1
|
Zaeneldin A, Chu CH, Yu OY. Diffusion of Silver Diamine Fluoride Solution in Dentine: An In Vitro Study. Int Dent J 2025; 75:767-776. [PMID: 39097438 PMCID: PMC11976465 DOI: 10.1016/j.identj.2024.06.002] [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: 04/14/2024] [Revised: 05/30/2024] [Accepted: 06/01/2024] [Indexed: 08/05/2024] Open
Abstract
OBJECTIVES The aim of this research was to assess the diffusion dynamics of silver and fluoride ions after 38% silver diamine fluoride (SDF) solution application on dentine of varying thicknesses over 24 weeks. METHODS Bovine dentine discs of 5.5 mm in diameter were prepared and separated into 3 groups with thicknesses of 0.5 mm (group 1), 1.0 mm (group 2), and 1.5 mm (group 3). The diameter and number of dentinal tubules of discs were assessed. Each disc received a topical application of 0.05 mL 38% SDF solution. The deionised water in the tube was collected weekly for 24 weeks. The silver and the fluoride ion concentrations in the collected deionised water were determined. Generalised estimating equations was used to explore the potential effects of the key factors on the silver/fluoride diffusion. RESULTS The amount of silver and fluoride ion diffusion through dentine almost levelled off after 4 weeks and showed a decline trend over 24 weeks. The mean (SD) 24-week cumulative ion diffusion through dentine in groups 1, 2, and 3 was as follows: 20 (4) μg, 10 (2) μg, and 5 (1) μg for silver (P < .05) and 18 (2) μg, 13 (2) μg, and 7 (1) μg for fluoride (P < .05), respectively. CONCLUSIONS Silver and fluoride ion diffusion through dentine showed a decline trend over 24 weeks. The diameter and the number of dentinal tubules on dentine with different thicknesses affects the ion diffusion dynamics. This study provides indications on the pattern of silver/fluoride ions diffusion through dentine to pulp after 38% SDF application. An increased amount of silver/fluoride diffuses through dentine into the pulp with decreased remaining dentine thickness.
Collapse
Affiliation(s)
- Ahmed Zaeneldin
- Faculty of Dentistry, The University of Hong Kong, Hong Kong, S.A.R., China
| | - Chun Hung Chu
- Faculty of Dentistry, The University of Hong Kong, Hong Kong, S.A.R., China
| | - Ollie Yiru Yu
- Faculty of Dentistry, The University of Hong Kong, Hong Kong, S.A.R., China.
| |
Collapse
|
2
|
Jităreanu A, Agoroaei L, Caba IC, Cojocaru FD, Vereștiuc L, Vieriu M, Mârțu I. The Evolution of In Vitro Toxicity Assessment Methods for Oral Cavity Tissues-From 2D Cell Cultures to Organ-on-a-Chip. TOXICS 2025; 13:195. [PMID: 40137522 PMCID: PMC11946525 DOI: 10.3390/toxics13030195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 03/03/2025] [Accepted: 03/06/2025] [Indexed: 03/29/2025]
Abstract
Since the oral cavity comes into contact with several xenobiotics (dental materials, oral hygiene formulations, drugs, or tobacco products), it is one major site for toxicity manifestation. Multiple parameters are assessed during toxicity testing (cell viability and proliferation, apoptosis, morphological changes, genotoxicity, oxidative stress, and inflammatory response). Due to the complexity of the oral cavity environment, researchers have made great efforts to design better in vitro models that mimic natural human anatomic and functional features. The present review describes the in vitro methods currently used to investigate the toxic potential of various agents on oral cavity tissues and their evolution from simple 2D cell culture systems to complex organ-a-chip designs.
Collapse
Affiliation(s)
- Alexandra Jităreanu
- Department of Toxicology, Faculty of Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy Iasi, 700115 Iasi, Romania;
| | - Luminița Agoroaei
- Department of Toxicology, Faculty of Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy Iasi, 700115 Iasi, Romania;
| | - Ioana-Cezara Caba
- Department of Toxicology, Faculty of Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy Iasi, 700115 Iasi, Romania;
| | - Florina-Daniela Cojocaru
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, “Grigore T. Popa” University of Medicine and Pharmacy Iasi, 700115 Iasi, Romania; (F.-D.C.); (L.V.)
| | - Liliana Vereștiuc
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, “Grigore T. Popa” University of Medicine and Pharmacy Iasi, 700115 Iasi, Romania; (F.-D.C.); (L.V.)
| | - Mădălina Vieriu
- Department of Analytical Chemistry, Faculty of Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy Iasi, 700115 Iasi, Romania;
| | - Ioana Mârțu
- Department of Dental Technology, Faculty of Dental Medicine, “Grigore T. Popa” University of Medicine and Pharmacy Iasi, 700115 Iasi, Romania;
| |
Collapse
|
3
|
Machla F, Monou PK, Artemiou P, Angelopoulos I, Zisis V, Panteris E, Katsamenis O, Williams E, Tzimtzimis E, Tzetzis D, Andreadis D, Tsouknidas A, Fatouros D, Bakopoulou A. Design, additive manufacturing, and characterization of an organ-on-chip microfluidic device for oral mucosa analogue growth. J Mech Behav Biomed Mater 2025; 163:106877. [PMID: 39729779 DOI: 10.1016/j.jmbbm.2024.106877] [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: 10/04/2024] [Revised: 12/15/2024] [Accepted: 12/17/2024] [Indexed: 12/29/2024]
Abstract
INTRODUCTION Α customized organ-on-a-chip microfluidic device was developed for dynamic culture of oral mucosa equivalents (Oral_mucosa_chip-OMC). MATERIALS AND METHODS Additive Manufacturing (AM) was performed via stereolithography (SLA) printing. The dimensional accuracy was evaluated via microfocus computed tomography (mCT), the surface characteristics via scanning electron microscopy (SEM), while the mechanical properties via nanoindentation and compression tests. Computational fluid dynamics (CFD) optimized net forces towards the culture area. An oral mucosa equivalent comprising a multilayered epithelium derived by culture of TR146 cells at the air-liquid interface (ALI) and a lamina propria-analogue based on a collagen-I/fibrin hydrogel was maintained under ultra-precise flow conditions. RESULTS An open-type device concept encompassing two interconnected chambers for long-term dynamic culture was developed and characterized for AM parameters, mechanical and biological properties. The split-inlet flow channel architecture allowed even distribution and symmetric flow velocity to the culture area. Cell viability exceeded 90%, while mCT and SEM indicated the 0° building angle as the most accurate SLA condition. CFD further showed that the 0° and 30° building angles most accurately reproduced the channel flow velocity predicted by the initial CAD model. CONCLUSION This study developed a customized, easy-to-produce, and cell-friendly OMC device, providing a 3D tool for biocompatibility assessment of biomaterials.
Collapse
Affiliation(s)
- Foteini Machla
- Department of Prosthodontics, Dental and Craniofacial Bioengineering and Applied Biomaterials, School of Dentistry, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, 54124, Greece
| | - Paraskevi Kyriaki Monou
- Department of Pharmaceutical Technology, School of Pharmacy, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, 54124, Greece; Center for Interdisciplinary Research and Innovation (CIRI-AUTH), Thessaloniki, 57001, Greece
| | - Panagiotis Artemiou
- Department of Mechanical Engineering, University of Western Macedonia, Kozani, 50100, Greece
| | - Ioannis Angelopoulos
- Department of Prosthodontics, Dental and Craniofacial Bioengineering and Applied Biomaterials, School of Dentistry, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, 54124, Greece
| | - Vasileios Zisis
- Department of Oral Medicine/Pathology, School of Dentistry, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, 54124, Greece
| | - Emmanuel Panteris
- Department of Botany, School of Biology, Faculty of Sciences, Aristotle University of Thessaloniki, Thessaloniki, 54124, Greece
| | - Orestis Katsamenis
- μ-VIS X-ray Imaging Centre, Faculty of Engineering and the Environment, University of Southampton, Southampton, SO17 1BJ, United Kingdom; Institute for Life Sciences, University of Southampton, SO17 1BJ, Southampto, United Kingdom
| | - Eric Williams
- μ-VIS X-ray Imaging Centre, Faculty of Engineering and the Environment, University of Southampton, Southampton, SO17 1BJ, United Kingdom
| | - Emmanouil Tzimtzimis
- Digital Manufacturing and Materials Characterization Laboratory, School of Science and Technology, International Hellenic University, 57001, Thermi, Greece
| | - Dimitrios Tzetzis
- Digital Manufacturing and Materials Characterization Laboratory, School of Science and Technology, International Hellenic University, 57001, Thermi, Greece
| | - Dimitrios Andreadis
- Digital Manufacturing and Materials Characterization Laboratory, School of Science and Technology, International Hellenic University, 57001, Thermi, Greece
| | - Alexander Tsouknidas
- Department of Mechanical Engineering, University of Western Macedonia, Kozani, 50100, Greece
| | - Dimitrios Fatouros
- Department of Pharmaceutical Technology, School of Pharmacy, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, 54124, Greece; Center for Interdisciplinary Research and Innovation (CIRI-AUTH), Thessaloniki, 57001, Greece
| | - Athina Bakopoulou
- Department of Prosthodontics, Dental and Craniofacial Bioengineering and Applied Biomaterials, School of Dentistry, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, 54124, Greece.
| |
Collapse
|
4
|
Rosa V, Cavalcanti BN, Nör JE, Tezvergil-Mutluay A, Silikas N, Bottino MC, Kishen A, Soares DG, Franca CM, Cooper PR, Duncan HF, Ferracane JL, Watts DC. Guidance for evaluating biomaterials' properties and biological potential for dental pulp tissue engineering and regeneration research. Dent Mater 2025; 41:248-264. [PMID: 39674710 PMCID: PMC11875114 DOI: 10.1016/j.dental.2024.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 12/10/2024] [Indexed: 12/16/2024]
Abstract
BACKGROUND Dental pulp regeneration is a complex and advancing field that requires biomaterials capable of supporting the pulp's diverse functions, including immune defense, sensory perception, vascularization, and reparative dentinogenesis. Regeneration involves orchestrating the formation of soft connective tissues, neurons, blood vessels, and mineralized structures, necessitating materials with tailored biological and mechanical properties. Numerous biomaterials have entered clinical practice, while others are being developed for tissue engineering applications. The composition and a broad range of material properties, such as surface characteristics, degradation rate, and mechanical strength, significantly influence cellular behavior and tissue outcomes. This underscores the importance of employing robust evaluation methods and ensuring precise and comprehensive reporting of findings to advance research and clinical translation. AIMS This article aims to present the biological foundations of dental pulp tissue engineering alongside potential testing methodologies and their advantages and limitations. It provides guidance for developing research protocols to evaluate the properties of biomaterials and their influences on cell and tissue behavior, supporting progress toward effective dental pulp regeneration strategies.
Collapse
Affiliation(s)
- Vinicius Rosa
- Faculty of Dentistry, National University of Singapore, Singapore; ORCHIDS: Oral Care Health Innovations and Designs Singapore, National University of Singapore, Singapore.
| | - Bruno Neves Cavalcanti
- Department of Cariology, Restorative Sciences, and Endodontics, Division of Endodontics, School of Dentistry, University of Michigan, Ann Arbor, United States.
| | - Jacques E Nör
- Department of Cariology, Restorative Sciences, and Endodontics, Division of Endodontics, School of Dentistry, University of Michigan, Ann Arbor, United States.
| | - Arzu Tezvergil-Mutluay
- Department of Cariology and Restorative Dentistry, Institute of Dentistry, University of Turku, Turku, Finland; Turku University Hospital, TYKS, Turku, Finland.
| | - Nikolaos Silikas
- Division of Dentistry, School of Medical Sciences, University of Manchester, Manchester, United Kingdom.
| | - Marco C Bottino
- Department of Cariology, Restorative Sciences, and Endodontics, Division of Endodontics, School of Dentistry, University of Michigan, Ann Arbor, United States; Department of Biomedical Engineering, College of Engineering, University of Michigan, Ann Arbor, United States.
| | - Anil Kishen
- Faculty of Dentistry, University of Toronto, Toronto, Canada; Department of Dentistry, Mount Sinai Health System, Mount Sinai Hospital, Toronto, Canada.
| | - Diana Gabriela Soares
- Department of Operative Dentistry, Endodontics and Dental Materials, School of Dentistry, São Paulo University, Bauru, Brazil.
| | - Cristiane M Franca
- Department of Oral Rehabilitation and Biosciences, School of Dentistry, Oregon Health & Science University (OHSU), Portland, USA; Knight Cancer Precision Biofabrication Hub, Oregon Health & Science University (OHSU), Portland, USA.
| | - Paul Roy Cooper
- Sir John Walsh Research Institute, Department of Oral Sciences, Faculty of Dentistry, University of Otago, New Zealand.
| | - Henry F Duncan
- Division of Restorative Dentistry and Periodontology, Dublin Dental University Hospital, Trinity College Dublin, University of Dublin, Dublin, Ireland.
| | - Jack L Ferracane
- Department of Oral Rehabilitation and Biosciences, School of Dentistry, Oregon Health & Science University (OHSU), Portland, USA.
| | - David C Watts
- Division of Dentistry, School of Medical Sciences, University of Manchester, Manchester, United Kingdom.
| |
Collapse
|
5
|
Mbitta Akoa D, Avril A, Hélary C, Poliard A, Coradin T. Evaluation of Silica and Bioglass Nanomaterials in Pulp-like Living Materials. ACS Biomater Sci Eng 2025; 11:891-902. [PMID: 39803670 DOI: 10.1021/acsbiomaterials.4c01898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2025]
Abstract
Although silicon is a widespread constituent in dental materials, its possible influence on the formation and repair of teeth remains largely unexplored. Here, we studied the effect of two silicic acid-releasing nanomaterials, silica and bioglass, on a living model of pulp consisting of dental pulp stem cells seeded in dense type I collagen hydrogels. Silica nanoparticles and released silicic acid had little effect on cell viability and mineralization efficiency but impacted metabolic activity, delayed matrix remodeling, and led to heterogeneous cell distribution. Bioglass improved cell metabolic activity and led to a homogeneous dispersion of cells and mineral deposits within the hydrogel. These results suggest that the presence of calcium ions in bioglass is not only favorable to cell proliferation but can also counterbalance the negative effects of silicon. Both chemical and biological processes should therefore be considered when investigating the effects of silicon-containing materials on dental tissues.
Collapse
Affiliation(s)
- Daline Mbitta Akoa
- Sorbonne Université, CNRS, Laboratoire de Chimie de la Matière Condensée de Paris, Paris 75252, France
| | - Anthony Avril
- Sorbonne Université, CNRS, Laboratoire de Chimie de la Matière Condensée de Paris, Paris 75252, France
| | - Christophe Hélary
- Sorbonne Université, CNRS, Laboratoire de Chimie de la Matière Condensée de Paris, Paris 75252, France
| | - Anne Poliard
- Université de Paris Cité, UR2496 Pathologies, Imagerie et Biothérapies Orofaciales, FHU-DDS-Net, Dental School, Montrouge 92120, France
| | - Thibaud Coradin
- Sorbonne Université, CNRS, Laboratoire de Chimie de la Matière Condensée de Paris, Paris 75252, France
| |
Collapse
|
6
|
Rosa V, Silikas N, Yu B, Dubey N, Sriram G, Zinelis S, Lima AF, Bottino MC, Ferreira JN, Schmalz G, Watts DC. Guidance on the assessment of biocompatibility of biomaterials: Fundamentals and testing considerations. Dent Mater 2024; 40:1773-1785. [PMID: 39129079 DOI: 10.1016/j.dental.2024.07.020] [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: 06/09/2024] [Revised: 07/22/2024] [Accepted: 07/24/2024] [Indexed: 08/13/2024]
Abstract
BACKGROUND Assessing the biocompatibility of materials is crucial for ensuring the safety and well-being of patients by preventing undesirable, toxic, immune, or allergic reactions, and ensuring that materials remain functional over time without triggering adverse reactions. To ensure a comprehensive assessment, planning tests that carefully consider the intended application and potential exposure scenarios for selecting relevant assays, cell types, and testing parameters is essential. Moreover, characterizing the composition and properties of biomaterials allows for a more accurate understanding of test outcomes and the identification of factors contributing to cytotoxicity. Precise reporting of methodology and results facilitates research reproducibility and understanding of the findings by the scientific community, regulatory agencies, healthcare providers, and the general public. AIMS This article aims to provide an overview of the key concepts associated with evaluating the biocompatibility of biomaterials while also offering practical guidance on cellular principles, testing methodologies, and biological assays that can support in the planning, execution, and reporting of biocompatibility testing.
Collapse
Affiliation(s)
- Vinicius Rosa
- Faculty of Dentistry, National University of Singapore, Singapore; ORCHIDS: Oral Care Health Innovations and Designs Singapore, National University of Singapore, Singapore.
| | - Nikolaos Silikas
- Dental Biomaterials, Dentistry, The University of Manchester, Manchester, United Kingdom.
| | - Baiqing Yu
- Faculty of Dentistry, National University of Singapore, Singapore.
| | - Nileshkumar Dubey
- ORCHIDS: Oral Care Health Innovations and Designs Singapore, National University of Singapore, Singapore; Division of Cariology and Operative Dentistry, Department of Comprehensive Dentistry, University of Maryland School of Dentistry, Baltimore, United States.
| | - Gopu Sriram
- Faculty of Dentistry, National University of Singapore, Singapore; ORCHIDS: Oral Care Health Innovations and Designs Singapore, National University of Singapore, Singapore.
| | - Spiros Zinelis
- School of Dentistry National and Kapodistrian University of Athens (NKUA), Greece.
| | - Adriano F Lima
- Dental Research Division, Paulista University, Sao Paulo, Brazil.
| | - Marco C Bottino
- School of Dentistry, University of Michigan, Ann Arbor, USA.
| | - Joao N Ferreira
- Center of Excellence for Innovation for Oral Health and Healthy Longevity, Faculty of Dentistry, Chulalongkorn University, Thailand.
| | - Gottfried Schmalz
- Department of Conservative Dentistry and Periodontology, University Hospital Regensburg, Regensburg, Germany; Department of Periodontology, School of Dental Medicine, University of Bern, Bern, Switzerland.
| | - David C Watts
- School of Medical Sciences and Photon Science Institute, University of Manchester, United Kingdom.
| |
Collapse
|
7
|
Akoa DM, Hélary C, Foda A, Chaussain C, Poliard A, Coradin T. Silicon impacts collagen remodelling and mineralization by human dental pulp stem cells in 3D pulp-like matrices. Dent Mater 2024; 40:1390-1399. [PMID: 38908960 DOI: 10.1016/j.dental.2024.06.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 06/14/2024] [Accepted: 06/14/2024] [Indexed: 06/24/2024]
Abstract
OBJECTIVES Silicon-releasing biomaterials are widely used in the field of dentistry. However, unlike bone, very little is known about the role of silicon on dental tissue formation and repair. This study investigates the influence of silicic acid on the survival, differentiation and mineralizing ability of human dental pulp stem cells (hDPSCs) in 3D pulp-like environments METHODS: Dense type I collagen hydrogels seeded with hDPSCs were cultured over 4 weeks in the presence of silicic acid at physiological (10 μM) and supraphysiological (100 μM) concentrations. Cell viability and proliferation were studied by Alamar Blue and live/dead staining. The collagen network was investigated using second harmonic generation imaging. Mineral deposition was monitored by histology and scanning electron microscopy. Gene expression of mineralization- and matrix remodeling-associated proteins was studied by qPCR. RESULTS Presence of silicic acid did not show any significant influence on cell survival, metabolic activity and gene expression of key mineralization-related proteins (ALP, OCN, BSP). However, it induced enhanced cell clustering and delayed expression of matrix remodeling-associated proteins (MMP13, Col I). OPN expression and mineral deposition were inhibited at 100 μM. It could be inferred that silicic acid has no direct cellular effect but rather interacts with the collagen network, leading to a modification of the cell-matrix interface. SIGNIFICANCE Our results offer advanced insights on the possible role of silicic acid, as released by pulp capping calcium silicates biomaterials, in reparative dentine formation. More globally, these results interrogate the possible role of Si in pulp pathophysiology.
Collapse
Affiliation(s)
- Daline Mbitta Akoa
- Sorbonne Université, CNRS, Laboratoire de Chimie de la Matière Condensée de Paris, Paris, France
| | - Christophe Hélary
- Sorbonne Université, CNRS, Laboratoire de Chimie de la Matière Condensée de Paris, Paris, France
| | - Asmaa Foda
- Université de Paris Cité, UR2496 Pathologies, Imagerie et Biothérapies Orofaciales, FHU-DDS-net, Dental School, Montrouge, France
| | - Catherine Chaussain
- Université de Paris Cité, UR2496 Pathologies, Imagerie et Biothérapies Orofaciales, FHU-DDS-net, Dental School, Montrouge, France; AP-HP Service de médecine bucco-dentaire, Hôpital Bretonneau, Paris, France
| | - Anne Poliard
- Université de Paris Cité, UR2496 Pathologies, Imagerie et Biothérapies Orofaciales, FHU-DDS-net, Dental School, Montrouge, France
| | - Thibaud Coradin
- Sorbonne Université, CNRS, Laboratoire de Chimie de la Matière Condensée de Paris, Paris, France.
| |
Collapse
|
8
|
Zhang H, Li L, Sun X, Hou B, Luo C. Research and development of microenvironment's influence on stem cells from the apical papilla - construction of novel research microdevices: tooth-on-a-chip. Biomed Microdevices 2024; 26:33. [PMID: 39023652 DOI: 10.1007/s10544-024-00715-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/29/2024] [Indexed: 07/20/2024]
Abstract
Stem cells are crucial in tissue engineering, and their microenvironment greatly influences their behavior. Among the various dental stem cell types, stem cells from the apical papilla (SCAPs) have shown great potential for regenerating the pulp-dentin complex. Microenvironmental cues that affect SCAPs include physical and biochemical factors. To research optimal pulp-dentin complex regeneration, researchers have developed several models of controlled biomimetic microenvironments, ranging from in vivo animal models to in vitro models, including two-dimensional cultures and three-dimensional devices. Among these models, the most powerful tool is a microfluidic microdevice, a tooth-on-a-chip with high spatial resolution of microstructures and precise microenvironment control. In this review, we start with the SCAP microenvironment in the regeneration of pulp-dentin complexes and discuss research models and studies related to the biological process.
Collapse
Affiliation(s)
- Hexuan Zhang
- Center for Microscope Enhanced Dentistry, School of Stomatology, Capital Medical University, Beijing, China
- Department of Endodontics and Operative Dentistry, School of Stomatology, Capital Medical University, Beijing, China
| | - Lingjun Li
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China
| | - Xiaoqiang Sun
- Department of Endodontics and Operative Dentistry, School of Stomatology, Capital Medical University, Beijing, China.
| | - Benxiang Hou
- Center for Microscope Enhanced Dentistry, School of Stomatology, Capital Medical University, Beijing, China.
| | - Chunxiong Luo
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China.
- The State Key Laboratory for Artificial Microstructures and Mesoscopic Physics, School of Physics, Peking University, Beijing, China.
| |
Collapse
|
9
|
Zhang H, Li L, Wang S, Sun X, Luo C, Hou B. Construction of dentin-on-a-chip based on microfluidic technology and tissue engineering. J Dent 2024; 146:105028. [PMID: 38719135 DOI: 10.1016/j.jdent.2024.105028] [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: 02/05/2024] [Revised: 04/23/2024] [Accepted: 04/25/2024] [Indexed: 05/19/2024] Open
Abstract
AIM Three-dimensional (3D) cell culture systems perform better in resembling tissue or organism structures compared with traditional 2D models. Organs-on-chips (OoCs) are becoming more efficient 3D models. This study aimed to create a novel simplified dentin-on-a-chip using microfluidic chip technology and tissue engineering for screening dental materials. METHODOLOGY A microfluidic device with three channels was designed for creating 3D dental tissue constructs using stem cells from the apical papilla (SCAP) and gelatin methacrylate (GelMA). The study investigated the effect of varying cell densities and GelMA concentrations on the layer features formed within the microfluidic chip. Cell viability and distribution were evaluated through live/dead staining and nuclei/F-actin staining. The osteo/odontogenic potential was assessed through ALP staining and Alizarin red staining. The impact of GelMA concentrations (5 %, 10 %) on the osteo/odontogenic differentiation trajectory of SCAP was also studied. RESULTS The 3D tissue constructs maintained high viability and favorable spreading within the microfluidic chip for 3-7 days. A cell seeding density of 2 × 104 cells/μL was found to be the most optimal choice, ensuring favorable cell proliferation and even distribution. GelMA concentrations of 5 % and 10 % proved to be most effective for promoting cell growth and uniform distribution. Within the 5 % GelMA group, SCAP demonstrated higher osteo/odontogenic differentiation than that in the 10 % GelMA group. CONCLUSION In 3D culture, GelMA concentration was found to regulate the osteo/odontogenic differentiation of SCAP. The study recommends a seeding density of 2 × 104 cells/μL of SCAP within 5 % GelMA for constructing simplified dentin-on-a-chip. CLINICAL SIGNIFICANCE This study built up the 3D culture protocol, and induced odontogenic differentiation of SCAP, thus forming the simplified dentin-on-a-chip and paving the way to be used as a well-defined biological model for regenerative endodontics. It may serve as a potential testing platform for cell differentiation.
Collapse
Affiliation(s)
- Hexuan Zhang
- Center for Microscope Enhanced Dentistry, School of Stomatology, Capital Medical University, Beijing 100162, PR China; Department of Endodontics and Operative Dentistry, School of Stomatology, Capital Medical University, Beijing 100050, PR China
| | - Lingjun Li
- Wenzhou Institute University of Chinese Academy of Sciences, Wenzhou 325001, PR China.
| | - Shujing Wang
- Wenzhou Institute University of Chinese Academy of Sciences, Wenzhou 325001, PR China
| | - Xiaoqiang Sun
- Department of Endodontics and Operative Dentistry, School of Stomatology, Capital Medical University, Beijing 100050, PR China
| | - Chunxiong Luo
- Wenzhou Institute University of Chinese Academy of Sciences, Wenzhou 325001, PR China; The State Key Laboratory for Artificial Microstructures and Mesoscopic Physics, School of Physics, Peking University, Beijing 100871, PR China.
| | - Benxiang Hou
- Center for Microscope Enhanced Dentistry, School of Stomatology, Capital Medical University, Beijing 100162, PR China.
| |
Collapse
|
10
|
Zheng L, Zhang Y, Bai Y, Zhang Z, Wu Q. Study on the mechanical and aging properties of an antibacterial composite resin loaded with fluoride-doped nano-zirconia fillers. Front Bioeng Biotechnol 2024; 12:1397459. [PMID: 38846803 PMCID: PMC11153679 DOI: 10.3389/fbioe.2024.1397459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 04/29/2024] [Indexed: 06/09/2024] Open
Abstract
Preventing the occurrence of secondary caries serves as one of the significant issues in dental clinic, thus make it indispensable to improving the properties of conventional composite resin (CR) by developing a novel CR. In present study, two groups of experimental CRs loaded with different contents of fluoride-doped nano-zirconia fillers (25 wt% and 50 wt%) were fabricated. The surface topography, mechanical performance, fluoride release, antibacterial effect, aging property and cytotoxicity of the experimental CRs were evaluated subsequently. A uniform distribution of the F-zirconia fillers over the whole surface of resin matrix could be observed. The experimental CRs showed continuous fluoride release within 28 days, which was positively correlated with the content of F-zirconia fillers. Moreover, the amount of fluoride release increased in the acidic buffer. Addition of F-zirconia fillers could improve the color stability, wear resistance and microhardness of the experimental CRs, without reducing the flexure strength. Furtherly, the fluoride ions released continuously from the experimental CRs resulted in effective contact and antibacterial properties, while they showed no cytotoxicity. As a consequence, considerations can be made to employ this new kind of composite resin loaded with fluoride-doped nano-zirconia fillers to meet clinical requirements when the antimicrobial benefits are desired.
Collapse
Affiliation(s)
- Liyuan Zheng
- Department of Prosthodontics, Stomatological Hospital of Xiamen Medical College, Xiamen Key Laboratory of Stomatological Disease Diagnosis and Treatment, Xiamen, China
| | - Yi Zhang
- Department of Prosthodontics, Stomatological Hospital of Xiamen Medical College, Xiamen Key Laboratory of Stomatological Disease Diagnosis and Treatment, Xiamen, China
| | - Yuming Bai
- Department of Orthodontics, Stomatological Hospital of Xiamen Medical College, Xiamen Key Laboratory of Stomatological Disease Diagnosis and Treatment, Xiamen, China
| | - Zhisheng Zhang
- Department of Prosthodontics, Stomatological Hospital of Xiamen Medical College, Xiamen Key Laboratory of Stomatological Disease Diagnosis and Treatment, Xiamen, China
| | - Qianju Wu
- Department of Prosthodontics, Stomatological Hospital of Xiamen Medical College, Xiamen Key Laboratory of Stomatological Disease Diagnosis and Treatment, Xiamen, China
| |
Collapse
|
11
|
Koutrouli A, Machla F, Arapostathis K, Kokoti M, Bakopoulou A. "Biological responses of two calcium-silicate-based cements on a tissue-engineered 3D organotypic deciduous pulp analogue". Dent Mater 2024; 40:e14-e25. [PMID: 38431482 DOI: 10.1016/j.dental.2024.02.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 02/14/2024] [Accepted: 02/18/2024] [Indexed: 03/05/2024]
Abstract
OBJECTIVES The biological responses of MTA and Biodentine™ has been assessed on a three-dimensional, tissue-engineered organotypic deciduous pulp analogue. METHODS Human endothelial (HUVEC) and dental mesenchymal stem cells (SHED) at a ratio of 3:1, were incorporated into a collagen I/fibrin hydrogel; succeeding Biodentine™ and MTA cylindrical specimens were placed in direct contact with the pulp analogue 48 h later. Cell viability/proliferation and morphology were evaluated through live/dead staining, MTT assay and Scanning Electron Microscopy (SEM), and expression of angiogenic, odontogenic markers through real time PCR. RESULTS Viable cells dominated at day 3 after treatment presenting typical morphology, firmly attached within the hydrogel structures, as shown by live/dead staining and SEM images. MTT assay at day 1 presented a significant increase of cell proliferation in Biodentine™ group. Real-time PCR showed significant upregulation of odontogenic markers DSPP, BMP-2 (day 3,6), RUNX2, ALP (day 3) in contact with Biodentine™ compared to MTA and the control, whereas MTA promoted significant upregulation of DSPP, BMP-2, RUNX2, Osterix (day 3) and ALP (day 6) compared to the control. MSX1 presented downregulation in both experimental groups. Expression of angiogenic markers VEGFa and ANGPT-1 at day 3 was significantly upregulated in contact with Biodentine™ and MTA respectively, while the receptors VEGFR1, VEGFR2 and Tie-2, as well as PECAM-1 were downregulated. SIGNIFICANCE Both calcium silicate-based materials are biocompatible and exert positive angiogenic and odontogenic effects, although Biodentine™ during the first days of culture, seems to induce higher cell proliferation and provoke a more profound odontogenic and angiogenic response from SHED.
Collapse
Affiliation(s)
- A Koutrouli
- Department of Paediatric Dentistry, School of Dentistry, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki GR-54124, Greece
| | - F Machla
- Department of Prosthodontics, School of Dentistry, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki GR-54124, Greece
| | - K Arapostathis
- Department of Paediatric Dentistry, School of Dentistry, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki GR-54124, Greece
| | - M Kokoti
- Department of Prosthodontics, School of Dentistry, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki GR-54124, Greece
| | - A Bakopoulou
- Department of Prosthodontics, School of Dentistry, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki GR-54124, Greece.
| |
Collapse
|
12
|
López-García S, Aznar-Cervantes SD, Pagán A, Llena C, Forner L, Sanz JL, García-Bernal D, Sánchez-Bautista S, Ceballos L, Fuentes V, Melo M, Rodríguez-Lozano FJ, Oñate-Sánchez RE. 3D Graphene/silk fibroin scaffolds enhance dental pulp stem cell osteo/odontogenic differentiation. Dent Mater 2024; 40:431-440. [PMID: 38114344 DOI: 10.1016/j.dental.2023.12.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 09/24/2023] [Accepted: 12/11/2023] [Indexed: 12/21/2023]
Abstract
OBJECTIVES The current in vitro study aims to evaluate silk fibroin with and without the addition of graphene as a potential scaffold material for regenerative endodontics. MATERIAL AND METHODS Silk fibroin (SF), Silk fibroin/graphene oxide (SF/GO) and silk fibroin coated with reduced graphene oxide (SF/rGO) scaffolds were prepared (n = 30). The microarchitectures and mechanical properties of scaffolds were evaluated using field emission scanning electron microscopy (FESEM), pore size and water uptake, attenuated total reflectance fourier transformed infrared spectroscopy (ATR-FTIR), Raman spectroscopy and mechanical compression tests. Next, the study analyzed the influence of these scaffolds on human dental pulp stem cell (hDPSC) viability, apoptosis or necrosis, cell adhesion, odontogenic differentiation marker expression and mineralized matrix deposition. The data were analyzed with ANOVA complemented with the Tukey post-hoc test (p < 0.005). RESULTS SEM analysis revealed abundant pores with a size greater than 50 nm on the surface of tested scaffolds, primarily between 50 nm and 600 µm. The average value of water uptake obtained in pure fibroin scaffolds was statistically higher than that of those containing GO or rGO (p < 0.05). ATR-FTIR evidenced that the secondary structures did not present differences between pure fibroin and fibroin coated with graphene oxide, with a similar infrared spectrum in all tested scaffolds. Raman spectroscopy showed a greater number of defects in the links in SF/rGO scaffolds due to the reduction of graphene. In addition, adequate mechanical properties were exhibited by the tested scaffolds. Regarding biological properties, hDPSCs attached to scaffolds were capable of proliferating at a rate similar to the control, without affecting their viability over time. A significant upregulation of ALP, ON and DSPP markers was observed with SF/rGO and SF/GO groups. Finally, SF/GO and SF/rGO promoted a significantly higher mineralization than the control at 21 days. SIGNIFICANCE Data obtained suggested that SF/GO and SF/rGO scaffolds promote hDPSC differentiation at a genetic level, increasing the expression of key osteo/odontogenic markers, and supports the mineralization of the extracellular matrix. However, results from this study are to be interpreted with caution, requiring further in vivo studies to confirm the potential of these scaffolds.
Collapse
Affiliation(s)
- Sergio López-García
- Departament d'Estomatologia, Facultat de Medicina I Odontologia, Universitat de València, Valencia 46010, Spain
| | - Salvador D Aznar-Cervantes
- Biotechnology, Genomics and PlantBreedingDepartment, Instituto Murciano de Investigación y Desarrollo Agrario y Ambiental (IMIDA), La Alberca 30150, Murcia, Spain
| | - Ana Pagán
- Biotechnology, Genomics and PlantBreedingDepartment, Instituto Murciano de Investigación y Desarrollo Agrario y Ambiental (IMIDA), La Alberca 30150, Murcia, Spain
| | - Carmen Llena
- Departament d'Estomatologia, Facultat de Medicina I Odontologia, Universitat de València, Valencia 46010, Spain
| | - Leopoldo Forner
- Departament d'Estomatologia, Facultat de Medicina I Odontologia, Universitat de València, Valencia 46010, Spain
| | - José L Sanz
- Departament d'Estomatologia, Facultat de Medicina I Odontologia, Universitat de València, Valencia 46010, Spain
| | - David García-Bernal
- Department of Biochemistry, Molecular Biology B and Immunology, Faculty of Medicine, University of Murcia, Biomedical Research Institute (IMIB), Murcia 30120, Spain
| | | | - Laura Ceballos
- IDIBO Research Group, Area of Stomatology, Health Sciences Faculty, Rey Juan Carlos University, Alcorcón, Madrid, Spain
| | - Victoria Fuentes
- IDIBO Research Group, Area of Stomatology, Health Sciences Faculty, Rey Juan Carlos University, Alcorcón, Madrid, Spain
| | - María Melo
- Departament d'Estomatologia, Facultat de Medicina I Odontologia, Universitat de València, Valencia 46010, Spain
| | - Francisco J Rodríguez-Lozano
- Department of Dermatology, Stomatology, Radiology and Physical Medicine, Morales Meseguer Hospital, Biomedical Research Institute (IMIB), Regional Campus of International Excellence "Campus Mare Nostrum", Faculty of Medicine, University of Murcia, Murcia 30008, Spain.
| | - Ricardo E Oñate-Sánchez
- Department of Dermatology, Stomatology, Radiology and Physical Medicine, Morales Meseguer Hospital, Biomedical Research Institute (IMIB), Regional Campus of International Excellence "Campus Mare Nostrum", Faculty of Medicine, University of Murcia, Murcia 30008, Spain
| |
Collapse
|
13
|
Liu C, Staples R, Gómez-Cerezo MN, Ivanovski S, Han P. Emerging Technologies of Three-Dimensional Printing and Mobile Health in COVID-19 Immunity and Regenerative Dentistry. Tissue Eng Part C Methods 2023; 29:163-182. [PMID: 36200626 DOI: 10.1089/ten.tec.2022.0160] [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: 11/13/2022] Open
Abstract
The ongoing coronavirus disease 2019 (COVID-19) pandemic highlights the importance of developing point-of-care (POC) antibody tests for monitoring the COVID-19 immune response upon viral infection or following vaccination, which requires three key aspects to achieve optimal monitoring, including three-dimensional (3D)-printed POC devices, mobile health (mHealth), and noninvasive sampling. As a critical tissue engineering concept, additive manufacturing (AM, also known as 3D printing) enables accurate control over the dimensional and architectural features of the devices. mHealth refers to the use of portable digital devices, such as smartphones, tablet computers, and fitness and medical wearables, to support health, which facilitates contact tracing, and telehealth consultations during the pandemic. Compared with invasive biosample (blood), saliva is of great importance in the spread and surveillance of COVID-19 as a noninvasive diagnostic method for virus detection and immune status monitoring. However, investigations into 3D-printed POC antibody test and mHealth using noninvasive saliva are relatively limited. Further exploration of 3D-printed antibody POC tests and mHealth applications to monitor antibody production for either disease onset or immune response following vaccination is warranted. This review briefly describes the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus and immune response after infection and vaccination, then discusses current widely used binding antibody tests using blood samples and enzyme-linked immunosorbent assays on two-dimensional microplates before focusing upon emerging POC technological platforms, such as field-effect transistor biosensors, lateral flow assay, microfluidics, and AM for fabricating immunoassays, and the possibility of their combination with mHealth. This review proposes that noninvasive biofluid sampling combined with 3D POC antibody tests and mHealth technologies is a promising and novel approach for POC detection and surveillance of SARS-CoV-2 immune response. Furthermore, as key concepts in dentistry, the application of 3D printing and mHealth was also included to facilitate the appreciation of cutting edge techniques in regenerative dentistry. This review highlights the potential of 3D printing and mHealth in both COVID-19 immunity monitoring and regenerative dentistry.
Collapse
Affiliation(s)
- Chun Liu
- School of Dentistry, The University of Queensland, Brisbane, Queensland, Australia
- Center for Oral-Facial Regeneration, Rehabilitation and Reconstruction (COR3), School of Dentistry, The University of Queensland, Brisbane, Queensland, Australia
| | - Reuben Staples
- Center for Oral-Facial Regeneration, Rehabilitation and Reconstruction (COR3), School of Dentistry, The University of Queensland, Brisbane, Queensland, Australia
| | - Maria Natividad Gómez-Cerezo
- Center for Oral-Facial Regeneration, Rehabilitation and Reconstruction (COR3), School of Dentistry, The University of Queensland, Brisbane, Queensland, Australia
| | - Sašo Ivanovski
- School of Dentistry, The University of Queensland, Brisbane, Queensland, Australia
- Center for Oral-Facial Regeneration, Rehabilitation and Reconstruction (COR3), School of Dentistry, The University of Queensland, Brisbane, Queensland, Australia
| | - Pingping Han
- School of Dentistry, The University of Queensland, Brisbane, Queensland, Australia
- Center for Oral-Facial Regeneration, Rehabilitation and Reconstruction (COR3), School of Dentistry, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
14
|
Kunrath MF, Shah FA, Dahlin C. Bench-to-bedside: Feasibility of nano-engineered and drug-delivery biomaterials for bone-anchored implants and periodontal applications. Mater Today Bio 2023; 18:100540. [PMID: 36632628 PMCID: PMC9826856 DOI: 10.1016/j.mtbio.2022.100540] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 12/03/2022] [Accepted: 12/29/2022] [Indexed: 12/31/2022] Open
Abstract
Nanotechnology and drug-release biomaterials have been thoroughly explored in the last few years aiming to develop specialized clinical treatments. However, it is rare to find biomaterials associated with drug delivery properties in the current dental market for application in oral bone- and periodontal-related procedures. The gap between basic scientific evidence and translation to a commercial product remains wide. Several challenges have been reported regarding the clinical translation of biomaterials with drug-delivery systems (BDDS) and nanofeatures. Therefore, processes for BDDS development, application in preclinical models, drug delivery doses, sterilization processes, storage protocols and approval requirements were explored in this review, associated with tentative solutions for these issues. The diversity of techniques and compounds/molecules applied to develop BDDS demands a case-by-case approach to manufacturing and validating a commercial biomaterial. Promising outcomes such as accelerated tissue healing and higher antibacterial response have been shown through basic and preclinical studies using BDDS and nano-engineered biomaterials; however, the adequate process for sterilization, storage, cost-effectiveness and possible cytotoxic effects remains unclear for multifunctional biomaterials incorporated with different chemical compounds; then BDDSs are rarely translated into products. The future benefits of BDDS and nano-engineered biomaterials have been reported suggesting personalized clinical treatment and a promising reduction in the use of systemic antibiotics. Finally, the launch of these specialized biomaterials with solid data and controlled traceability onto the market will generate strong specificity for healthcare treatments.
Collapse
Affiliation(s)
- Marcel F. Kunrath
- Department of Biomaterials, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, P.O. Box 412, SE 405 30, Göteborg, Sweden
- Department of Dentistry, School of Health and Life Sciences, Pontifical Catholic University of Rio Grande Do Sul (PUCRS), Porto Alegre, Brazil
| | - Furqan A. Shah
- Department of Biomaterials, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, P.O. Box 412, SE 405 30, Göteborg, Sweden
| | - Christer Dahlin
- Department of Biomaterials, Institute of Clinical Sciences, Sahlgrenska Academy at University of Gothenburg, P.O. Box 412, SE 405 30, Göteborg, Sweden
| |
Collapse
|
15
|
Anada R, Hara ES, Nagaoka N, Okada M, Kamioka H, Matsumoto T. Important roles of odontoblast membrane phospholipids in early dentin mineralization. J Mater Chem B 2023; 11:657-666. [PMID: 36541228 DOI: 10.1039/d2tb02351b] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The objective of this study was to first identify the timing and location of early mineralization of mouse first molar, and subsequently, to characterize the nucleation site for mineral formation in dentin from a materials science viewpoint and evaluate the effect of environmental cues (pH) affecting early dentin formation. Early dentin mineralization in mouse first molars began in the buccal central cusp on post-natal day 0 (P0), and was first hypothesized to involve collagen fibers. However, elemental mapping indicated the co-localization of phospholipids with collagen fibers in the early mineralization area. Co-localization of phosphatidylserine and annexin V, a functional protein that binds to plasma membrane phospholipids, indicated that phospholipids in the pre-dentin matrix were derived from the plasma membrane. A 3-dimensional in vitro biomimetic mineralization assay confirmed that phospholipids from the plasma membrane are critical factors initiating mineralization. Additionally, the direct measurement of the tooth germ pH, indicated it to be alkaline. The alkaline environment markedly enhanced the mineralization of cell membrane phospholipids. These results indicate that cell membrane phospholipids are nucleation sites for mineral formation, and could be important materials for bottom-up approaches aiming for rapid and more complex fabrication of dentin-like structures.
Collapse
Affiliation(s)
- Risa Anada
- Department of Biomaterials, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan. .,Department of Orthodontics, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Emilio Satoshi Hara
- Department of Biomaterials, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan.
| | - Noriyuki Nagaoka
- Advanced Research Center for Oral and Craniofacial Sciences, Dental School, Okayama University, Okayama, Japan
| | - Masahiro Okada
- Department of Biomaterials, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan.
| | - Hiroshi Kamioka
- Department of Orthodontics, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Takuya Matsumoto
- Department of Biomaterials, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan.
| |
Collapse
|
16
|
Rothermund K, Calabrese TC, Syed-Picard FN. Differential Effects of Escherichia coli- Versus Porphyromonas gingivalis-derived Lipopolysaccharides on Dental Pulp Stem Cell Differentiation in Scaffold-free Engineered Tissues. J Endod 2022; 48:1378-1386.e2. [PMID: 36108879 PMCID: PMC9764159 DOI: 10.1016/j.joen.2022.08.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 08/30/2022] [Accepted: 08/30/2022] [Indexed: 11/30/2022]
Abstract
INTRODUCTION To leverage the therapeutic capabilities of dental pulp stem cells (DPSCs) for regenerative endodontic applications, a better understanding of their innate defense and reparative processes is needed. Lipopolysaccharide (LPS) is a major virulent factor of gram-negative bacteria and contributor to endodontic infections. We have developed 3-dimensional scaffold-free DPSC tissues that self-organize into dentin-pulp organoids comprising a mineralized dentin-like tissue on the periphery and an unmineralized pulp-like core. In this study, scaffold-free DPSC constructs were used as controllable experimental models to study the DPSC response to bacterial challenge. METHODS Scaffold-free constructs were engineered using DPSCs isolated from human third molars. To simulate bacterial exposure, DPSC constructs were exposed to either Porphyromonas gingivalis-derived LPS or Escherichia coli-derived LPS. The effects of LPS on DPSC differentiation, proliferation, and apoptosis were evaluated. RESULTS Engineered tissues lacking LPS treatment self-organized into dentin-pulp organoids. LPS treatment did not negatively affect DPSC proliferation or apoptosis in the engineered tissues. Both E. coli LPS and P. gingivalis LPS inhibited the up-regulation of RUNX2 messenger RNA expression and reduced the expression of the odontoblast-associated proteins (P < .05), suggesting that LPS is inhibiting odontoblastic differentiation. However, only E. coli LPS treatment significantly reduced mineral deposition in the DPSC (P < .05) constructs, indicating that E. coli LPS but not P. gingivalis LPS reduced functional differentiation of DPSCs and prevented DPSCs from self-organizing into a dentin-pulp complex-like structure. CONCLUSIONS This study establishes scaffold-free DPSC constructs as models of oral disease. Furthermore, it emphasizes the diversity of LPS derived from different bacterial species and highlights the necessity of using LPS derived from clinically relevant bacteria in basic science investigations.
Collapse
Affiliation(s)
- Kristi Rothermund
- Center for Craniofacial Regeneration, School of Dental Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Tia C Calabrese
- Center for Craniofacial Regeneration, School of Dental Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Fatima N Syed-Picard
- Center for Craniofacial Regeneration, School of Dental Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania; Department of Oral and Craniofacial Sciences, School of Dental Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania; McGowan Institute for Regenerative Medicine, Pittsburgh, Pennsylvania.
| |
Collapse
|
17
|
Franca CM, Balbinot GDS, Cunha D, Saboia VDPA, Ferracane J, Bertassoni LE. In-vitro models of biocompatibility testing for restorative dental materials: From 2D cultures to organs on-a-chip. Acta Biomater 2022; 150:58-66. [PMID: 35933103 PMCID: PMC9814917 DOI: 10.1016/j.actbio.2022.07.060] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 07/13/2022] [Accepted: 07/28/2022] [Indexed: 02/08/2023]
Abstract
Dental caries is a biofilm-mediated, diet-modulated, multifactorial and dynamic disease that affects more than 90% of adults in Western countries. The current treatment for decayed tissue is based on using materials to replace the lost enamel or dentin. More than 500 million dental restorations are placed annually worldwide, and materials used for these purposes either directly or indirectly interact with dentin and pulp tissues. The development and understanding of the effects of restorative dental materials are based on different in-vitro and in-vivo tests, which have been evolving with time. In this review, we first discuss the characteristics of the tooth and the dentin-pulp interface that are unique for materials testing. Subsequently, we discuss frequently used in-vitro tests to evaluate the biocompatibility of dental materials commonly used for restorative procedures. Finally, we present our perspective on the future directions for biological research on dental materials using tissue engineering and organs on-a-chip approaches. STATEMENT OF SIGNIFICANCE: Dental caries is still the most prevalent infectious disease globally, requiring more than 500 million restorations to be placed every year. Regrettably, the failure rates of such restorations are still high. Those rates are partially based on the fact that current platforms to test dental materials are somewhat inaccurate in reproducing critical components of the complex oral microenvironment. Thus, there is a collective effort to develop new materials while evolving the platforms to test them. In this context, the present review critically discusses in-vitro models used to evaluate the biocompatibility of restorative dental materials and brings a perspective on future directions for tissue-engineered and organs-on-a-chip platforms for testing new dental materials.
Collapse
Affiliation(s)
- Cristiane Miranda Franca
- Department of Restorative Dentistry, School of Dentistry, Oregon Health & Science University, Portland, OR, United States
| | - Gabriela de Souza Balbinot
- Dental Materials Laboratory, School of Dentistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Diana Cunha
- Post-Graduation Program in Dentistry, Federal University of Ceará, Fortaleza, Ceará, Brazil
| | | | - Jack Ferracane
- Department of Restorative Dentistry, School of Dentistry, Oregon Health & Science University, Portland, OR, United States
| | - Luiz E Bertassoni
- Department of Restorative Dentistry, School of Dentistry, Oregon Health & Science University, Portland, OR, United States; Center for Regenerative Medicine, School of Medicine, Oregon Health & Science University, Portland, OR, United States; Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, OR, United States; Cancer Early Detection Advanced Research Center (CEDAR), Knight Cancer Institute, Portland, OR, United States.
| |
Collapse
|
18
|
Hu S, Muniraj G, Mishra A, Hong K, Lum JL, Hong CHL, Rosa V, Sriram G. Characterization of silver diamine fluoride cytotoxicity using microfluidic tooth-on-a-chip and gingival equivalents. Dent Mater 2022; 38:1385-1394. [PMID: 35778310 DOI: 10.1016/j.dental.2022.06.025] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 06/16/2022] [Accepted: 06/22/2022] [Indexed: 01/18/2023]
Abstract
OBJECTIVE This study aims to characterize the cytotoxicity potential of silver diamine fluoride (SDF) on dental pulp stem cells (DPSC) and gingival equivalents. METHODS DPSC cultured on 96-well plates was exposed directly to SDF (0.0001-0.01%) and cell viability (IC50) quantified. Effect of SDF on DPSC viability under flow (with dentin barrier) conditions was evaluated using a custom-designed microfluidic "tooth-on-a-chip". Permeability of dentin discs (0.5-1.5 mm thickness) was evaluated using lucifer yellow permeation assay. Dentin discs were treated with 38% SDF (up to 3 h), and cell viability (live/dead assay) of the DPSC cultured in the inlet (unexposed) and outlet (exposed) regions of the pulp channel was evaluated. To assess the mucosal corrosion potential, gingival equivalents were treated with 38% SDF for 3 or 60 min (OECD test guideline 431) and characterized by MTT assay and histomorphometric analysis. RESULTS DPSC exposed directly to SDF showed a dose-dependent reduction in cell viability (IC50: 0.001%). Inlet channels (internal control) of the tooth-on-a-chip exposed to PBS and SDF-exposed dentin discs showed> 85% DPSC viability. In contrast, the outlet channels of SDF-exposed dentin discs showed a decreased viability of< 31% and 0% (1.5 and ≤1.0 mm thick dentin disc, respectively) (p < 0.01). The gingiva equivalents treated with SDF for 3 and 60 min demonstrated decreased epithelial integrity, loss of intercellular cohesion and corneal layer detachment with significant reduction in intact epithelial thickness (p < 0.05). SIGNIFICANCE SDF penetrated the dentin (≤1 mm thick) inducing significant death of the pulp cells. SDF also disrupted gingival epithelial integrity resulting in mucosal corrosion.
Collapse
Affiliation(s)
- Shijia Hu
- Faculty of Dentistry, National University of Singapore, Singapore.
| | | | - Apurva Mishra
- Faculty of Dentistry, National University of Singapore, Singapore
| | - Kanglun Hong
- National University Centre for Oral Health Singapore, National University Hospital, Singapore
| | - Jing Li Lum
- National University Centre for Oral Health Singapore, National University Hospital, Singapore
| | | | - Vinicius Rosa
- Faculty of Dentistry, National University of Singapore, Singapore; ORCHIDS: Oral Care Health Innovations and Designs Singapore, National University of Singapore, Singapore.
| | - Gopu Sriram
- Faculty of Dentistry, National University of Singapore, Singapore; ORCHIDS: Oral Care Health Innovations and Designs Singapore, National University of Singapore, Singapore.
| |
Collapse
|
19
|
Rosa V, Sriram G, McDonald N, Cavalcanti BN. A critical analysis of research methods and biological experimental models to study pulp regeneration. Int Endod J 2022; 55 Suppl 2:446-455. [PMID: 35218576 PMCID: PMC9311820 DOI: 10.1111/iej.13712] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 02/23/2022] [Accepted: 02/24/2022] [Indexed: 12/01/2022]
Abstract
With advances in knowledge and treatment options, pulp regeneration is now a clear objective in clinical dental practice. For this purpose, many methodologies have been developed in attempts to address the putative questions raised both in research and in clinical practice. In the first part of this review, laboratory‐based methods will be presented, analysing the advantages, disadvantages, and benefits of cell culture methodologies and ectopic/semiorthotopic animal studies. This will also demonstrate the need for alignment between two‐dimensional and three‐dimensional laboratory techniques to accomplish the range of objectives in terms of cell responses and tissue differentiation. The second part will cover observations relating to orthotopic animal studies, describing the current models used for this purpose and how they contribute to the translation of regenerative techniques to the clinic.
Collapse
Affiliation(s)
- Vinicius Rosa
- Faculty of Dentistry, National University of Singapore, Singapore
| | - Gopu Sriram
- Faculty of Dentistry, National University of Singapore, Singapore
| | - Neville McDonald
- Department of Cariology, Restorative Sciences and Endodontics, University of Michigan School of Dentistry, Ann Arbor, MI, USA
| | - Bruno Neves Cavalcanti
- Department of Cariology, Restorative Sciences and Endodontics, University of Michigan School of Dentistry, Ann Arbor, MI, USA
| |
Collapse
|
20
|
Hadjichristou C, About I, Koidis P, Bakopoulou A. Advanced in Vitro Experimental Models for Tissue Engineering-based Reconstruction of a 3D Dentin/pulp Complex: a Literature Review. Stem Cell Rev Rep 2020; 17:785-802. [PMID: 33145672 DOI: 10.1007/s12015-020-10069-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/25/2020] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Experimental procedures have been used to monitor cellular responses at the dentin/pulp interface. Aiming to divert from in vivo studies and oversimplified two-dimensional assays, three-dimensional (3D) models have been developed. This review provides an overview of existing literature, regarding 3D in vitro dentin/pulp reconstruction. MATERIAL & METHODS PubMed, Scopus, Cochrane Library and Web of Science- were systematically searched for attributes between 1998 and 2020. The search focused on articles on the development of three-dimensional tools for the reconstruction of a dentin/pulp complex under in vitro conditions, which were then screened and qualitatively assessed. Article grouping according to mode of implementation, resulted in five categories: the customised cell perfusion chamber (CPC) (n = 8), the tooth bud model (TBM) (n = 3), the 3D dentin/pulp complex manufactured by tissue engineering (DPC) (n = 6), the entire tooth culture (ETC) (n = 4) and the tooth slice culture model (TSC) (n = 5). RESULTS A total of 26 publications, applying nine and eight substances for pulp and dentin representation respectively, were included. Natural materials and dentin components were the most widely utilized. The most diverse category was the DPC, while the CPC group was the test with the highest longevity. The most consistent categories were the ETC and TSC models, while the TBM presented as the most complete de novo approach. CONCLUSIONS All studies presented with experimental protocols with potential upgrades. Solving the limitations of each category will provide a complete in vitro testing and monitoring tool of dental responses to exogenous inputs. CLINICAL RELEVANCE The 3D dentin/pulp complexes are valid supplementary tools for in vivo studies and clinical testing. Graphical Abstract.
Collapse
Affiliation(s)
- Christina Hadjichristou
- Department of Prosthodontics, School of Dentistry, Faculty of Health Sciences, Aristotle University of Thessaloniki (A.U.Th), GR-54124, Thessaloniki, Greece.
| | - Imad About
- Centre National de la Recherche Scientifique, Institute of Movement Sciences, Aix Marseille University, Marseille, France
| | - Petros Koidis
- Department of Prosthodontics, School of Dentistry, Faculty of Health Sciences, Aristotle University of Thessaloniki (A.U.Th), GR-54124, Thessaloniki, Greece
| | - Athina Bakopoulou
- Department of Prosthodontics, School of Dentistry, Faculty of Health Sciences, Aristotle University of Thessaloniki (A.U.Th), GR-54124, Thessaloniki, Greece
| |
Collapse
|
21
|
Hong H, Chen X, Li K, Wang N, Li M, Yang B, Yu X, Wei X. Dental follicle stem cells rescue the regenerative capacity of inflamed rat dental pulp through a paracrine pathway. Stem Cell Res Ther 2020; 11:333. [PMID: 32746910 PMCID: PMC7397608 DOI: 10.1186/s13287-020-01841-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 06/27/2020] [Accepted: 07/17/2020] [Indexed: 02/07/2023] Open
Abstract
Background Pulpitis is a common dental disease characterized by sustained inflammation and impaired pulp self-repair. Mesenchymal stem cell-based minimally invasive vital pulp therapy (MSC-miVPT) is a potential treatment method, but its application is limited by the difficulty in acquiring MSCs. We recently revealed the immunomodulatory effects of rat dental follicle stem cells (rDFSCs) on acute lung injury. The present study focused on the paracrine effects of rDFSCs on the inflammation and regeneration of rat injured dental pulp to detect whether DFSCs are a potential candidate for MSC-miVPT. Methods Conditioned medium from rDFSCs (rDFSC-CM) was applied to lipopolysaccharide (LPS)-induced inflammatory rat dental pulp cells (rDPCs). The inflammation and regeneration of rDPCs were detected by RT-qPCR, Western blotting, immunofluorescence staining, Cell Counting Kit-8 (CCK-8) assay, flow cytometry, wound-healing assay, and Masson’s staining. The effects of rDFSC-CM on inflamed rat dental pulp were further evaluated by hematoxylin-eosin and immunohistochemical staining. Results rDFSC-CM downregulated the ERK1/2 and NF-κB signaling pathways, which resulted in suppression of the expression of IL-1β, IL-6, and TNF-α and promotion of the expression of IL-4 and TGF-β, and these findings lead to the attenuation of rDPC inflammation. rDFSC-CM enhanced the in vitro proliferation, migration, and odontogenic differentiation of inflammatory rDPCs and their in vivo ectopic dentinogenesis. Furthermore, rDFSC-CM inhibited inflammatory cell infiltration in rat pulpitis and triggered Runx2 expression in some of the odontoblast-like cells surrounding the injured site, and these effects were conducive to the repair of inflamed dental pulp. Conclusions rDFSC-CM exhibits therapeutic potential by rescuing the regeneration of the inflamed rat dental pulp through an immunomodulatory mechanism, indicating the application prospects of DFSCs in biological regenerative endodontics.
Collapse
Affiliation(s)
- Hong Hong
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510055, People's Republic of China
| | - Xiaochuan Chen
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510055, People's Republic of China.,Department of Stomatology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, People's Republic of China
| | - Kun Li
- Key Laboratory of Green Chemistry and Technology, Ministry of Education, College of Chemistry, Sichuan University, Chengdu, 610064, People's Republic of China
| | - Nan Wang
- Key Laboratory of Green Chemistry and Technology, Ministry of Education, College of Chemistry, Sichuan University, Chengdu, 610064, People's Republic of China
| | - Mengjie Li
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510055, People's Republic of China
| | - Bo Yang
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510055, People's Republic of China
| | - Xiaoqi Yu
- Key Laboratory of Green Chemistry and Technology, Ministry of Education, College of Chemistry, Sichuan University, Chengdu, 610064, People's Republic of China.
| | - Xi Wei
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, 510055, People's Republic of China.
| |
Collapse
|