the gastrointestinal tract are nascent and require refinement. A working group was tasked by the International Life Sciences Institute NanoRelease Food Additive project to review existing models of the gastrointestinal tract in health and disease, and the utility of these models for the assessment of the uptake of NMs intended for food. Gastrointestinal digestion and absorption could be addressed in a tiered approach using in silico computational models, in vitro non-cellular fluid systems and in vitro cell culture models, after which the necessity of ex vivo organ culture and in vivo animal studies can be considered. Examples of NM quantification in gastrointestinal tract fluids and tissues are emerging; however, few standardized analytical techniques are available. Coupling of these techniques to gastrointestinal models, along with further standardization, will further strengthen methodologies for risk assessment.
Collapse
Affiliation(s)
- David E Lefebvre
- Regulatory Toxicology Research Division, Food Directorate, Health Canada , Ottawa , Canada
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Morales JO, McConville JT. Novel strategies for the buccal delivery of macromolecules. Drug Dev Ind Pharm 2014; 40:579-90. [DOI: 10.3109/03639045.2014.892960] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
43
|
Szabo EK, Maccallum DM. A novel renal epithelial cell in vitro assay to assess Candida albicans virulence. Virulence 2013; 5:286-96. [PMID: 24225657 DOI: 10.4161/viru.27046] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Candida albicans, an opportunistic fungal pathogen, can cause severe systemic infections in susceptible patient groups. Systemic candidiasis is mainly studied in the mouse intravenous challenge model, where progressive infection correlates with increased early renal chemokine levels. To develop a new in vitro assay to assess C. albicans virulence, which reflects the events occurring in the murine infection model, renal M-1 cortical collecting duct epithelial cells were evaluated as the early producers of cytokines in response to C. albicans. We show that renal epithelial cells respond only to live C. albicans cells capable of forming hyphae, producing chemokines KC and MIP-2, with levels correlating with epithelial cell damage. By assaying epithelial cell responses to strains of known virulence in the murine intravenous challenge model we demonstrate that renal epithelial cells can discriminate between virulent and attenuated strains. This simple, novel assay is a useful initial screen for altered virulence of C. albicans mutants or clinical isolates in vitro and provides an alternative to the mouse systemic infection model.
Collapse
Affiliation(s)
- Edina K Szabo
- Aberdeen Fungal Group; University of Aberdeen; School of Medical Sciences; Institute of Medical Sciences; Foresterhill, Aberdeen UK
| | - Donna M Maccallum
- Aberdeen Fungal Group; University of Aberdeen; School of Medical Sciences; Institute of Medical Sciences; Foresterhill, Aberdeen UK
| |
Collapse
|
44
|
Wayakanon K, Thornhill MH, Douglas CWI, Lewis AL, Warren NJ, Pinnock A, Armes SP, Battaglia G, Murdoch C. Polymersome-mediated intracellular delivery of antibiotics to treat Porphyromonas gingivalis-infected oral epithelial cells. FASEB J 2013; 27:4455-65. [PMID: 23921377 DOI: 10.1096/fj.12-225219] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The gram-negative anaerobe Porphyromonas gingivalis colonizes the gingival crevice and is etiologically associated with periodontal disease that can lead to alveolar bone damage and resorption, promoting tooth loss. Although susceptible to antibiotics, P. gingivalis can evade antibiotic killing by residing within gingival keratinocytes. This provides a reservoir of organisms that may recolonize the gingival crevice once antibiotic therapy is complete. Polymersomes are nanosized amphiphilic block copolymer vesicles that can encapsulate drugs. Cells internalize polymersomes by endocytosis into early endosomes, where they are disassembled by the low pH, causing intracellular release of their drug load. In this study, polymersomes were used as vehicles to deliver antibiotics in an attempt to kill intracellular P. gingivalis within monolayers of keratinocytes and organotypic oral mucosal models. Polymersome-encapsulated metronidazole or doxycycline, free metronidazole, or doxycycline, or polymersomes alone as controls, were used, and the number of surviving intracellular P. gingivalis was quantified after host cell lysis. Polymersome-encapsulated metronidazole or doxycycline significantly (P<0.05) reduced the number of intracellular P. gingivalis in both monolayer and organotypic cultures compared to free antibiotic or polymersome alone controls. Polymersomes are effective delivery vehicles for antibiotics that do not normally gain entry to host cells. This approach could be used to treat recurrent periodontitis or other diseases caused by intracellular-dwelling organisms.
Collapse
Affiliation(s)
- Kornchanok Wayakanon
- 1C.M., Unit of Oral and Maxillofacial Medicine and Surgery, School of Clinical Dentistry, University of Sheffield, Claremont Crescent, Sheffield, S10 2TA, UK.
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Colley HE, Eves PC, Pinnock A, Thornhill MH, Murdoch C. Tissue-engineered oral mucosa to study radiotherapy-induced oral mucositis. Int J Radiat Biol 2013; 89:907-14. [PMID: 23718176 DOI: 10.3109/09553002.2013.809171] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE Oral mucositis is a severe and often dose-limiting side-effect of cancer therapy that occurs in patients receiving radiotherapy for head and neck cancers. Although radiation-induced effects on keratinocytes have been studied, little is known about its effect on fibroblasts or endothelial cells or, more importantly, when all these cells are combined in an engineered oral mucosal model. MATERIALS AND METHODS Monolayer cultures of normal oral keratinocytes, normal oral fibroblasts, human dermal microvascular endothelial cells or tissue-engineered oral mucosa (TEOM) were exposed to 20 Gy irradiation. Cell damage and cytokine release was measured for 72 h for monolayer cultures and for up to 21 d for TEOM. RESULTS Compared to non-irradiated cells, the viability of all monolayer and co-cultures was significantly reduced 72 h post-irradiation while levels of secreted interleukin IL-6 and CXCL8 were increased. The viability of irradiated TEOM models was significantly reduced compared to controls at all time-points. Histologically, irradiated TEOM displayed thinner epithelium, increased apoptosis and more extensive damage than non-irradiated models. IL-6, CXCL8 and granulocyte macrophage colony-stimulating factor release was reduced whereas IL-1α levels were increased in irradiated TEOM models compared to controls. CONCLUSIONS TEOM models comprising of mixed cell populations may prove useful in examining the pathobiology of radiation-induced mucositis.
Collapse
Affiliation(s)
- Helen E Colley
- Academic Unit of Oral and Maxillofacial Medicine and Surgery, School of Clinical Dentistry, University of Sheffield , Sheffield , UK
| | | | | | | | | |
Collapse
|
46
|
Moharamzadeh K, Colley H, Murdoch C, Hearnden V, Chai WL, Brook IM, Thornhill MH, Macneil S. Tissue-engineered oral mucosa. J Dent Res 2012; 91:642-50. [PMID: 22266525 DOI: 10.1177/0022034511435702] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Advances in tissue engineering have permitted the three-dimensional (3D) reconstruction of human oral mucosa for various in vivo and in vitro applications. Tissue-engineered oral mucosa have been further optimized in recent years for clinical applications as a suitable graft material for intra-oral and extra-oral repair and treatment of soft-tissue defects. Novel 3D in vitro models of oral diseases such as cancer, Candida, and bacterial invasion have been developed as alternatives to animal models for investigation of disease phenomena, their progression, and treatment, including evaluation of drug delivery systems. The introduction of 3D oral mucosal reconstructs has had a significant impact on the approaches to biocompatibility evaluation of dental materials and oral healthcare products as well as the study of implant-soft tissue interfaces. This review article discusses the recent advances in tissue engineering and applications of tissue-engineered human oral mucosa.
Collapse
Affiliation(s)
- K Moharamzadeh
- School of Clinical Dentistry, University of Sheffield, Claremont Crescent, Sheffield, S10 2TA, UK.
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Development of tissue-engineered models of oral dysplasia and early invasive oral squamous cell carcinoma. Br J Cancer 2011; 105:1582-92. [PMID: 21989184 PMCID: PMC3242522 DOI: 10.1038/bjc.2011.403] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Background: Current organotypic models of dysplasia and oral squamous cell carcinoma (OSCC) lack the complexity that mimics in vivo tissue. Here we describe a three-dimensional in vitro model of the oral epithelium that replicates tumour progression from dysplasia to an invasive phenotype. Methods: The OSCC cell lines were seeded as a cell suspension (D20, Cal27) or as multicellular tumour spheroids (FaDu) with oral fibroblasts on to a de-epidermised acellular dermis to generate tissue-engineered models and compared with patient biopsies. Results: The D20 and Cal27 cells generated a model of epithelial dysplasia. Overtime Cal27 cells traversed the basement membrane and invaded the connective tissue to reproduce features of early invasive OSCC. When seeded onto a model of the normal oral mucosa, FaDu spheroids produced a histological picture mimicking carcinoma in situ with severe cellular atypia juxtaposed to normal epithelium. Conclusion: It is possible to culture in vitro models with the morphological appearance and histological characteristics of dysplasia and tumour cell invasion seen in vivo using native dermis. Such models could facilitate study of the molecular processes involved in malignant transformation, invasion and tumour growth as well as in vitro testing of new treatments, diagnostic tests and drug delivery systems for OSCC.
Collapse
|