The Evolution of In Vitro Toxicity Assessment Methods for Oral Cavity Tissues—From 2D Cell Cultures to Organ-on-a-Chip
Abstract
:1. Introduction
2. Materials and Methods
3. Two-Dimensional Cell Culture Testing
3.1. Cell Viability and Cell Proliferation Assays
Method | Type of Cells | Tested Agent | Results | Reference |
---|---|---|---|---|
TUNEL assay | Human gingival fibroblasts | Hydrogen peroxide | Hydrogen peroxide promoted apoptosis in human gingival fibroblasts. | [78] |
Oral mucosa epithelial cells | Mobile phone use (radiofrequency radiation) | Apoptosis levels were not significantly influenced. | [79] | |
Human gingival fibroblasts | Nicotine | Nicotine increased the number of TUNEL-positive cells. | [80] | |
Human gingival epithelium keratinocytes | Traditional cigarette smoke and electronic cigarette aerosols | Exposure to traditional cigarette smoke and electronic cigarette aerosols increased the number of TUNEL-positive cells. | [39] | |
Human oral fibroblasts | Cigarette smoke and e-vapor condensates | The DNA fragmentation assay indicated an increased number of TUNEL-positive apoptotic fibroblasts with broken nuclei. | [40] | |
Human gingival epithelial cells | E-cigarette vapor | E-cigarette vapor exposure increased the proportion of TUNEL-positive cells. | [58] | |
Caspase activity assay | Human gingival fibroblasts | Nicotine | Nicotine induced apoptosis via the caspase-3 pathway. | [80] |
Human gingival epithelial cells | Cigarette smoke total particulate matter (TPM), smokeless tobacco extracted with complete artificial saliva (ST/CAS), and whole-smoke conditioned media (WS-CM). | TPM and WS-CM (combusted tobacco) significantly increased caspase-3 activity, while ST/CAS (non-combusted) showed no or only minimal activation. | [55] | |
Human gingival epithelial cells | E-cigarette vapor | A significant induction of caspase-3 proteins was observed in e-cigarette vapor-exposed cells. | [58] | |
Human gingival fibroblasts | E-cigarette vapors and cigarette smoke | No significant differences were recorded for the caspase 3/7 activity in the tested groups. | [81] | |
Odontoblast-like MDPC-23 cells | NaF | NaF exposure induced the activation of caspase-3. | [82] | |
Cementoblasts | Zinc-oxide eugenol-based root-canal sealer | The root-canal sealer stimulated caspase-3, -8, and -9 activity in a dose-dependent manner. | [76] | |
Human oral fibroblasts | Antioxidant mixtures of resveratrol (R), ferulic acid (F), phloretin (P), and tetrahydrocurcuminoids (T)—RFT, PFR, and PFT | The tested mixtures did not increase apoptosis. | [43] | |
Human gingival fibroblasts | Epicatechin gallate | Epicatechin gallate did not induce caspase-3 activity. | [52] | |
Image-iTTM LIVE green caspase detection | Human oral fibroblasts | Antioxidant mixtures of resveratrol (R), ferulic acid (F), phloretin (P), and tetrahydrocurcuminoids (T)—RFT, PFR, and PFT | The tested mixtures did not increase apoptosis. | [43] |
DAPI staining and microscopy analysis | Human gingival fibroblasts | Nicotine | Apoptotic cellular bodies, nuclear condensation, and DNA fragmentation were present. | [80] |
Human gingival epithelium keratinocytes | Traditional cigarette smoke and electronic cigarette aerosols | Apoptotic morphology was detected; there were no significant differences in cell morphology between t-cigs and e-cigs. | [39] | |
Annexin- V-FITC | Human gingival fibroblasts | Commercial topical fluoride varnishes | Some of the tested samples exhibited high cytotoxicity, associated with certain components present in the fluoride varnishes (cetylpyridinium chloride and ethyl acetate). | [83] |
Human normal oral epithelial cells | Cisplatin and thymoquinone (alone and in combination) | Thymoquinone (in combination with cisplatin) decreased cisplatin-induced apoptosis and necrosis in normal cells. | [49] | |
Human gingival fibroblasts | Vitamin C | Vitamin C reduced apoptosis rates in human gingival fibroblasts exposed to Porphyromonas gingivalis. | [84] | |
Human gingival fibroblasts | Curcumin | The proportion of apoptotic cells increased with increasing concentrations of curcumin. | [85] | |
Human oral fibroblast | Menthol and eucalyptol | The results indicated a concentration-dependent and exposure-time-dependent cytotoxicity for eucalyptol and menthol. | [86] | |
Human gingival fibroblasts | Methacrylate-based resins | Methacrylate-based resins produced cell death via necrosis or apoptosis, depending on the sample. | [87] | |
Human gingival fibroblasts | N-acetylcysteine incorporated in methacrylate-based resin cement | N-acetylcysteine diminished cytotoxicity, cell apoptosis, and necrosis in cells. | [88] | |
Human gingival fibroblast | Dental resin polymerization initiators | Dental resin polymerization initiators induced cell death via necrosis. | [89] | |
Western blot analysis of apoptotic proteins | Human gingival fibroblasts | Nicotine | Nicotine increased BAX expression (pro-apoptotic protein) and decreased BCL-2 expression (anti-apoptotic protein). | [80] |
Human normal oral epithelial cells | Cisplatin and thymoquinone (alone and in combination) | Cisplatin and thymoquinone (alone and in combination) showed a significant upregulation of p53 and caspase-9 (pro-apoptotic), and a decrease in the anti-apoptotic Bcl-2 protein expression. | [49] | |
Human gingival fibroblasts | Hydrogen peroxide | H2O2 increased the BAX expression and decreased the Bcl-2 expression. | [78] | |
Mitochondrial membrane potential assay | Human gingival fibroblasts | Curcumin | Curcumin induced early apoptotic events characterized by the loss of the mitochondrial membrane potential. | [85] |
Human gingival fibroblasts | TEGDMA released from resin composite | TEGDMA induced mitochondrial damage and mitochondrial membrane potential collapse. | [90] |
3.2. Morphological Changes Assessment
3.3. Genotoxicity Assessment
Method | Type of Cells | Tested Agent | Results | Reference |
---|---|---|---|---|
Micronucleus assay | Oral epithelial cells | Liquids for electronic cigarettes | Positivity for micronuclei was recorded for flavored and unflavored e-liquids. | [54] |
Human oral keratinocytes | Dental photoinitiators (BAPO and TPO) | No genotoxic effect was observed. | [30] | |
Human gingival fibroblasts | Single-wall carbon nanotubes | The frequency of micronuclei was higher after exposure to lower doses (50–100 μg/mL), and lower for high-dose treatments (125–150 μg/mL). | [41] | |
Human pulp fibroblasts | Aloe vera associated with endodontic medication (calcium hydroxide) and laser photo-biomodulation | The highest micronucleus rate was obtained for the association Aloe vera + laser therapy. | [126] | |
Human gingival fibroblasts | Toothpastes | The tested toothpastes did not exhibit genotoxic effects. | [127] | |
Human gingival fibroblasts | Static magnetic field produced by dental magnetic attachments | The micronuclei rates increased in the exposed groups, but no mitotic changes appeared. | [128] | |
Human gingival fibroblasts | Achyrocline satureioides extract | The micronucleus count increased in a concentration-dependent manner; no genotoxic effects were recorded for concentrations up to 6.25 mg/mL. | [129] | |
Human gingival fibroblasts | Methacrylic monomer BAPP | A dose-related increase in the number of micronuclei was recorded. | [130] | |
Comet assay | Human gingival fibroblasts | Single-wall carbon nanotubes | The nanotubes induced a significant increase in tail moments. | [41] |
Human gingival fibroblasts | Bracket alloys | All samples exhibited genotoxicity, but the Ni-Ti combination showed the highest DNA damage. | [131] | |
Human gingival fibroblasts and buccal epithelial cells | Corrosion eluates from stainless-steel brackets, nanoparticle-coated brackets, and polymeric-coated brackets | The stainless-steel bracket eluates caused increased nuclear damage in cells. | [132] | |
Human gingival fibroblasts | Metals released from three orthodontic alloys (stainless-steel, nickel-free, and titanium) | The greatest DNA damage was produced by the stainless-steel alloy. | [133] | |
Human gingival keratinocytes | Corrosion eluates produced from orthodontic materials | No apparent DNA damage was recorded. | [134] | |
Human oral keratinocytes | TEGDMA | The increase in tail moments after TEGDMA exposure was concentration-dependent. | [135] | |
Human gingival fibroblasts | BAPP | The incidence of DNA strand breaks increased in a concentration-dependent manner. | [130] | |
Human gingival fibroblasts | HEMA | HEMA induced significant fragmentation of the DNA. | [136] | |
Human gingival fibroblasts | Non-irradiated campherquinone | The exposure to campherquinone caused significant DNA damage. | [137] | |
Oral epithelial cells | Liquids for electronic cigarettes | No significant increase in DNA damage assessed by the comet assay was recorded. | [54] | |
Human periodontal ligament fibroblasts | E-cigarette vapors with flavoring | An increase in fluorescent tail length was observed, indicating DNA damage. | [138] | |
γH2AX assay | Human gingival fibroblasts | HEMA | A significant extent of DNA double-strand breaks was observed. | [136] |
Human gingival fibroblasts | Methacrylate-based adhesives | The highest level of DNA damage induction was exhibited by the adhesive with diphenyliodonium hexafluorophosphate as a co-initiator. | [46] | |
Human gingival fibroblasts | N-acetylcysteine incorporated in methacrylate-based resin cement | Incorporating N-acetylcysteine resulted in an anticytotoxic effect. | [88] | |
Human gingival fibroblasts | Methacrylate-based monomers in association with antioxidants (N-acetylcysteine and ascorbic acid) | The addition of antioxidants reduced the number of DNA double-strand breaks after exposure to methacrylate-based monomers. | [125] | |
Human gingival fibroblasts | Titanium dioxide-modified glass ionomer cements | No genotoxic effects were observed for the titanium dioxide-modified glass ionomer cements. | [139] | |
Human gingival fibroblasts | Dental resin restoration components (TEEGDMA, Neopen, DPIC, TPSB, TPP) | The induction of DNA double-strand breaks in exposed cells was observed, with the highest incidence for diphenyliodoniumchloride and neopentylglycol dimethacrylate. | [140] | |
DNA precipitation assay | Buccal fibroblasts | Arecoline | No DNA single-strand breaks appeared after exposure to arecoline. | [141] |
Integrity of genomic DNA (electrophoresis) | Human gingival fibroblasts and buccal epithelial cells | Stainless-steel brackets, nanoparticle-coated brackets, and polymeric-coated brackets exposed to corrosion eluates | The number of cellular alterations was insignificant. | [132] |
Human gingival fibroblasts | Silver tungstate microcrystals | DNA integrity was not affected. | [62] |
3.4. Oxidative Stress
3.5. Inflammatory Response
4. Three-Dimensional Cell Culture Models
Type of 3D Model | Investigated Parameter | Tested Agent | Results | Reference |
---|---|---|---|---|
Dental materials and oral healthcare formulations | ||||
Human gingival epithelial culture model EpiGingival™ | Cell viability and retention of o-cymen-5-ol and zinc | Toothpaste with o-cymen-5-ol and zinc | No cytotoxic effect appeared and the delivery of o-cymen-5-ol and zinc was successful. | [173] |
Human oral epithelial culture model EpiOral™ | Tissue viability and cytokine release | Prototype oral care formulations | The models were useful to screen the irritation effect of oral products. | [174] |
Three-dimensional human oral mucosal model | Histology, tissue viability, and IL-1β release | Dental composite resins | TEGDMA-based composite resin increased IL-1β release and caused mucotoxicity. | [175] |
Human oral epithelial culture model EpiOral™ | Tissue viability, histology, and caffeine permeability | Caffeine, ethanol, and mouthrinses | Ethanol-containing mouthwashes did not exhibit a cytotoxic effect on the mucosal tissue; the permeability of caffeine was not affected. | [176] |
Human oral epithelial culture model EpiOral™ | Viability, cytokine release, levels of protein markers for apoptosis, DNA damage, cell proliferation, cell–cell adhesion | Mucoadhesive polymer blend (pullulan, tamarindus indica polysaccharide, and sodium hyaluronate) | No cytotoxic effect was observed and the cytokine levels were not modified; there was no indication of DNA damage. | [177] |
Reconstituted human oral epithelial tissue models (3D) (SkinEthic Laboratories) | Cell viability and morphology | Point-welded, laser-welded, and silver-soldered orthodontic wires | Viability and histological evaluation did not reveal signs of severe toxicity. | [178] |
Oral mucosa tissue model (3D) | Transepithelial electrical resistance, cell viability, and cytokine secretion | Mouth rinsing solutions | Cell viability was not affected, but only one solution had no impact on TEER. One product increased IL 8 secretion. | [179] |
DentCytoTool | Viability, cytotoxicity, and odontogenesis- and angiogenesis-related markers | HEMA; TEGDMA | The cytotoxic potential increased in a time- and concentration-dependent manner, and the expression of angiogenesis-related markers was reduced; the LPS/TEGDMA co-treatment aggravated the cytotoxic effects. | [172] |
Pulp analogue (3D) | Cell viability/proliferation, morphology, and expression of angiogenic and odontogenic markers | Calcium-silicate-based cements | The materials were biocompatible, with positive angiogenic and odontogenic effects. | [180] |
Oral mucosal models | Cytotoxicity, histological, and permeation analyses | Mucoadhesive patches with clobetasol-17-propionate | No tissue damage was recorded. | [181] |
Nicotine, ENDS, and tobacco products | ||||
Human oral epithelial culture model EpiOral™ | Cytotoxicity, histological analysis, cytochrome P450 activity, production of pro-inflammatory mediators, and transcriptomics | Cigarette smoke and aerosols obtained from a modified-risk tobacco product | Cytotoxicity, morphological alterations, production of inflammatory mediators, and transcriptomic changes were significantly more pronounced for cigarette smoke. | [182] |
Human gingival epithelial culture model EpiGingival™ | Cytotoxicity, histological analysis, cytochrome P450 activity, production of pro-inflammatory mediators, transcriptomics, and metabolomics | Cigarette smoke and aerosols obtained from a modified-risk tobacco product | The aerosol from the modified-risk tobacco product caused minimal cytotoxicity and minor histopathological alterations, and had a low impact on transcriptomic and metabolomic data. | [183] |
Human gingival epithelial culture model EpiGingival™ | Pro-inflammatory cytokine levels, inflammation and DNA damage markers (RAGE, COX-2, and γH2A.X) | E-cigarette aerosols | E-cigarette exposure augmented inflammation and DNA damage markers. | [138] |
Human oral epithelial culture model EpiOral™ | Viability, morphology, and tissue absorption | E-cigarette aerosol | No decrease in viability and no change in morphology and tissue structure were observed. | [184] |
Human oral epithelial culture model EpiOral™ Human gingival epithelial culture model EpiGingival™ | Cytotoxicity (adenylate kinase assay), histology, inflammatory mediator secretion, transcriptomics, and targeted proteomics | Cigarette smoke and a carbon-heated tobacco-product aerosol | The carbon-heated tobacco-product aerosol exhibited a significantly lower biological impact in comparison to cigarette smoke. | [185] |
Human gingival epithelial culture model EpiGingival™ | Irritant potential | Extracts from nicotine pouch products | The tested products were not irritants. | [186] |
Oral mucosa tissue model (3D) with normal fibroblasts and cancerous TR146 keratinocytes | Histological examination and viability | E-cigarette liquids | Prolonged exposure and high concentrations of e-liquids caused cytotoxic effects for normal cells but stimulated the growth of cancerous cells. | [187] |
Other agents | ||||
Gingival culture system (3D) | DNA analysis | Sodium butyrate | Sodium butyrate induced DNA release in a time- and dose-dependent manner, and stimulated the release of SAP130. | [188] |
Human oral epithelial culture model EpiOral™ | Cell viability and histological analysis | Sodium lauryl sulfate | EpiOral is a suitable system for oral mucosal irritation test. | [170] |
Human gingival epithelial culture model EpiGingival™ Human oral epithelial culture model EpiOral™ | DNA damage, CPD repair rate, and apoptotic cell numbers | UV radiation | UVB radiation presented a higher carcinogenic risk for oral tissues. | [189] |
Human gingival epithelial culture model EpiGingival™ Human oral epithelial culture model EpiOral™ | Cytokine release, DNA damage, CPD repair rate, and apoptotic cell numbers | UVB radiation | Increased interleukin-8 (IL-8) release, low rates of CPD repair, and decreased apoptotic cell numbers. | [190] |
Cell culture model (3D) (gingival fibroblasts in collagen matrix) | Cell viability and morphology, and gene expression of growth factors | Low-level laser therapy | Low-level laser therapy increased viability and promoted biostimulation of gingival fibroblasts. | [191] |
Reconstituted human oral epithelial tissue models (3D) (EpiSkin Laboratories) | Cell viability and morphology | Carrageenan | No loss of viability was recorded for the carrageenan-treated sample. | [31] |
Human gingival epithelial culture model EpiGingival™ | Cytotoxicity, viability, histology, immunohistochemistry (Ki67, VEGF-A), and TUNEL assay | Low-temperature plasma | Low-temperature plasma had low cytotoxicity and high cellular viability. | [192] |
Human gingival epithelial culture model EpiGingival™ | Viability | Lipophilic vehicles | No signs of cytotoxicity were recorded. | [193] |
4.1. Cell Viability
4.2. Morphological Changes
4.3. Genotoxicity Assessment
4.4. Oxidative Stress and Inflammatory Response
5. Organ-on-a-Chip Systems
6. In Vitro Models for Toxicity Assessment: Advantages, Challenges, and Future Perspectives
7. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Conflicts of Interest
Abbreviations
5-CFDA-AM | 5-carboxyfluorescein diacetate acetoxymethyl ester |
3H-TdR | tritiated thymidine |
8OHdG | 8-hydroxy-2′-deoxyguanosine |
8-oxo-Gua | 8-oxoguanine |
ATP | adenosine triphosphate |
BAPO | phenyl-bis(acyl) phosphine oxide |
BAPP | 2,2-bis[4-(acryloxypropoxy) phenyl] propane |
BAX | Bcl-2-associated X protein |
BisGMA | bisphenol A-glycidylmethacrylate |
BrdU | 5-bromo-2′-deoxyuridine |
C | catechin |
CAPE | caffeic acid phenethyl ester |
CAT | catalase |
CG | catechin gallate |
CLFPs | cytoplasm-localized fluorescent probes |
CPD | cyclobutane pyrimidine dimer |
CQ | camphorquinone |
CRP1.1 | a smokeless tobacco reference product—snus |
CSFs | colony-stimulating factors |
DCF | 2′,7′-dichlorofluorescein |
DPIC | diphenyliodonium chloride |
DNPH | 2,4-dinitrophenylhydrazine |
EC | epicatechin |
ECG | epicatechin gallate |
EGC | epigallocatechin |
EGCG | epigallocatechin gallate |
ENDS | electronic nicotine delivery systems |
EdU | 5-ethynyl-2-deoxyuridine |
Er:YAG | erbium-doped yttrium aluminum garnet |
FACS | fluorescence-activated cell sorting |
G6PD | glucose-6-phosphate dehydrogenase |
GPx | glutathione peroxidase family |
GR | glutathione reductase |
GSH | glutathione |
H2DCF | 2′,7′-dichlorodihydrofluorescein diacetate |
HEMA | 2-hydroxy-ethyl-methacrylate |
IWGT WG | the International Workshop on Genotoxicity Testing Working Group |
LDH | lactate dehydrogenase |
LPS | lipopolysaccharides |
MDA | malondialdehyde |
MIP | macrophage inflammatory proteins |
MMPs | matrix metalloproteinases |
MTS | dimethylthiazolcarboxymethoxyphenylsulfophenyl-tetrazolium |
MTT | methylthiazolyldiphenyltetrazolium bromide |
NADPH | nicotinamide adenine dinucleotide phosphate |
NRU | neutral red uptake |
PARP1 | poly-(ADP-ribose) polymerase-1 |
PCNA | proliferating cell nuclear antigen |
PHH3 | phosphohistone H3 |
RAGE | receptor for advanced glycation end |
RIPK1 | receptor interacting serine/threonine kinase 1 |
SAP130 | Sin3A associated protein 130 kDa |
SDF-1 | stromal cell-derived factor 1 |
SOD | superoxide dismutase |
SRB | sulforhodamine B |
TBA | thiobarbituric acid |
TEGD-MA | triethyleneglycol-dimethacrylate |
TEEGDMA | tetraethyleneglycol dimethacrylate |
TEER | transepithelial electrical resistance |
TGFs | transforming growth factors |
TNFα | tumor necrosis factor α |
TPO | diphenyl(acyl) phosphine oxide |
TPSB | triphenyl-stibane |
TPP | triphenylphosphane |
UDMA | urethane dimethacrylate |
VEGF | vascular endothelial growth factor |
WST | water-soluble tetrazolium salt |
XTT | methoxynitrosulfophenyl-tetrazolium carboxanilide |
References
- Poh, W.T.; Stanslas, J. The new paradigm in animal testing—“3Rs alternatives”. Regul. Toxicol. Pharmacol. 2024, 153, 105705. [Google Scholar] [CrossRef] [PubMed]
- ISO 10993-5:2009; Biological Evaluation of Medical Devices Part 5: Tests for in Vitro Cytotoxicity. International Organization for Standardization: Geneva, Switzerland, 2009.
- Srivastava, G.K.; Alonso-Alonso, M.L.; Fernandez-Bueno, I.; Garcia-Gutierrez, M.T.; Rull, F.; Medina, J.; Coco, R.M.; Pastor, J.C. Comparison between direct contact and extract exposure methods for PFO cytotoxicity evaluation. Sci. Rep. 2018, 8, 1425. [Google Scholar] [CrossRef]
- Liu, X.; Rodeheaver, D.P.; White, J.C.; Wright, A.M.; Walker, L.M.; Zhang, F.; Shannon, S. A comparison of in vitro cytotoxicity assays in medical device regulatory studies. Regul. Toxicol. Pharmacol. 2018, 97, 24–32. [Google Scholar] [CrossRef]
- Gruber, S.; Nickel, A. Toxic or not toxic? The specifications of the standard ISO 10993-5 are not explicit enough to yield comparable results in the cytotoxicity assessment of an identical medical device. Front. Med. Technol. 2023, 5, 1195529. [Google Scholar] [CrossRef]
- Schwarze, T.; Fiedler, I.; Leyhausen, G.; Geurtsen, W. The cellular compatibility of five endodontic sealers during the setting period. J. Endod. 2002, 28, 784–786. [Google Scholar] [CrossRef] [PubMed]
- Badole, G.P.; Warhadpande, M.M.; Meshram, G.K.; Bahadure, R.N.; Tawani, S.G.; Tawani, G.; Badole, S.G. A comparative evaluation of cytotoxicity of root canal sealers: An in vitro study. Restor. Dent. Endod. 2013, 38, 204–209. [Google Scholar] [CrossRef]
- Kumar, A.; Kour, S.; Kaul, S.; Malik, A.; Dhani, R.; Kaul, R. Cytotoxicity evaluation of Bio-C, CeraSeal, MTA—Fillapex, and AH Plus root canal sealers by microscopic and 3-(4, 5 dimethythiazol-2yl)-2, 5-diphenyltetrazolium bromide (MTT) assay. J. Conserv. Dent. 2023, 26, 73–78. [Google Scholar] [CrossRef] [PubMed]
- Sazonova, E.V.; Chesnokov, M.S.; Zhivotovsky, B.; Kopeina, G.S. Drug toxicity assessment: Cell proliferation versus cell death. Cell Death Discov. 2022, 8, 417. [Google Scholar] [CrossRef]
- Riss, T.L.; Moravec, R.A.; Niles, A.L.; Duellman, S.; Benink, H.A.; Worzella, T.J.; Minor, L. Cell Viability Assays. In Assay Guidance Manual [Internet]; Markossian, S., Grossman, A., Arkin, M., Auld, D., Austin, C., Baell, J., Brimacombe, K., Chung, T.D.Y., Coussens, N.P., Dahlin, J.L., et al., Eds.; Eli Lilly & Company and the National Center for Advancing Translational Sciences: Bethesda, MD, USA, 2004. Available online: https://www.ncbi.nlm.nih.gov/sites/books/NBK53196/ (accessed on 20 November 2024).
- Kamiloglu, S.; Sari, G.; Ozdal, T.; Capanoglu, E. Guidelines for cell viability assays. Food Front. 2020, 1, 332–349. [Google Scholar] [CrossRef]
- Aslantürk, Ö.S. In Vitro Cytotoxicity and Cell Viability Assays: Principles, Advantages, and Disadvantages [Internet]. In Genotoxicity—A Predictable Risk to Our Actual World; InTech: London, UK, 2018. [Google Scholar] [CrossRef]
- Adan, A.; Kiraz, Y.; Baran, Y. Cell Proliferation and Cytotoxicity Assays. Curr. Pharm. Biotechnol. 2016, 17, 1213–1221. [Google Scholar] [CrossRef]
- Präbst, K.; Engelhardt, H.; Ringgeler, S.; Hübner, H. Basic Colorimetric Proliferation Assays: MTT, WST, and Resazurin. In Cell Viability Assays. Methods in Molecular Biology; Gilbert, D., Friedrich, O., Eds.; Humana Press: New York, NY, USA, 2017; Volume 1601. [Google Scholar]
- Ates, G.; Vanhaecke, T.; Rogiers, V.; Rodrigues, R.M. Assaying Cellular Viability Using the Neutral Red Uptake Assay. In Cell Viability Assays. Methods in Molecular Biology; Gilbert, D., Friedrich, O., Eds.; Humana Press: New York, NY, USA, 2017; Volume 1601. [Google Scholar]
- Vichai, V.; Kirtikara, K. Sulforhodamine B colorimetric assay for cytotoxicity screening. Nat. Protoc. 2006, 1, 1112–1116. [Google Scholar] [CrossRef] [PubMed]
- Duellman, S.J.; Zhou, W.; Meisenheimer, P.; Vidugiris, G.; Cali, J.J.; Gautam, P.; Wennerberg, K.; Vidugiriene, J. Bioluminescent, Nonlytic, Real-Time Cell Viability Assay and Use in Inhibitor Screening. Assay. Drug Dev. Technol. 2015, 13, 456–465. [Google Scholar] [CrossRef]
- Drescher, H.; Weiskirchen, S.; Weiskirchen, R. Flow Cytometry: A Blessing and a Curse. Biomedicines 2021, 9, 1613. [Google Scholar] [CrossRef]
- Cummings, B.S.; Schnellmann, R.G. Measurement of Cell Death in Mammalian Cells. Curr. Protoc. 2021, 1, e210. [Google Scholar] [CrossRef]
- Franken, N.A.; Rodermond, H.M.; Stap, J.; Haveman, J.; van Bree, C. Clonogenic assay of cells in vitro. Nat. Protoc. 2006, 1, 2315–2319. [Google Scholar] [CrossRef] [PubMed]
- Flomerfelt, F.A.; Gress, R.E. Analysis of Cell Proliferation and Homeostasis Using EdU Labeling. Methods Mol. Biol. 2016, 1323, 211–220. [Google Scholar] [CrossRef] [PubMed]
- Salic, A.; Mitchison, T.J. A chemical method for fast and sensitive detection of DNA synthesis in vivo. Proc. Natl. Acad. Sci. USA 2008, 105, 2415–2420. [Google Scholar] [CrossRef]
- Boehm, E.M.; Gildenberg, M.S.; Washington, M.T. The Many Roles of PCNA in Eukaryotic DNA Replication. Enzymes 2016, 39, 231–254. [Google Scholar] [CrossRef]
- Stefanowicz-Hajduk, J.; Ochocka, J.R. Real-time cell analysis system in cytotoxicity applications: Usefulness and comparison with tetrazolium salt assays. Toxicol. Rep. 2020, 7, 335–344. [Google Scholar] [CrossRef]
- Urcan, E.; Haertel, U.; Styllou, M.; Hickel, R.; Scherthan, H.; Reichl, F.X. Real-time xCELLigence impedance analysis of the cytotoxicity of dental composite components on human gingival fibroblasts. Dent. Mater. 2010, 26, 51–58. [Google Scholar] [CrossRef]
- Larsson, P.; Engqvist, H.; Biermann, J.; Werner Rönnerman, E.; Forssell-Aronsson, E.; Kovács, A.; Karlsson, P.; Helou, K.; Parris, T.Z. Optimization of cell viability assays to improve replicability and reproducibility of cancer drug sensitivity screens. Sci. Rep. 2020, 10, 5798. [Google Scholar] [CrossRef] [PubMed]
- Frigaard, J.; Liaaen Jensen, J.; Kanli Galtung, H.; Hiorth, M. Stability and cytotoxicity of biopolymer-coated liposomes for use in the oral cavity. Int. J. Pharm. 2023, 645, 123407. [Google Scholar] [CrossRef] [PubMed]
- Basso, F.G.; Pansani, T.N.; de Oliveira, C.F.; Turrioni, A.P.; Soares, D.G.; Hebling, J.; Costa, C.A. Cytotoxic effects of zoledronic acid on human epithelial cells and gingival fibroblasts. Braz. Dent. J. 2013, 24, 551–558. [Google Scholar] [CrossRef]
- Modena, K.C.D.S.; Calvo, A.M.; Sipert, C.R.; Dionísio, T.J.; Navarro, M.F.L.; Atta, M.T.; Santos, C.F. Dental Pulp Fibroblasts Response after Stimulation with HEMA and Adhesive System. Braz. Dent. J. 2018, 29, 419–426. [Google Scholar] [CrossRef]
- Popal, M.; Volk, J.; Leyhausen, G.; Geurtsen, W. Cytotoxic and genotoxic potential of the type I photoinitiators BAPO and TPO on human oral keratinocytes and V79 fibroblasts. Dent. Mater. 2018, 34, 1783–1796. [Google Scholar] [CrossRef]
- Alli, B.Y.; Upadhyay, A.; Zhang, Y.; Nicolau, B.; Tran, S.D. In Vitro Toxicity Evaluation of Carrageenan on Cells and Tissues of the Oral Cavity. Mar. Drugs 2022, 20, 502. [Google Scholar] [CrossRef] [PubMed]
- Best, M.; Phillips, G.; Fowler, C.; Rowland, J.; Elsom, J. Characterisation and cytotoxic screening of metal oxide nanoparticles putative of interest to oral healthcare formulations in non-keratinised human oral mucosa cells in vitro. Toxicol. In Vitro 2015, 30 Pt B, 402–411. [Google Scholar] [CrossRef]
- Pagano, S.; Lombardo, G.; Costanzi, E.; Balloni, S.; Bruscoli, S.; Flamini, S.; Coniglio, M.; Valenti, C.; Cianetti, S.; Marinucci, L. Morpho-functional effects of different universal dental adhesives on human gingival fibroblasts: An in vitro study. Odontology 2021, 109, 524–539. [Google Scholar] [CrossRef]
- Sampath Kumar, L.; Shantha, V.; Janardhana, K. Cell viability studies using human gingival fibroblast, human oral fibroblast cells and mechanical characterization of Calcite/Zincite nanoparticles. World J. Adv. Res. Rev. 2022, 16, 812–824. [Google Scholar] [CrossRef]
- Nirwana, I.; Munadziroh, E.; Yogiartono, R.M.; Thiyagu, C.; Ying, C.S.; Dinaryanti, A. Cytotoxicity and proliferation evaluation on fibroblast after combining calcium hydroxide and ellagic acid. J. Adv. Pharm. Technol. Res. 2021, 12, 27–31. [Google Scholar] [CrossRef]
- Strauss, F.J.; Stähli, A.; Beer, L.; Mitulović, G.; Gilmozzi, V.; Haspel, N.; Schwab, G.; Gruber, R. Acid bone lysate activates TGFβ signalling in human oral fibroblasts. Sci. Rep. 2018, 8, 16065. [Google Scholar] [CrossRef] [PubMed]
- Soares, D.G.; Basso, F.G.; Hebling, J.; de Souza Costa, C.A. Concentrations of and application protocols for hydrogen peroxide bleaching gels: Effects on pulp cell viability and whitening efficacy. J. Dent. 2014, 42, 185–198. [Google Scholar] [CrossRef] [PubMed]
- Boza, Y.; Yefi, R.; Rudolph, M.I.; Smith, P.C.; Oberyszyn, T.M.; Tober, K.L.; Rojas, I.G. Single Exposure of Human Oral Mucosa Fibroblasts to Ultraviolet B Radiation Reduces Proliferation and Induces COX-2 Expression and Activation. Rev. Clín. Periodoncia Implant. Rehabil. Oral 2010, 3, 123–127. [Google Scholar] [CrossRef]
- Ramenzoni, L.L.; Schneider, A.; Fox, S.C.; Meyer, M.; Meboldt, M.; Attin, T.; Schmidlin, P.R. Cytotoxic and Inflammatory Effects of Electronic and Traditional Cigarettes on Oral Gingival Cells Using a Novel Automated Smoking Instrument: An In Vitro Study. Toxics 2022, 10, 179. [Google Scholar] [CrossRef]
- Alanazi, H.; Park, H.J.; Chakir, J.; Semlali, A.; Rouabhia, M. Comparative study of the effects of cigarette smoke and electronic cigarettes on human gingival fibroblast proliferation, migration and apoptosis. Food Chem. Toxicol. 2018, 118, 390–398. [Google Scholar] [CrossRef]
- Cicchetti, R.; Divizia, M.; Valentini, F.; Argentin, G. Effects of single-wall carbon nanotubes in human cells of the oral cavity: Geno-cytotoxic risk. Toxicol. In Vitro 2011, 25, 1811–1819. [Google Scholar] [CrossRef] [PubMed]
- Tu, M.-G.; Liang, W.-M.; Wu, T.-C.; Chen, S.-Y. Evaluation of cytotoxicity of resin bonding materials toward human oral epithelial cells using three assay systems. J. Dent. Sci. 2009, 4, 178–186. [Google Scholar] [CrossRef]
- San Miguel, S.M.; Opperman, L.A.; Allen, E.P.; Zielinski, J.; Svoboda, K.K. Bioactive antioxidant mixtures promote proliferation and migration on human oral fibroblasts. Arch. Oral Biol. 2011, 56, 812–822. [Google Scholar] [CrossRef]
- Silva, D.; Arancibia, R.; Tapia, C.; Acuña-Rougier, C.; Diaz-Dosque, M.; Cáceres, M.; Martínez, J.; Smith, P.C. Chitosan and platelet-derived growth factor synergistically stimulate cell proliferation in gingival fibroblasts. J. Periodontal Res. 2013, 48, 677–686. [Google Scholar] [CrossRef]
- Soares, A.S.L.S.; Scelza, M.Z.; Spoladore, J.; Gallito, M.A.; Oliveira, F.; Moraes, R.C.M.; Alves, G.G. Comparison of primary human gingival fibroblasts from an older and a young donor on the evaluation of cytotoxicity of denture adhesives. J. Appl. Oral Sci. 2018, 26, e20160594. [Google Scholar] [CrossRef]
- Taubmann, A.; Willershausen, I.; Walter, C.; Al-Maawi, S.; Kaina, B.; Gölz, L. Genotoxic and cytotoxic potential of methacrylate-based orthodontic adhesives. Clin. Oral Investig. 2021, 25, 2569–2581. [Google Scholar] [CrossRef]
- Lee, J.H.; Seo, S.H.; Lee, S.B.; Om, J.Y.; Kim, K.M.; Kim, K.N. Cytotoxicity and terminal differentiation of human oral keratinocyte by indium ions from a silver-palladium-gold-indium dental alloy. Dent. Mater. 2015, 31, 123–133. [Google Scholar] [CrossRef] [PubMed]
- Kong, S.; Aoki, A.; Iwasaki, K.; Mizutani, K.; Katagiri, S.; Suda, T.; Ichinose, S.; Ogita, M.; Pavlic, V.; Izumi, Y. Biological effects of Er:YAG laser irradiation on the proliferation of primary human gingival fibroblasts. J. Biophotonics 2018, 11, e201700157. [Google Scholar] [CrossRef]
- Alaufi, O.M.; Noorwali, A.; Zahran, F.; Al-Abd, A.M.; Al-Attas, S. Cytotoxicity of thymoquinone alone or in combination with cisplatin (CDDP) against oral squamous cell carcinoma in vitro. Sci. Rep. 2017, 7, 13131. [Google Scholar] [CrossRef] [PubMed]
- Babich, H.; Sinensky, M.C. Indirect cytotoxicity of dental materials: A study with Transwell inserts and the neutral red uptake assay. Altern. Lab. Anim. 2001, 29, 9–13. [Google Scholar] [CrossRef]
- Tavares, S.J.d.O.; Caldas, I.P.; Gonçalves, F.P.; Scelza, P.; Oliveira, F.; Alves, G.; Scelza, M.F.Z. Cytotoxicity in fibroblasts from young and elderly donors from two mouthwashes used to prevent the spread of SARS-CoV-2. Res. Soc. Dev. 2021, 10, e56810414587. [Google Scholar] [CrossRef]
- Babich, H.; Krupka, M.E.; Nissim, H.A.; Zuckerbraun, H.L. Differential in vitro cytotoxicity of (-)-epicatechin gallate (ECG) to cancer and normal cells from the human oral cavity. Toxicol. In Vitro 2005, 19, 231–242. [Google Scholar] [CrossRef]
- Babich, H.; Pinsky, S.M.; Muskin, E.T.; Zuckerbraun, H.L. In vitro cytotoxicity of a theaflavin mixture from black tea to malignant, immortalized, and normal cells from the human oral cavity. Toxicol. In Vitro 2006, 20, 677–688. [Google Scholar] [CrossRef]
- Tellez, C.S.; Juri, D.E.; Phillips, L.M.; Do, K.; Yingling, C.M.; Thomas, C.L.; Dye, W.W.; Wu, G.; Kishida, S.; Kiyono, T.; et al. Cytotoxicity and Genotoxicity of E-Cigarette Generated Aerosols Containing Diverse Flavoring Products and Nicotine in Oral Epithelial Cell Lines. Toxicol. Sci. 2021, 179, 220–228. [Google Scholar] [CrossRef]
- Gao, H.; Prasad, G.L.; Zacharias, W. Differential cell-specific cytotoxic responses of oral cavity cells to tobacco preparations. Toxicol. In Vitro 2013, 27, 282–291. [Google Scholar] [CrossRef]
- Szkaradkiewicz, A.K.; Karpiński, T.M.; Szkaradkiewicz, A. Effect of novobiocin on the viability of human gingival fibroblasts (HGF-1). BMC Pharmacol. Toxicol. 2014, 15, 25. [Google Scholar] [CrossRef] [PubMed]
- Chen, F.C.; Huang, C.M.; Yu, X.W.; Chen, Y.Y. Effect of nano zinc oxide on proliferation and toxicity of human gingival cells. Hum. Exp. Toxicol. 2022, 41, 9603271221080236. [Google Scholar] [CrossRef]
- Rouabhia, M.; Park, H.J.; Semlali, A.; Zakrzewski, A.; Chmielewski, W.; Chakir, J. E-Cigarette Vapor Induces an Apoptotic Response in Human Gingival Epithelial Cells Through the Caspase-3 Pathway. J. Cell Physiol. 2017, 232, 1539–1547. [Google Scholar] [CrossRef] [PubMed]
- Aati, S.; Shrestha, B.; Fawzy, A. Cytotoxicity and antimicrobial efficiency of ZrO2 nanoparticles reinforced 3D printed resins. Dent. Mater. 2022, 38, 1432–1442. [Google Scholar] [CrossRef]
- Poulsen, C.; Mehalick, L.A.; Fischer, C.L.; Lanzel, E.A.; Bates, A.M.; Walters, K.S.; Cavanaugh, J.E.; Guthmiller, J.M.; Johnson, G.K.; Wertz, P.W.; et al. Differential cytotoxicity of long-chain bases for human oral gingival epithelial keratinocytes, oral fibroblasts, and dendritic cells. Toxicol. Lett. 2015, 237, 21–29. [Google Scholar] [CrossRef]
- Adil, N.; Ali, H.; Siddiqui, A.J.; Ali, A.; Ahmed, A.; El-Seedi, H.R.; Musharraf, S.G. Evaluation of cytotoxicity of areca nut and its commercial products on normal human gingival fibroblast and oral squamous cell carcinoma cell lines. J. Hazard. Mater. 2021, 403, 123872. [Google Scholar] [CrossRef] [PubMed]
- Haro Chávez, N.L.; de Avila, E.D.; Barbugli, P.A.; de Oliveira, R.C.; de Foggi, C.C.; Longo, E.; Vergani, C.E. Promising effects of silver tungstate microcrystals on fibroblast human cells and three dimensional collagen matrix models: A novel non-cytotoxic material to fight oral disease. Colloids Surf. B Biointerfaces 2018, 170, 505–513. [Google Scholar] [CrossRef] [PubMed]
- Willershausen, I.; Wolf, T.; Weyer, V.; Sader, R.; Ghanaati, S.; Willershausen, B. Influence of E-smoking liquids on human periodontal ligament fibroblasts. Head. Face Med. 2014, 10, 39. [Google Scholar] [CrossRef]
- Ji, E.H.; Sun, B.; Zhao, T.; Shu, S.; Chang, C.H.; Messadi, D.; Xia, T.; Zhu, Y.; Hu, S. Characterization of Electronic Cigarette Aerosol and Its Induction of Oxidative Stress Response in Oral Keratinocytes. PLoS ONE 2016, 11, e0154447, Erratum in PLoS ONE 2016, 11, e0169380. https://doi.org/10.1371/journal.pone.0169380. [Google Scholar] [CrossRef]
- Bagchi, M.; Balmoori, J.; Bagchi, D.; Ray, S.D.; Kuszynski, C.; Stohs, S.J. Smokeless tobacco, oxidative stress, apoptosis, and antioxidants in human oral keratinocytes. Free Radic. Biol. Med. 1999, 26, 992–1000, Erratum in Free Radic. Biol. Med. 1999, 26, 1599. [Google Scholar] [CrossRef]
- Cotrim, P.; Martelli-Junior, H.; Graner, E.; Sauk, J.J.; Coletta, R.D. Cyclosporin A induces proliferation in human gingival fibroblasts via induction of transforming growth factor-beta1. J. Periodontol. 2003, 74, 1625–1633. [Google Scholar] [CrossRef]
- Thomsen, A.R.; Aldrian, C.; Luka, B.; Hornhardt, S.; Gomolka, M.; Moertl, S.; Hess, J.; Zitzelsberger, H.; Heider, T.; Schlueter, N.; et al. Biopsy-derived oral keratinocytes—A model to potentially test for oral mucosa radiation sensitivity. Clin. Transl. Radiat. Oncol. 2022, 34, 51–56. [Google Scholar] [CrossRef] [PubMed]
- Nguyen, P.A.; Pham, T.A.V. Effects of platelet-rich plasma on human gingival fibroblast proliferation and migration in vitro. J. Appl. Oral Sci. 2018, 26, e20180077. [Google Scholar] [CrossRef] [PubMed]
- Trivedy, C.; Meghji, S.; Warnakulasuriya, K.A.; Johnson, N.W.; Harris, M. Copper stimulates human oral fibroblasts in vitro: A role in the pathogenesis of oral submucous fibrosis. J. Oral Pathol. Med. 2001, 30, 465–470. [Google Scholar] [CrossRef]
- Tipton, D.A.; Dabbous, M.K. Effects of nicotine on proliferation and extracellular matrix production of human gingival fibroblasts in vitro. J. Periodontol. 1995, 66, 1056–1064. [Google Scholar] [CrossRef] [PubMed]
- Wyganowska-Swiatkowska, M.; Kotwicka, M.; Urbaniak, P.; Nowak, A.; Skrzypczak-Jankun, E.; Jankun, J. Clinical implications of the growth-suppressive effects of chlorhexidine at low and high concentrations on human gingival fibroblasts and changes in morphology. Int. J. Mol. Med. 2016, 37, 1594–1600. [Google Scholar] [CrossRef]
- Robertson, J.D.; Orrenius, S. Molecular mechanisms of apoptosis induced by cytotoxic chemicals. Crit. Rev. Toxicol. 2000, 30, 609–627. [Google Scholar] [CrossRef]
- Elmore, S. Apoptosis: A review of programmed cell death. Toxicol. Pathol. 2007, 35, 495–516. [Google Scholar] [CrossRef]
- Darzynkiewicz, Z.; Galkowski, D.; Zhao, H. Analysis of apoptosis by cytometry using TUNEL assay. Methods 2008, 44, 250–254. [Google Scholar] [CrossRef]
- Payne, A.M.; Zorman, J.; Horton, M.; Dubey, S.; ter Meulen, J.; Vora, K.A. Caspase activation as a versatile assay platform for detection of cytotoxic bacterial toxins. J. Clin. Microbiol. 2013, 51, 2970–2976. [Google Scholar] [CrossRef]
- Huang, F.-M.; Kuan, Y.-H.; Lee, S.-S.; Chang, Y.-C. Caspase activation by a zinc-oxide eugenol-based root-canal sealer in cementoblasts. J. Dent. Sci. 2015, 10, 338–340. [Google Scholar] [CrossRef]
- Kaushal, V.; Herzog, C.; Haun, R.S.; Kaushal, G.P. Caspase protocols in mice. Methods Mol. Biol. 2014, 1133, 141–154. [Google Scholar] [CrossRef] [PubMed]
- Gutiérrez-Venegas, G.; Guadarrama-Solís, A.; Muñoz-Seca, C.; Arreguín-Cano, J.A. Hydrogen peroxide-induced apoptosis in human gingival fibroblasts. Int. J. Clin. Exp. Pathol. 2015, 8, 15563–15572. [Google Scholar] [PubMed]
- Khalil, A.M.; Alemam, I.F.; Al-Qaoud, K.M. Association between Mobile Phone Using and DNA Damage of Epithelial Cells of the Oral Mucosa. J. Biotechnol. Biomed. 2020, 3, 50–66. [Google Scholar] [CrossRef]
- Kang, S.W.; Park, H.J.; Ban, J.Y.; Chung, J.H.; Chun, G.S.; Cho, J.O. Effects of nicotine on apoptosis in human gingival fibroblasts. Arch. Oral Biol. 2011, 56, 1091–1097. [Google Scholar] [CrossRef]
- Vermehren, M.F.; Wiesmann, N.; Deschner, J.; Brieger, J.; Al-Nawas, B.; Kämmerer, P.W. Comparative analysis of the impact of e-cigarette vapor and cigarette smoke on human gingival fibroblasts. Toxicol. In Vitro 2020, 69, 105005. [Google Scholar] [CrossRef]
- Karube, H.; Nishitai, G.; Inageda, K.; Kurosu, H.; Matsuoka, M. NaF activates MAPKs and induces apoptosis in odontoblast-like cells. J. Dent. Res. 2009, 88, 461–465. [Google Scholar] [CrossRef]
- López-García, S.; Pecci-Lloret, M.P.; Pecci-Lloret, M.R.; Guerrero-Gironés, J.; Rodríguez-Lozano, F.J.; García-Bernal, D. Topical fluoride varnishes promote several biological responses on human gingival cells. Ann. Anat. 2021, 237, 151723. [Google Scholar] [CrossRef]
- Staudte, H.; Güntsch, A.; Völpel, A.; Sigusch, B.W. Vitamin C attenuates the cytotoxic effects of Porphyromonas gingivalis on human gingival fibroblasts. Arch. Oral Biol. 2010, 55, 40–45. [Google Scholar] [CrossRef]
- Atsumi, T.; Tonosaki, K.; Fujisawa, S. Induction of early apoptosis and ROS-generation activity in human gingival fibroblasts (HGF) and human submandibular gland carcinoma (HSG) cells treated with curcumin. Arch. Oral Biol. 2006, 51, 913–921. [Google Scholar] [CrossRef]
- Puig-Herreros, C.; Sanz, J.L.; García-Bernal, D.; Rodríguez-Lozano, F.J.; Murcia, L.; Forner, L.; Ghilotti, J.; Oñate-Sánchez, R.E.; López-García, S. Comparative Cytotoxicity of Menthol and Eucalyptol: An In Vitro Study on Human Gingival Fibroblasts. Pharmaceutics 2024, 16, 521. [Google Scholar] [CrossRef] [PubMed]
- Sulek, J.; Luczaj-Cepowicz, E.; Marczuk-Kolada, G.; Rosłan, M.; Holownia, A. Cytotoxicity of Methacrylate Dental Resins to Human Gingival Fibroblasts. J. Funct. Biomater. 2022, 13, 56. [Google Scholar] [CrossRef]
- Yang, Y.; Wang, L.; Huang, Z.; Ge, L.; Shi, J. N-acetylcysteine as a novel methacrylate-based resin cement component: Effect on cell apoptosis and genotoxicity in human gingival fibroblasts. BMC Oral Health 2024, 24, 222. [Google Scholar] [CrossRef]
- Masuki, K.; Nomura, Y.; Bhawal, U.K.; Sawajiri, M.; Hirata, I.; Nahara, Y.; Okazaki, M. Apoptotic and necrotic influence of dental resin polymerization initiators in human gingival fibroblast cultures. Dent. Mater. J. 2007, 26, 861–869. [Google Scholar] [CrossRef]
- Lefeuvre, M.; Amjaad, W.; Goldberg, M.; Stanislawski, L. TEGDMA induces mitochondrial damage and oxidative stress in human gingival fibroblasts. Biomaterials 2005, 26, 5130–5137. [Google Scholar] [CrossRef] [PubMed]
- Nassiri, I.; McCall, M.N. Systematic exploration of cell morphological phenotypes associated with a transcriptomic query. Nucleic Acids Res. 2018, 46, e116. [Google Scholar] [CrossRef] [PubMed]
- Prasad, A.; Alizadeh, E. Cell Form and Function: Interpreting and Controlling the Shape of Adherent Cells. Trends Biotechnol. 2019, 37, 347–357. [Google Scholar] [CrossRef]
- Pincus, Z.; Theriot, J.A. Comparison of quantitative methods for cell-shape analysis. J. Microsc. 2007, 227 Pt 2, 140–156. [Google Scholar] [CrossRef]
- Lai, F.; Shen, Z.; Wen, H.; Chen, J.; Zhang, X.; Lin, P.; Yin, D.; Cui, H.; Chen, X. A Morphological identification cell cytotoxicity assay using cytoplasm-localized fluorescent probe (CLFP) to distinguish living and dead cells. Biochem. Biophys. Res. Commun. 2017, 482, 257–263. [Google Scholar] [CrossRef]
- Son, R.; Yamazawa, K.; Oguchi, A.; Suga, M.; Tamura, M.; Yanagita, M.; Murakawa, Y.; Kume, S. Morphomics via next-generation electron microscopy. J. Mol. Cell Biol. 2024, 15, mjad081. [Google Scholar] [CrossRef]
- Available online: https://www.the-scientist.com/enhancing-cell-morphology-based-analysis-71249 (accessed on 25 November 2024).
- Available online: https://www.drugdiscoverytrends.com/deepcells-rem-i-platform-marries-ai-with-morphological-analysis-for-drug-discovery (accessed on 25 November 2024).
- Tromans-Coia, C.; Jamali, N.; Abbasi, H.S.; Giuliano, K.A.; Hagimoto, M.; Jan, K.; Kaneko, E.; Letzsch, S.; Schreiner, A.; Sexton, J.Z.; et al. Assessing the performance of the Cell Painting assay across different imaging systems. Cytom. A 2023, 103, 915–926. [Google Scholar] [CrossRef]
- Bray, M.A.; Gustafsdottir, S.M.; Rohban, M.H.; Singh, S.; Ljosa, V.; Sokolnicki, K.L.; Bittker, J.A.; Bodycombe, N.E.; Dancík, V.; Hasaka, T.P.; et al. A dataset of images and morphological profiles of 30,000 small-molecule treatments using the Cell Painting assay. Gigascience 2017, 6, giw014. [Google Scholar] [CrossRef]
- Bray, M.A.; Singh, S.; Han, H.; Davis, C.T.; Borgeson, B.; Hartland, C.; Kost-Alimova, M.; Gustafsdottir, S.M.; Gibson, C.C.; Carpenter, A.E. Cell Painting, a high-content image-based assay for morphological profiling using multiplexed fluorescent dyes. Nat. Protoc. 2016, 11, 1757–1774. [Google Scholar] [CrossRef] [PubMed]
- Nyffeler, J.; Willis, C.; Harris, F.R.; Foster, M.J.; Chambers, B.; Culbreth, M.; Brockway, R.E.; Davidson-Fritz, S.; Dawson, D.; Shah, I.; et al. Application of Cell Painting for chemical hazard evaluation in support of screening-level chemical assessments. Toxicol. Appl. Pharmacol. 2023, 468, 116513. [Google Scholar] [CrossRef] [PubMed]
- Willis, C.; Nyffeler, J.; Harrill, J. Phenotypic Profiling of Reference Chemicals across Biologically Diverse Cell Types Using the Cell Painting Assay. SLAS Discov. 2020, 25, 755–769. [Google Scholar] [CrossRef] [PubMed]
- Way, G.P.; Kost-Alimova, M.; Shibue, T.; Harrington, W.F.; Gill, S.; Piccioni, F.; Becker, T.; Shafqat-Abbasi, H.; Hahn, W.C.; Carpenter, A.E.; et al. Predicting cell health phenotypes using image-based morphology profiling. Mol. Biol. Cell 2021, 32, 995–1005. [Google Scholar] [CrossRef]
- Beacham, D.; Mandavilli, B.; Holly, R.; Chambers, K.; Oon, C.; Mohr, B.; Hu, Y.-Z.; Yan, J. New-Generation, Functional Probes for Caspase 3/7 and Mitochondrial Superoxide and Ready-Use Phenotypic Cell Painting Tools. Thermo Fisher Scientific, Eugene, Oregon, USA. Available online: https://assets.thermofisher.com/TFS-Assets/BID/posters/caspase-3-7-mitochondrial-superoxide-cell-painting-poster.pdf (accessed on 15 January 2025).
- Belton, C.M.; Izutsu, K.T.; Goodwin, P.C.; Park, Y.; Lamont, R.J. Fluorescence image analysis of the association between Porphyromonas gingivalis and gingival epithelial cells. Cell Microbiol. 1999, 1, 215–223. [Google Scholar] [CrossRef]
- Sunny, S.P.; Khan, A.I.; Rangarajan, M.; Hariharan, A.; Birur N, P.; Pandya, H.J.; Shah, N.; Kuriakose, M.A.; Suresh, A. Oral epithelial cell segmentation from fluorescent multichannel cytology images using deep learning. Comput. Methods Programs Biomed. 2022, 227, 107205. [Google Scholar] [CrossRef]
- Akner, G.; Mossberg, K.; Wikström, A.C.; Sundqvist, K.G.; Gustafsson, J.A. Evidence for colocalization of glucocorticoid receptor with cytoplasmic microtubules in human gingival fibroblasts, using two different monoclonal anti-GR antibodies, confocal laser scanning microscopy and image analysis. J. Steroid Biochem. Mol. Biol. 1991, 39, 419–432. [Google Scholar] [CrossRef]
- Al-Nazhan, S.; Spangberg, L. Morphological cell changes due to chemical toxicity of a dental material: An electron microscopic study on human periodontal ligament fibroblasts and L929 cells. J. Endod. 1990, 16, 129–134. [Google Scholar] [CrossRef]
- Cecato, R.C.; Martinez, E.F.; Benfatti, C.A.M. Analysis of the Viability and Morphology of Gingival Cells on Materials Used in Novel Prosthetic Components: In Vitro Study. J. Contemp. Dent. Pract. 2022, 23, 22–30. [Google Scholar] [CrossRef] [PubMed]
- Yildiz, H.; Sen, E.; Dalcik, H.; Meseli, S.E. Evaluation of cell morphology and adhesion capacity of human gingival fibroblasts on titanium discs with different roughened surfaces: An in vitro scanning electron microscope analysis and cell culture study. Folia Morphol. 2023, 82, 63–71. [Google Scholar] [CrossRef] [PubMed]
- Esfahanizadeh, N.; Motalebi, S.; Daneshparvar, N.; Akhoundi, N.; Bonakdar, S. Morphology, proliferation, and gene expression of gingival fibroblasts on Laser-Lok, titanium, and zirconia surfaces. Lasers Med. Sci. 2016, 31, 863–873. [Google Scholar] [CrossRef] [PubMed]
- Chan, C.P.; Jeng, J.H.; Hsieh, C.C.; Lin, C.L.; Lei, D.; Chang, M.C. Morphological alterations associated with the cytotoxic and cytostatic effects of citric acid on cultured human dental pulp cells. J. Endod. 1999, 25, 354–358. [Google Scholar] [CrossRef] [PubMed]
- Noites, R.; Tavares, I.; Cardoso, M.; Carreira, I.M.; Bartolomeu, M.; Duarte, A.S.; Ribeiro, I.P. Human Gingival Fibroblasts Response to Different Endodontic Sealers: An In Vitro Study. Appl. Sci. 2023, 13, 10976. [Google Scholar] [CrossRef]
- Saramet, V.; Stan, M.S.; Ripszky Totan, A.; Țâncu, A.M.C.; Voicu-Balasea, B.; Enasescu, D.S.; Rus-Hrincu, F.; Imre, M. Analysis of Gingival Fibroblasts Behaviour in the Presence of 3D-Printed versus Milled Methacrylate-Based Dental Resins-Do We Have a Winner? J. Funct. Biomater. 2024, 15, 147. [Google Scholar] [CrossRef]
- Lee, H.; Lee, J.Y.; Ha, D.H.; Jeong, J.H.; Park, J.B. Effects of Valproic Acid on Morphology, Proliferation, and Differentiation of Mesenchymal Stem Cells Derived from Human Gingival Tissue. Implant. Dent. 2018, 27, 33–42. [Google Scholar] [CrossRef]
- Ha, D.H.; Yong, C.S.; Kim, J.O.; Jeong, J.H.; Park, J.B. Effects of tacrolimus on morphology, proliferation and differentiation of mesenchymal stem cells derived from gingiva tissue. Mol. Med. Rep. 2016, 14, 69–76. [Google Scholar] [CrossRef]
- Messer, R.L.; Bishop, S.; Lucas, L.C. Effects of metallic ion toxicity on human gingival fibroblasts morphology. Biomaterials 1999, 20, 1647–1657. [Google Scholar] [CrossRef]
- Jeong, S.H.; Lee, J.E.; Jin, S.H.; Ko, Y.; Park, J.B. Effects of Asiasari radix on the morphology and viability of mesenchymal stem cells derived from the gingiva. Mol. Med. Rep. 2014, 10, 3315–3319. [Google Scholar] [CrossRef]
- Dusinska, M.; Rundén-Pran, E.; Schnekenburger, J.; Kanno, J. Toxicity Tests: In Vitro and In Vivo. In Adverse Effects of Engineered Nanomaterials, 2nd ed.; Fadeel, B., Pietroiusti, A., Shvedova, A.A., Eds.; Academic Press: Cambridge, MA, USA, 2017; pp. 51–82. [Google Scholar]
- Corvi, R.; Madia, F. In vitro genotoxicity testing-Can the performance be enhanced? Food Chem. Toxicol. 2017, 106 Pt B, 600–608. [Google Scholar] [CrossRef]
- Sommer, S.; Buraczewska, I.; Kruszewski, M. Micronucleus Assay: The State of Art, and Future Directions. Int. J. Mol. Sci. 2020, 21, 1534. [Google Scholar] [CrossRef] [PubMed]
- Beedanagari, S. Genetic Toxicology. In Comprehensive Medicinal Chemistry III; Chackalamannil, S., Rotella, D., Ward, S.E., Eds.; Elsevier: Amsterdam, The Netherlands, 2017; pp. 195–203. [Google Scholar]
- Ivashkevich, A.; Redon, C.E.; Nakamura, A.J.; Martin, R.F.; Martin, O.A. Use of the γ-H2AX assay to monitor DNA damage and repair in translational cancer research. Cancer Lett. 2012, 327, 123–133. [Google Scholar] [CrossRef] [PubMed]
- Kuo, L.J.; Yang, L.X. Gamma-H2AX—A novel biomarker for DNA double-strand breaks. In Vivo 2008, 22, 305–309. [Google Scholar] [PubMed]
- Lottner, S.; Shehata, M.; Hickel, R.; Reichl, F.-X.; Durner, J. Effects of antioxidants on DNA-double strand breaks in human gingival fibroblasts exposed to methacrylate based monomers. Dent. Mater. 2013, 29, 991–998. [Google Scholar] [CrossRef]
- Carvalho, N.C.; Guedes, S.A.G.; Albuquerque-Júnior, R.L.C.; de Albuquerque, D.S.; de Souza Araújo, A.A.; Paranhos, L.R.; Camargo, S.E.A.; Ribeiro, M.A.G. Analysis of Aloe vera cytotoxicity and genotoxicity associated with endodontic medication and laser photobiomodulation. J. Photochem. Photobiol. B 2018, 178, 348–354. [Google Scholar] [CrossRef]
- Rode, S.M.; Sato, T.D.P.; Matos, F.S.; Correia, A.M.O.; Camargo, S.E.A. Toxicity and effect of whitening toothpastes on enamel surface. Braz. Oral Res. 2021, 35, e025. [Google Scholar] [CrossRef] [PubMed]
- Yagci, F.; Kesim, B. Cytotoxic and genotoxic effects on gingival fibroblasts from static magnetic fields produced by dental magnetic attachments. Gerodontology 2016, 33, 421–427. [Google Scholar] [CrossRef]
- Ferreira, E.C.C.; Gonçalves, T.M.; Pereira, T.C.; Hasna, A.A.; Oliveira, F.E.d.; Jorjão, A.L.; Camargo, S.E.A.; Oliveira, L.D.d.; Spalding, M. The biocompatibility of Achyrocline satureioides plant extract over human gingival fibroblasts. Res. Soc. Dev. 2021, 10, e37610111902. [Google Scholar] [CrossRef]
- Yang, M.L. The effects of cytotoxicity and genotoxicity induced by 2,2-bis[4-(acryloxypropoxy)phenyl]propane via caspases in human gingival fibroblasts. Toxicol. Ind. Health 2014, 30, 755–764. [Google Scholar] [CrossRef]
- Loyola-Rodríguez, J.P.; Lastra-Corso, I.; García-Cortés, J.O.; Loyola-Leyva, A.; Domínguez-Pérez, R.A.; Avila-Arizmendi, D.; Contreras-Palma, G.; González-Calixto, C. In Vitro Determination of Genotoxicity Induced by Brackets Alloys in Cultures of Human Gingival Fibroblasts. J. Toxicol. 2020, 2020, 1467456, Erratum in J. Toxicol. 2021, 2021, 4028481. https://doi.org/10.1155/2021/4028481. [Google Scholar] [CrossRef]
- Ahuja, D.; Jose, N.P.; Kamal, R.; Panduranga, V.; Nambiar, S.; Isloor, A.M. In vitro determination of genotoxicity and cytotoxicity induced by stainless steel brackets with and without surface coating in cultures of oral mucosal cells. BMC Oral Health 2024, 24, 1233. [Google Scholar] [CrossRef] [PubMed]
- Ortiz, A.J.; Fernández, E.; Vicente, A.; Calvo, J.L.; Ortiz, C. Metallic ions released from stainless steel, nickel-free, and titanium orthodontic alloys: Toxicity and DNA damage. Am. J. Orthod. Dentofac. Orthop. 2011, 140, e115–e122, Erratum in Am. J. Orthod. Dentofacial Orthop. 2018, 153, 765. https://doi.org/10.1016/j.ajodo.2018.03.012. [Google Scholar] [CrossRef]
- Tomakidi, P.; Koke, U.; Kern, R.; Erdinger, L.; Krüger, H.; Kohl, A.; Komposch, G. Assessment of acute cyto- and genotoxicity of corrosion eluates obtained from orthodontic materials using monolayer cultures of immortalized human gingival keratinocytes. J. Orofac. Orthop. 2000, 61, 2–19, English, German. [Google Scholar] [CrossRef] [PubMed]
- Volk, J.; Leyhausen, G.; Geurtsen, W. Glutathione level and genotoxicity in human oral keratinocytes exposed to TEGDMA. J. Biomed. Mater. Res. B Appl. Biomater. 2012, 100, 391–399. [Google Scholar] [CrossRef] [PubMed]
- Szczepanska, J.; Poplawski, T.; Synowiec, E.; Pawlowska, E.; Chojnacki, C.J.; Chojnacki, J.; Blasiak, J. 2-hydroxylethyl methacrylate (HEMA), a tooth restoration component, exerts its genotoxic effects in human gingival fibroblasts through methacrylic acid, an immediate product of its degradation. Mol. Biol. Rep. 2012, 39, 1561–1574. [Google Scholar] [CrossRef]
- Volk, J.; Ziemann, C.; Leyhausen, G.; Geurtsen, W. Non-irradiated campherquinone induces DNA damage in human gingival fibroblasts. Dent. Mater. 2009, 25, 1556–1563. [Google Scholar] [CrossRef]
- Sundar, I.K.; Javed, F.; Romanos, G.E.; Rahman, I. E-cigarettes and flavorings induce inflammatory and pro-senescence responses in oral epithelial cells and periodontal fibroblasts. Oncotarget 2016, 7, 77196–77204. [Google Scholar] [CrossRef]
- Laiteerapong, A.; Reichl, F.X.; Yang, Y.; Hickel, R.; HÖgg, C. Induction of DNA double-strand breaks in human gingival fibroblasts by eluates from titanium dioxide modified glass ionomer cements. Dent. Mater. 2018, 34, 282–287. [Google Scholar] [CrossRef]
- Shehata, M.; Durner, J.; Eldenez, A.; Van Landuyt, K.; Styllou, P.; Rothmund, L.; Hickel, R.; Scherthan, H.; Geurtsen, W.; Kaina, B.; et al. Cytotoxicity and induction of DNA double-strand breaks by components leached from dental composites in primary human gingival fibroblasts. Dent. Mater. 2013, 29, 971–979. [Google Scholar] [CrossRef]
- Chang, Y.C.; Tai, K.W.; Cheng, M.H.; Chou, L.S.; Chou, M.Y. Cytotoxic and non-genotoxic effects of arecoline on human buccal fibroblasts in vitro. J. Oral Pathol. Med. 1998, 27, 68–71. [Google Scholar] [CrossRef] [PubMed]
- Żukowski, P.; Maciejczyk, M.; Waszkiel, D. Sources of free radicals and oxidative stress in the oral cavity. Arch. Oral Biol. 2018, 92, 8–17. [Google Scholar] [CrossRef] [PubMed]
- Kumar, J.; Teoh, S.L.; Das, S.; Mahakknaukrauh, P. Oxidative Stress in Oral Diseases: Understanding Its Relation with Other Systemic Diseases. Front. Physiol. 2017, 8, 693. [Google Scholar] [CrossRef] [PubMed]
- Goya, L.; Martín, M.A.; Ramos, S.; Mateos, R.; Bravo, L. A cell culture model for the assessment of the chemopreventive potential of dietary compounds. Curr. Nutr. Food Sci. 2009, 5, 56–64. [Google Scholar] [CrossRef]
- Van Landuyt, K.L.; Krifka, S.; Hiller, K.A.; Bolay, C.; Waha, C.; Van Meerbeek, B.; Schmalz, G.; Schweikl, H. Evaluation of cell responses toward adhesives with different photoinitiating systems. Dent. Mater. 2015, 31, 916–927. [Google Scholar] [CrossRef]
- Tejchman, K.; Kotfis, K.; Sieńko, J. Biomarkers and Mechanisms of Oxidative Stress-Last 20 Years of Research with an Emphasis on Kidney Damage and Renal Transplantation. Int. J. Mol. Sci. 2021, 22, 8010. [Google Scholar] [CrossRef]
- Alomari, E.; Bruno, S.; Ronda, L.; Paredi, G.; Bettati, S.; Mozzarelli, A. Protein carbonylation detection methods: A comparison. Data Brief. 2018, 19, 2215–2220. [Google Scholar] [CrossRef]
- Weber, D.; Davies, M.J.; Grune, T. Determination of protein carbonyls in plasma, cell extracts, tissue homogenates, isolated proteins: Focus on sample preparation and derivatization conditions. Redox Biol. 2015, 5, 367–380. [Google Scholar] [CrossRef]
- Krifka, S.; Seidenader, C.; Hiller, K.A.; Schmalz, G.; Schweikl, H. Oxidative stress and cytotoxicity generated by dental composites in human pulp cells. Clin. Oral Investig. 2012, 16, 215–224. [Google Scholar] [CrossRef]
- Chen, C.-S.; Lee, S.-S.; Yu, H.-C.; Huang, F.-M.; Chang, Y.-C. Effects of nicotine on cell growth, migration, and production of inflammatory cytokines and reactive oxygen species by cementoblasts. J. Dent. Sci. 2015, 10, 154–160. [Google Scholar] [CrossRef]
- Sancilio, S.; Gallorini, M.; Cataldi, A.; di Giacomo, V. Cytotoxicity and apoptosis induction by e-cigarette fluids in human gingival fibroblasts. Clin. Oral Investig. 2016, 20, 477–483. [Google Scholar] [CrossRef]
- Lleperuma, R.P.; Kim, D.K.; Park, Y.J.; Son, H.K.; Kim, J.Y.; Kim, J.; Lee, D.Y.; Kim, K.Y.; Jung, D.W.; Tilakaratne, W.M.; et al. Areca nut exposure increases secretion of tumor-promoting cytokines in gingival fibroblasts that trigger DNA damage in oral keratinocytes. Int. J. Cancer 2015, 137, 2545–2557. [Google Scholar] [CrossRef]
- Pinheiro, A.C.; Ombredane, A.S.; Pinheiro, W.O.; Andrade, L.R.; Silva, V.R.P.; Felice, G.J.; Alves, D.S.; Albernaz, A.F.; Silveira, A.P.; Lima, M.C.F.; et al. Evaluation of Biocompatibility, Anti-Inflammatory, and Antinociceptive Activities of Pequi Oil-Based Nanoemulsions in In Vitro and In Vivo Models. Nanomaterials 2022, 12, 4260. [Google Scholar] [CrossRef] [PubMed]
- Liu, C.; Chu, D.; Kalantar-Zadeh, K.; George, J.; Young, H.A.; Liu, G. Cytokines: From Clinical Significance to Quantification. Adv. Sci. 2021, 8, e2004433. [Google Scholar] [CrossRef] [PubMed]
- Kollmuss, M.; Edelhoff, D.; Schwendicke, F.; Wuersching, S.N. In Vitro Cytotoxic and Inflammatory Response of Gingival Fibroblasts and Oral Mucosal Keratinocytes to 3D Printed Oral Devices. Polymers 2024, 16, 1336. [Google Scholar] [CrossRef] [PubMed]
- Brenner, D.R.; Scherer, D.; Muir, K.; Schildkraut, J.; Boffetta, P.; Spitz, M.R.; Le Marchand, L.; Chan, A.T.; Goode, E.L.; Ulrich, C.M.; et al. A review of the application of inflammatory biomarkers in epidemiologic cancer research. Cancer Epidemiol. Biomark. Prev. 2014, 23, 1729–1751. [Google Scholar] [CrossRef]
- Atanasova, T.; Stankova, T.; Bivolarska, A.; Vlaykova, T. Matrix Metalloproteinases in Oral Health-Special Attention on MMP-8. Biomedicines 2023, 11, 1514. [Google Scholar] [CrossRef]
- Trubiani, O.; Toniato, E.; Di Iorio, D.; Diomede, F.; Merciaro, I.; D’ Arcangelo, C.; Caputi, S. Morphological analysis and interleukin release in human gingival fibroblasts seeded on different denture base acrylic resins. Int. J. Immunopathol. Pharmacol. 2012, 25, 637–643. [Google Scholar] [CrossRef]
- Massaro, H.; Zambelli, L.F.A.; Britto, A.A.; Vieira, R.P.; Ligeiro-de-Oliveira, A.P.; Andia, D.C.; Oliveira, M.T.; Lima, A.F. Solvent and HEMA Increase Adhesive Toxicity and Cytokine Release from Dental Pulp Cells. Materials 2019, 12, 2750. [Google Scholar] [CrossRef]
- Rinaldi, S.; Pieper, E.; Schulz, T.; Zimmermann, R.; Luch, A.; Laux, P.; Mallock-Ohnesorg, N. Oral nicotine pouches with an aftertaste? Part 2: In vitro toxicity in human gingival fibroblasts. Arch. Toxicol. 2023, 97, 2343–2356. [Google Scholar] [CrossRef]
- Kurek-Górecka, A.; Kłósek, M.; Pietsz, G.; Czuba, Z.P.; Kolayli, S.; Can, Z.; Balwierz, R.; Olczyk, P. The Phenolic Profile and Anti-Inflammatory Effect of Ethanolic Extract of Polish Propolis on Activated Human Gingival Fibroblasts-1 Cell Line. Molecules 2023, 28, 7477. [Google Scholar] [CrossRef] [PubMed]
- Jung, J.I.; Kim, S.; Baek, S.M.; Choi, S.I.; Kim, G.H.; Imm, J.Y. Ecklonia cava Extract Exerts Anti-Inflammatory Effect in Human Gingival Fibroblasts and Chronic Periodontitis Animal Model by Suppression of Pro-Inflammatory Cytokines and Chemokines. Foods 2021, 10, 1656. [Google Scholar] [CrossRef] [PubMed]
- Hao, C.; Wu, B.; Hou, Z.; Xie, Q.; Liao, T.; Wang, T.; Ma, D. Asiatic acid inhibits LPS-induced inflammatory response in human gingival fibroblasts. Int. Immunopharmacol. 2017, 50, 313–318, Erratum in Int. Immunopharmacol. 2019, 72, 523. https://doi.org/10.1016/j.intimp.2019.01.042. [Google Scholar] [CrossRef] [PubMed]
- Lee, H.J.; Choi, C.H. Anti-inflammatory effects of bamboo salt and sodium fluoride in human gingival fibroblasts—An in vitro study. Kaohsiung J. Med. Sci. 2015, 31, 303–308. [Google Scholar] [CrossRef]
- Wang, P.-L.; Tachi, Y.; Masuno, K.; Okusa, N.; Imamura, Y. The Study of Ozone Ointment on Human Gingival Fibroblasts Cell Proliferation Ability and Anti-Inflammatory. J. Hard Tissue Biol. 2018, 27, 209–212. [Google Scholar] [CrossRef]
- Ono, M.; Kantoh, K.; Ueki, J.; Shimada, A.; Wakabayashi, H.; Matsuta, T.; Sakagami, H.; Kumada, H.; Hamada, N.; Kitajima, M.; et al. Quest for anti-inflammatory substances using IL-1β-stimulated gingival fibroblasts. Vivo 2011, 25, 763–768. [Google Scholar]
- AlFatlawi, Z.; Huang, M.; Chau, D.Y.S.; D’Aiuto, F. Three dimensional (3D) gingival models in periodontal research: A systematic review. J. Mater. Sci. Mater. Med. 2023, 34, 58. [Google Scholar] [CrossRef]
- Klausner, M.; Handa, Y.; Aizawa, S. In vitro three-dimensional organotypic culture models of the oral mucosa. Vitro Cell Dev. Biol. Anim. 2021, 57, 148–159. [Google Scholar] [CrossRef]
- Riaz, A.; Gidvall, S.; Prgomet, Z.; Hernandez, A.R.; Ruzgas, T.; Nilsson, E.J.; Davies, J.; Valetti, S. Three-Dimensional Oral Mucosal Equivalents as Models for Transmucosal Drug Permeation Studies. Pharmaceutics 2023, 15, 1513. [Google Scholar] [CrossRef]
- Aizawa, S.; Yoshida, H.; Umeshita, K.; Watanabe, S.; Takahashi, Y.; Sakane, S.; Sakaguchi, H.; Kataoka, S. Development of an oral mucosal irritation test using a three-dimensional human buccal oral mucosal model. Toxicol. Vitro 2023, 87, 105519. [Google Scholar] [CrossRef]
- Available online: www.mattek.com/mattekproduct/epioral-epigingival/ (accessed on 10 January 2025).
- Hadjichristou, C.; Papachristou, E.; Bonovolias, I.; Bakopoulou, A. Three-dimensional tissue engineering-based Dentin/Pulp tissue analogue as advanced biocompatibility evaluation tool of dental restorative materials. Dent. Mater. 2020, 36, 229–248. [Google Scholar] [CrossRef]
- Yang, J.; Deol, G.; Myangar, N. Retention of o-cymen-5-ol and zinc on reconstructed human gingival tissue from a toothpaste formulation. Int. Dent. J. 2011, 61 (Suppl. 3), 41–45. [Google Scholar] [CrossRef] [PubMed]
- Klausner, M.; Ayehunie, S.; Breyfogle, B.A.; Wertz, P.W.; Bacca, L.; Kubilus, J. Organotypic human oral tissue models for toxicological studies. Toxicol. Vitro 2007, 21, 938–949. [Google Scholar] [CrossRef] [PubMed]
- Moharamzadeh, K.; Brook, I.M.; Scutt, A.M.; Thornhill, M.H.; Van Noort, R. Mucotoxicity of dental composite resins on a tissue-engineered human oral mucosal model. J. Dent. 2008, 36, 331–336. [Google Scholar] [CrossRef] [PubMed]
- Koschier, F.; Kostrubsky, V.; Toole, C.; Gallo, M.A. In vitro effects of ethanol and mouthrinse on permeability in an oral buccal mucosal tissue construct. Food Chem. Toxicol. 2011, 49, 2524–2529, Erratum in Food Chem. Toxicol. 2012, 50, 1815. [Google Scholar] [CrossRef]
- Song, G.; Banov, D.; Bassani, A.S.; Valdez, B.C. Evaluation of the Safety, Cell Migration, and Mucoadhesive Properties of a Mucoadhesive Polymer Blend in Human Oral Mucosa. AAPS PharmSciTech 2017, 18, 1617–1623. [Google Scholar] [CrossRef]
- Vande Vannet, B.; Hanssens, J.L.; Wehrbein, H. The use of three-dimensional oral mucosa cell cultures to assess the toxicity of soldered and welded wires. Eur. J. Orthod. 2007, 29, 60–66. [Google Scholar] [CrossRef]
- Zwicker, P.; Zumpe, M.; Kramer, A.; Müller, G. A 3D Model of Human Buccal Mucosa for Compatibility Testing of Mouth Rinsing Solutions. Pharmaceutics 2023, 15, 721. [Google Scholar] [CrossRef]
- 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. [Google Scholar] [CrossRef]
- Colley, H.E.; Said, Z.; Santocildes-Romero, M.E.; Baker, S.R.; D’Apice, K.; Hansen, J.; Madsen, L.S.; Thornhill, M.H.; Hatton, P.V.; Murdoch, C. Pre-clinical evaluation of novel mucoadhesive bilayer patches for local delivery of clobetasol-17-propionate to the oral mucosa. Biomaterials 2018, 178, 134–146. [Google Scholar] [CrossRef]
- Zanetti, F.; Sewer, A.; Mathis, C.; Iskandar, A.R.; Kostadinova, R.; Schlage, W.K.; Leroy, P.; Majeed, S.; Guedj, E.; Trivedi, K.; et al. 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, 1252–1269. [Google Scholar] [CrossRef]
- Zanetti, F.; Titz, B.; Sewer, A.; Lo Sasso, G.; Scotti, E.; Schlage, W.K.; Mathis, C.; Leroy, P.; Majeed, S.; Torres, L.O.; et al. Comparative systems toxicology analysis of cigarette smoke and aerosol from a candidate modified risk tobacco product in organotypic human gingival epithelial cultures: A 3-day repeated exposure study. Food Chem. Toxicol. 2017, 101, 15–35. [Google Scholar] [CrossRef] [PubMed]
- Miyauchi, M.; Ishikawa, S.; Kurachi, T.; Sakamoto, K.; Sakai, H. Oral Absorption across Organotypic Culture Models of the Human Buccal Epithelium after E-cigarette Aerosol Exposure. ACS Omega 2022, 7, 45574–45581. [Google Scholar] [CrossRef]
- Zanetti, F.; Sewer, A.; Scotti, E.; Titz, B.; Schlage, W.K.; Leroy, P.; Kondylis, A.; Vuillaume, G.; Iskandar, A.R.; Guedj, E.; et al. Assessment of the impact of aerosol from a potential modified risk tobacco product compared with cigarette smoke on human organotypic oral epithelial cultures under different exposure regimens. Food Chem. Toxicol. 2018, 115, 148–169. [Google Scholar] [CrossRef] [PubMed]
- Miller-Holt, J.; Baskerville-Abraham, I.; Sakimura, M.; Fukushima, T.; Puglisi, A.; Gafner, J. In vitro evaluation of mutagenic, cytotoxic, genotoxic and oral irritation potential of nicotine pouch products. Toxicol. Rep. 2022, 9, 1316–1324. [Google Scholar] [CrossRef] [PubMed]
- Shaikh, Z.N.; Alqahtani, A.; Almela, T.; Franklin, K.; Tayebi, L.; Moharamzadeh, K. Effects of electronic cigarette liquid on monolayer and 3D tissue-engineered models of human gingival mucosa. J. Adv. Periodontol. Implant. Dent. 2019, 11, 54–62. [Google Scholar] [CrossRef]
- Kurosawa, Y.; Yamaguchi, H.; Uemichi, K.; Shinozuka, K.; Kirihara, Y.; Tsuda, H. Butyrate-treatment induces gingival epithelial cell death in a three-dimensional gingival-connective tissue hybrid co-culture system. J. Dent. Sci. 2023, 18, 893–897. [Google Scholar] [CrossRef]
- Agrawal, A.; Shindell, E.; Jordan, F.; Baeva, L.; Pfefer, J.; Godar, D.E. UV radiation increases carcinogenic risks for oral tissues compared to skin. Photochem. Photobiol. 2013, 89, 1193–1198. [Google Scholar] [CrossRef]
- Breger, J.; Baeva, L.; Agrawal, A.; Shindell, E.; Godar, D.E. UVB-induced inflammatory cytokine release, DNA damage and apoptosis of human oral compared with skin tissue equivalents. Photochem. Photobiol. 2013, 89, 665–670. [Google Scholar] [CrossRef]
- Basso, F.G.; Soares, D.G.; de Souza Costa, C.A.; Hebling, J. Low-level laser therapy in 3D cell culture model using gingival fibroblasts. Lasers Med. Sci. 2016, 31, 973–978. [Google Scholar] [CrossRef]
- Carreiro, A.F.P.; Delben, J.A.; Guedes, S.; Silveira, E.J.D.; Janal, M.N.; Vergani, C.E.; Pushalkar, S.; Duarte, S. Low-temperature plasma on peri-implant-related biofilm and gingival tissue. J. Periodontol. 2019, 90, 507–515. [Google Scholar] [CrossRef]
- Moghaddam, B.; Yang, J.; Roohpour, N. Biologic evaluation of devices with chronic exposure using 3D human gingival model. In Proceedings of the 10th World Biomaterials Congress, Montréal, QC, Canada, 17–22 May 2016. [Google Scholar] [CrossRef]
- Avnet, S.; Di Pompo, G.; Borciani, G.; Fischetti, T.; Graziani, G.; Baldini, N. Advantages and limitations of using cell viability assays for 3Dbioprinted constructs. Biomed. Mater. 2024, 19, 025033. [Google Scholar] [CrossRef] [PubMed]
- Dominijanni, A.J.; Devarasetty, M.; Forsythe, S.D.; Votanopoulos, K.I.; Soker, S. Cell Viability Assays in Three-Dimensional Hydrogels: A Comparative Study of Accuracy. Tissue Eng. Part. C Methods 2021, 27, 401–410. [Google Scholar] [CrossRef]
- Pfuhler, S.; van Benthem, J.; Curren, R.; Doak, S.H.; Dusinska, M.; Hayashi, M.; Heflich, R.H.; Kidd, D.; Kirkland, D.; Luan, Y.; et al. Use of in vitro 3D tissue models in genotoxicity testing: Strategic fit, validation status and way forward. Report of the working group from the 7th International Workshop on Genotoxicity Testing (IWGT). Mutat. Res. Genet. Toxicol. Environ. Mutagen. 2020, 850–851, 503135. [Google Scholar] [CrossRef] [PubMed]
- Joseph, X.; Akhil, V.; Arathi, A.; Mohanan, P.V. Comprehensive Development in Organ-On-A-Chip Technology. J. Pharm. Sci. 2022, 111, 18–31. [Google Scholar] [CrossRef] [PubMed]
- Koyilot, M.C.; Natarajan, P.; Hunt, C.R.; Sivarajkumar, S.; Roy, R.; Joglekar, S.; Pandita, S.; Tong, C.W.; Marakkar, S.; Subramanian, L.; et al. Breakthroughs and Applications of Organ-on-a-Chip Technology. Cells 2022, 11, 1828. [Google Scholar] [CrossRef]
- Ingber, D.E. Human organs-on-chips for disease modelling, drug development and personalized medicine. Nat. Rev. Genet. 2022, 23, 467–491. [Google Scholar] [CrossRef]
- Deng, S.; Li, C.; Cao, J.; Cui, Z.; Du, J.; Fu, Z.; Yang, H.; Chen, P. Organ-on-a-chip meets artificial intelligence in drug evaluation. Theranostics 2023, 13, 4526–4558. [Google Scholar] [CrossRef]
- Huang, C.; Sanaei, F.; Verdurmen, W.P.R.; Yang, F.; Ji, W.; Walboomers, X.F. The Application of Organs-on-a-Chip in Dental, Oral, and Craniofacial Research. J. Dent. Res. 2023, 102, 364–375. [Google Scholar] [CrossRef]
- Pierfelice, T.V.; D’Amico, E.; Petrini, M.; Romano, M.; D’Arcangelo, C.; Sbordone, L.; Barone, A.; Plebani, R.; Iezzi, G. A Systematic Review on Organ-on-a-Chip in PDMS or Hydrogel in Dentistry: An Update of the Literature. Gels 2024, 10, 102. [Google Scholar] [CrossRef]
- Hu, S.; Muniraj, G.; Mishra, A.; Hong, K.; Lum, J.L.; Hong, C.H.L.; 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. [Google Scholar] [CrossRef] [PubMed]
- Muniraj, G.; Tan, R.H.S.; Dai, Y.; Wu, R.; Alberti, M.; Sriram, G. Microphysiological Modeling of Gingival Tissues and Host-Material Interactions Using Gingiva-on-Chip. Adv. Healthc. Mater. 2023, 12, e2301472. [Google Scholar] [CrossRef]
- França, C.M.; Balbinot, G.S.; Cunha, D.; Saboia, V.P.A.; Ferracane, J.; Bertassoni, L.E. In-vitro models of biocompatibility testing for restorative dental materials: From 2D cultures to organs on-a-chip. Acta Biomater. 2022, 150, 58–66. [Google Scholar] [CrossRef]
- ISO 7405:2018; Dentistry—Evaluation of Biocompatibility of Medical Devices Used in Dentistry. International Organization for Standardization: Geneva, Switzerland, 2018.
- França, C.M.; Tahayeri, A.; Rodrigues, N.S.; Ferdosian, S.; Puppin Rontani, R.M.; Sereda, G.; Ferracane, J.L.; Bertassoni, L.E. The tooth on-a-chip: A microphysiologic model system mimicking the biologic interface of the tooth with biomaterials. Lab. Chip 2020, 20, 405–413. [Google Scholar] [CrossRef]
- 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. [Google Scholar] [CrossRef] [PubMed]
- Jin, L.; Kou, N.; An, F.; Gao, Z.; Tian, T.; Hui, J.; Chen, C.; Ma, G.; Mao, H.; Liu, H. Analyzing Human Periodontal Soft Tissue Inflammation and Drug Responses In Vitro Using Epithelium-Capillary Interface On-a-Chip. Biosensors 2022, 12, 345. [Google Scholar] [CrossRef] [PubMed]
- Makkar, H.; Zhou, Y.; Tan, K.S.; Lim, C.T.; Sriram, G. Modeling Crevicular Fluid Flow and Host-Oral Microbiome Interactions in a Gingival Crevice-on-Chip. Adv. Healthc. Mater. 2023, 12, 2202376. [Google Scholar] [CrossRef]
- Rodrigues, N.S.; França, C.M.; Tahayeri, A.; Ren, Z.; Saboia, V.P.A.; Smith, A.J.; Ferracane, J.L.; Koo, H.; Bertassoni, L.E. Biomaterial and Biofilm Interactions with the Pulp-Dentin Complex-on-a-Chip. J. Dent. Res. 2021, 100, 1136–1143. [Google Scholar] [CrossRef]
- Ly, K.L.; Rooholghodos, S.A.; Rahimi, C.; Rahimi, B.; Bienek, D.R.; Kaufman, G.; Raub, C.B.; Luo, X. An Oral-mucosa-on-a-chip sensitively evaluates cell responses to dental monomers. Biomed. Microdevices 2021, 23, 7. [Google Scholar] [CrossRef]
- Rahimi, C.; Rahimi, B.; Padova, D.; Rooholghodos, S.A.; Bienek, D.R.; Luo, X.; Kaufman, G.; Raub, C.B. Oral mucosa-on-a-chip to assess layer-specific responses to bacteria and dental materials. Biomicrofluidics 2018, 12, 054106. [Google Scholar] [CrossRef]
- Jensen, C.; Teng, Y. Is It Time to Start Transitioning from 2D to 3D Cell Culture? Front. Mol. Biosci. 2020, 7, 33. [Google Scholar] [CrossRef] [PubMed]
- Ardila, C.M.; Jiménez-Arbeláez, G.A.; Vivares-Builes, A.M. Potential Clinical Application of Organs-on-a-Chip in Periodontal Diseases: A Systematic Review of In Vitro Studies. Dent. J. 2023, 11, 158. [Google Scholar] [CrossRef] [PubMed]
- Farhang Doost, N.; Srivastava, S.K. A Comprehensive Review of Organ-on-a-Chip Technology and Its Applications. Biosensors 2024, 14, 225. [Google Scholar] [CrossRef] [PubMed]
Method | Type of Cells | Tested Agent | Results | Reference |
---|---|---|---|---|
Dye exclusion assays | ||||
Trypan blue dye exclusion test | Human oral keratinocytes | Biopolymer-coated liposomes (alginate, gellan gum, chitosan) | The formulations presented a low cytotoxicity; cell viability was above 70% in all cases. | [27] |
Human gingival fibroblasts | Zoledronic acid | An intense reduction in the number of viable cells was observed after exposure to zoledronic acid. | [28] | |
Human dental pulp fibroblasts | Dental materials (HEMA and adhesive) | The adhesive system showed significant cytotoxic effects at a concentration of 10−2; cytotoxic effects decreased when the material was polymerized before testing. | [29] | |
Colorimetric assays | ||||
MTT | Human oral keratinocytes | BAPO and TPO—alternative photoinitiators to CQ in dental resinous materials | BAPO and TPO presented higher cytotoxicity than CQ. | [30] |
Human salivary cell gland acinar cell line | Carrageenan | Carrageenan is safe and non-toxic to the oral cavity tissues as an anti-human papillomavirus agent. | [31] | |
Non-keratinized human oral mucosa cells | ZnO, TiO2, SiO2, and hydroxyapatite (bulk particle-sized materials and nanomaterials) | ZnO (70 nm NP and bulk) caused a significant reduction in cell viability; TiO2 (bulk and nano form) was well tolerated by the cells. | [32] | |
Human gingival stroma fibroblasts | Dental adhesives | Short-term stimulation and long-term inhibition of cell viability were observed for the undiluted extracts. | [33] | |
Human gingival and oral fibroblasts | Calcite (calcium carbonate) and zincite (zinc oxide) nanoparticles | Cell viability decreased in a concentration-dependent manner, but the tested materials were regarded as non-cytotoxic. | [34] | |
Rat gingival fibroblasts | Calcium hydroxide and ellagic acid preparations (different ratios) | The mixture calcium hydroxide–ellagic acid was non-toxic and promoted the proliferation of gingival fibroblasts. | [35] | |
Human gingival fibroblasts | Zoledronic acid | Zoledronic acid decreased cell viability by 40%. | [28] | |
Human oral fibroblasts | Acid bone lysate | The viability of human gingival fibroblasts was maintained with a 5% concentration of acid bone lysate. | [36] | |
Odontoblast-like cells (MDPC-23), human dental pulp cells | H2O2 bleaching gel | The cell viability reduction was proportional to the concentration and contact time. | [37] | |
Human oral mucosa fibroblasts | Ultraviolet B radiation | UVB-irradiated cells exhibited significantly reduced proliferation. | [38] | |
Human gingival epithelium keratinocytes | Traditional cigarette smoke and electronic cigarette aerosols | Traditional cigarette smoke and electronic cigarette aerosols decreased cell viability; the acute toxic effect of e-cig aerosols was lower. | [39] | |
Human oral fibroblasts | Cigarette smoke and e-vapor condensates | Cigarette smoke and e-vapor condensates reduced cell viability. | [40] | |
MTS | Human gingival fibroblasts | Single-wall carbon nanotubes | Single-wall carbon nanotubes did not affect cell viability at low concentrations but showed strong cytotoxicity at high concentrations (150 μg/mL). | [41] |
Human oral epithelial cells | Commercial resin bonding agents | Most uncured materials had significant, concentration-dependent, cytotoxic effects, while most post-cured samples revealed no cytotoxicity. | [42] | |
Human oral fibroblasts | Antioxidant mixtures of resveratrol (R), ferulic acid (F), phloretin (P), and tetrahydrocurcuminoids (T)—RFT, PFR, and PFT | Low concentrations of antioxidant mixtures increased viability, but high concentrations reduced cell survival. | [43] | |
Human gingival fibroblasts | Chitosan particles | Chitosan increased cell viability. | [44] | |
XTT | Human oral fibroblasts | Denture adhesives | The tested denture adhesives exhibited no cytotoxic effect. | [45] |
Human gingival fibroblasts | Methacrylate-based adhesives | Cell viability decreased in a concentration-dependent manner. | [46] | |
WST | Human oral keratinocytes | Silver–palladium–gold–indium dental alloy (In3+ released by corrosion) | Low concentrations of In3+ increased cell viability, while higher concentrations (1.6 mM) significantly decreased the percentage of viable cells and might have damaged the human oral epithelium. | [47] |
Human oral epithelial cells | Commercial resin bonding agents | Significant cytotoxic effects were exhibited by most of the uncured materials. | [42] | |
Human oral keratinocytes | Biopolymer-coated liposomes (alginate, gellan gum, and chitosan) | The formulations showed cell proliferation results comparable to the control. | [27] | |
Human gingival fibroblasts | Er:YAG (erbium-doped yttrium–aluminum–garnet) laser irradiation | Cellular proliferation depended on the applied energy levels, with a maximum registered at 6.3 J cm−2. | [48] | |
Human oral epithelial cells | Cisplatin and thymoquinone (alone and in combination) | Thymoquinone presented lower cytotoxicity against normal cells in comparison to cisplatin, but the interaction between cisplatin and thymoquinone was synergistic, increasing the killing effect against normal cells. | [49] | |
NRU | Human gingival epithelial cell | Dental restorative materials | Transwell inserts and the NRU assay proved a reliable method to assess cytotoxicity of dental materials. | [50] |
Human gingival fibroblasts | Mouthwashes | The products presented a dose-dependent cytotoxicity but can be considered safe in diluted solutions. | [51] | |
Human gingival fibroblasts | Tea polyphenols (ECG, EGCG, EC, EGC, CG, and C) | ECG and CG exhibited high toxicity, EGCG was moderately toxic, and EGC, C, and EC presented low toxicity (after a 3-day exposure). | [52] | |
Human gingival fibroblasts | Theaflavin mixture purified from black tea | The theaflavin mixture exhibited cytotoxic effects only at high concentrations; normal cells were less susceptible to cytotoxicity than cancerous cells. | [53] | |
Human oral epithelial cells | Liquids for electronic cigarettes | There was heterogeneity across e-liquids in terms of cytotoxicity; the presence of certain flavoring agents (ethyl maltol, vanillin, maltol, and ethyl vanillin) increased the risks. | [54] | |
SRB | Human gingival epithelial cell | Cigarette smoke total particulate matter, smokeless tobacco extracted with complete artificial saliva, and whole-smoke conditioned media. | Combusted products exhibited higher cytotoxicity; nicotine alone did not show significant signs of cytotoxicity in the applied range. | [55] |
LDH | Human gingival fibroblasts | Novobiocin | The percentage of LDH-releasing cells increased in a concentration-dependent manner. | [56] |
Non-keratinized human oral mucosa cells | ZnO, TiO2, SiO2, and hydroxyapatite (bulk particle-sized materials and nanomaterials) | ZnO (bulk and 70 nm NP) caused a significant increase in LDH release. | [32] | |
Human gingival fibroblasts | ZnO nanoparticles | MTT assay proved to be a more sensitive marker for cytotoxicity. | [57] | |
Human gingival fibroblasts | Chitosan particles | ZnO NP increased cytoplasmic LDH release only at high concentrations. | [44] | |
Human gingival epithelial cells | E-cigarette vapor | LDH levels were not significantly altered. | [58] | |
Acid phosphatase | Human oral fibroblasts | ZrO2 nanoparticles reinforced 3D-printed dental resins | The addition of ZrO2 nanoparticles did not interfere with biocompatibility; 3D-printed specimens were noncytotoxic. | [59] |
Fluorimetric assays | ||||
Resazurin-based assays (alamarBlue, PrestoBlue) | Human oral gingival epithelial keratinocytes, oral gingival fibroblasts | Long-chain bases (sphingosine, dihydrosphingosine, and phytosphingosine) | Gingival epithelial keratinocytes were more resistant to the three long-chain bases than gingival fibroblasts. | [60] |
Human gingival fibroblasts | Areca nut (Areca catechu L.) products | Areca nut extracts exhibited necrotizing and cytotoxic effects. | [61] | |
Human gingival fibroblasts | Silver tungstate (α-Ag2WO4) microcrystals | Concentrations of 0.781 and 7.81 μg/mL of silver tungstate microcrystals were not cytotoxic. | [62] | |
Human periodontal ligament fibroblasts | E-smoking liquids | The proliferation rates were reduced in a time-dependent manner; some flavors (e.g., menthol) exhibited higher cytotoxicity. | [63] | |
Luminometric assays | ||||
CellTiter-Glo | Human oral epithelial cells | Commercial resin bonding agents | Most uncured materials presented cytotoxic effects; most post-cured samples revealed no cytotoxicity. | [42] |
ATP assay | Human gingival fibroblasts | Novobiocin | Novobiocin decreased cell viability; ATP assay was the most sensitive assay of cell viability. | [56] |
Normal human oral keratinocytes | Electronic cigarette aerosols | The e-cig aerosols exhibited cytotoxic effects. | [64] | |
Human periodontal ligament fibroblasts | E-smoking liquids | Flavors like hazelnut, lime, or menthol reduced ATP detection. | [63] | |
Flowcytometric assays | ||||
Flowcytometry | Human gingival fibroblasts | Areca nut (Areca catechu L.) products | Areca nut products exhibited necrotizing effects. | [61] |
Human gingival epithelial cells | Cigarette smoke total particulate matter (TPM), smokeless tobacco extracted with complete artificial saliva (ST/CAS), whole-smoke conditioned media (WS-CM). | TPM caused necrosis, WS-CM did not increase the percentage of apoptotic or necrotic cells (the mechanism of cytotoxicity was different—cell cycle arrest and/or inhibition of proliferation), the low cytotoxicity of ST-CAS was confirmed. | [55] | |
Human oral keratinocytes | Smokeless tobacco extract | The percentage of cells in apoptotic death increased. | [65] | |
Human gingival epithelial cells | E-cigarette vapor | E-cigarette smoke promoted apoptosis and necrosis in gingival epithelial cells. | [58] | |
Human gingival stroma fibroblasts | Dental adhesives | The dental adhesives inhibited fibroblasts in the G0/G1 phase and impaired their transition to G1–S; the high cell death rate was not due to apoptosis, but possibly to necrosis. | [33] | |
Cell proliferation assays | ||||
Labeling with BrdU | Human oral fibroblasts | Antioxidant mixtures of resveratrol (R), ferulic acid (F), phloretin (P), and tetrahydrocurcuminoids (T)—RFT, PFR, and PFT | Antioxidant-mixture treatment induced DNA synthesis stimulation. | [43] |
Human oral fibroblasts | Cigarette smoke and electronic cigarette condensates | Fibroblast proliferation was significantly reduced after exposure to cigarette smoke and e-cig condensates. | [40] | |
Human gingival fibroblasts | Chitosan particles | Chitosan promoted cell proliferation. | [44] | |
Human gingival fibroblast | Cyclosporin A | Cyclosporin A significantly stimulated the proliferation of cells in a dose-dependent manner. | [66] | |
Human oral keratinocytes | BAPO and TPO—alternative photoinitiators to CQ in dental resinous materials | TPO and BAPO did not exhibit toxic effects on human oral keratinocytes. | [30] | |
Clonogenic assay | Oral keratinocyte cell line | Radiation | Radiation impaired cellular proliferation and migration. | [67] |
Human gingival fibroblasts | Platelet-rich plasma | Platelet-rich plasma (5%) stimulated the clonogenic ability of cells. | [68] | |
Radiolabeled thymidine (3H-TdR) | Human oral fibroblasts | Copper chloride solution | The addition of copper chloride had no significant effect on the fibroblast proliferation rates. | [69] |
Human gingival fibroblasts | Nicotine | Nicotine significantly inhibited proliferation. | [70] | |
Human oral fibroblasts | Ultraviolet B radiation | UVB-irradiated cells exhibited significantly reduced proliferation. | [38] | |
PCNA expression | Human gingival fibroblasts | Cyclosporin A | Cyclosporin A significantly increased PCNA levels. | [66] |
Human gingival fibroblasts | Chitosan particles | Chitosan increased PCNA protein levels. | [44] | |
Ki67 assay | Human gingival fibroblasts | Chitosan particles | Chitosan increased Ki67 staining. | [44] |
Human gingival fibroblasts | Er:YAG (erbium-doped yttrium–aluminum–garnet) laser irradiation | Laser irradiation increased Ki67 staining and the number of proliferating cells. | [48] | |
Real-time cell analyses | ||||
Human gingival fibroblasts | Chlorhexidine | The inhibition of human gingival fibroblast proliferation after exposure to chlorhexidine was time- and concentration-dependent. | [71] | |
Human gingival fibroblasts | Monomers/comonomers in dental resin composites (BisGMA, HEMA, TEGDMA, and UDMA) | HEMA exhibited the lowest cytotoxic effect. | [25] |
Tested Agent | Type of Cells | Results | Reference |
---|---|---|---|
Dental materials | |||
Cold-curing acrylic resin | Human periodontal ligament fibroblasts | The cells became rounder because of cytoplasmic shrinkage, and vacuolization was stimulated. | [108] |
Dental adhesive | Human gingival fibroblasts | The fusiform-shaped cells eventually became detachable and round. | [33] |
Materials for prosthetic components (polyether-ether-ketone and stainless steel) | Human gingival keratinocytes and fibroblasts | The morphology of both cell lines was not affected. | [109] |
Titanium discs | Human gingival fibroblasts | The surface roughness of the titanium discs could influence cell morphology. | [110] |
Discs with Laser-Lok (a laser-modified titanium surface), zirconia, and titanium surfaces | Human gingival fibroblasts | Cell morphology was different for the three materials; in the Laser-Lok group, the cells were elongated and had pseudopods, while in the zirconia group, they were round. | [111] |
Silver tungstate (α-Ag2WO4) microcrystals | Human gingival fibroblasts | A normal morphology was observed after exposure to α-Ag2WO4 (7.81 μg/mL); a concentration of 78.1 μg/mL caused membrane disruption and complete cell death. | [62] |
Citric acid (root surface demineralization, smear layer removal, dentin etching) | Human dental pulp cells | Cell retraction and cell surface blebbing were observed. | [112] |
Endodontic sealers | Human gingival fibroblasts | Compromised cell membranes and loss of cell content were observed. | [113] |
Hydrogen peroxide bleaching gels | Human dental pulp cells | Alterations in cell morphology appeared (e.g., shrinkage of the cytoplasm), proportional to the peroxide concentration. | [37] |
Topical fluoride varnishes | Human gingival fibroblasts | Changes in cell morphology and in the actin cytoskeleton structure were observed. | [83] |
Methacrylate-based CAD/CAM milled and 3D-printed samples | Human gingival fibroblasts | Apoptotic features (cell shrinking, membrane blebbing, and apoptotic body formation) were identified in fibroblasts incubated with methacrylate-based 3D samples. | [114] |
Drugs/oral healthcare formulations | |||
ZnO, TiO2, SiO2, hydroxyapatite—bulk particle-sized materials and nanomaterials | Oral epithelial keratinocytes | Cells exposed to ZnO (bulk and nanomaterials) presented with reduced/absent filament network on cell surfaces, reduced diameter, and blebbing. | [32] |
Chlorhexidine | Human gingival fibroblasts | Low concentrations of chlorhexidine did not influence cell morphology; high concentrations caused the appearance of small, round-shaped cells. | [71] |
Valproic acid | Human gingival fibroblasts | High concentrations of valproic acid (8 mM) affected cell shape and proliferation. | [115] |
Tacrolimus | Human gingival fibroblasts | Only at 100 µg/mL, the cell morphology was affected. | [116] |
Zoledronic acid | Human gingival fibroblasts | Altered cell morphology and the presence of round-shaped cells with disrupted cytoplasmic membrane and numerous flake structures were observed. | [28] |
Other agents | |||
Cigarette smoke and e-vapor condensates | Human gingival fibroblasts | The e-vapor condensates (nicotine-rich and nicotine-free) and cigarette smoke caused alterations in cell shape. | [40] |
Traditional cigarette smoke and e-cigarette aerosol | Human epithelial gingival keratinocytes | Apoptotic features were detected after exposure (apoptotic cellular bodies, nuclear condensation, and DNA fragmentation). | [39] |
E-cigarette vapors | Human gingival epithelial cells | The morphology of cells was altered; the small cuboidal cells changed into large, elongated shapes. | [58] |
Metallic ions (beryllium (Be+2), chromium (Cr+6 and Cr+3), nickel (Ni+2), and molybdenum (Mo+6)) | Human gingival fibroblasts | Hexavalent chromium and nickel induced irregularly shaped nuclei; all metal ions reduced the number of polyribosomes and the size of mitochondria; and beryllium and molybdenum caused pseudopodia. | [117] |
Zinc oxide nanoparticles | Human gingival fibroblasts | Only at concentrations above 50 μg/mL did ZnO nanoparticles have altered cell morphology. | [57] |
Chitosan-coated and alginate-coated liposomes | Human oral keratinocytes | Exposed cells did not exhibit an altered morphology at visual inspection. | [27] |
Asiasari radix extracts | Human stem cells derived from the gingiva | Alterations in the cytoskeletal organization were observed at high concentrations of extracts (100 and 1000 μg/mL). | [118] |
Areca nut products | Human gingival fibroblasts | Reduced cytoplasmic volume and shrinkage of the actin cytoskeleton were observed. | [61] |
Er:YAG laser irradiation | Human gingival fibroblasts | Transient alterations of the mitochondria and ribosomal endoplasmic reticulum were observed. | [48] |
Investigated Parameter | Type of Cells | Tested Agent | Results | Reference |
---|---|---|---|---|
Dental materials | ||||
Intracellular ROS/RNS levels mRNA expression of redox-regulated proteins | Human oral keratinocytes | Photoinitiators BAPO and TPO | No increase in the intracellular ROS/RNS was observed; BAPO modified the expression of oxidatively regulated enzymes. | [30] |
ROS generation Expression of antioxidant enzymes | Human pulp- derived cells | Photoinitiators (CQ and TPO) | Adaptive changes in the expression of antioxidant enzymes were present. | [145] |
Intracellular ROS levels | Human oral keratinocyte | Silver–palladium–gold–indium dental alloy | Increasing indium concentration was associated with increased intracellular ROS production. | [47] |
Intracellular ROS levels | Human gingival fibroblast | Silver tungstate microcrystals | Only the highest tested concentration (78.1 μg/mL) caused an overproduction of ROS. | [62] |
Intracellular ROS levels | Human pulp cells | Dental composites | The components released from composites caused cytotoxicity via ROS formation. | [149] |
Intracellular GSH levels Lipid peroxidation | Human gingival fibroblasts | TEGDMA | The depletion of intracellular GSH was drastic after TEGDMA exposure. Signs of lipid peroxidation were recorded. | [90] |
Intracellular GSH levels | Human oral keratinocytes | TEGDMA | Moderate ROS formation was recorded. | [135] |
Nicotine and ENDS products | ||||
Intracellular ROS levels | Cementoblasts | Nicotine | The levels of intracellular ROS increased after nicotine exposure in a time-dependent manner. | [150] |
Intracellular GSH levels Expression of redox-regulated proteins | Normal human oral keratinocytes | Electronic cigarette aerosol | A significant decrease in intracellular GSH levels was recorded. The aerosols induced the expression of heme oxygenase 1. | [64] |
Intracellular ROS levels | Human gingival fibroblasts | E-cigarette fluids | The production of ROS was increased by both nicotine-containing and nicotine-free fluids. | [151] |
ROS generation | Human oral fibroblasts | Cigarette smoke and e-vapor condensates | After traditional cigarette smoke exposure, the formation of ROS was significantly higher than for e-vapor condensates. | [81] |
ROS generation Lipid peroxidation | Oral epithelial cells | E-cigarette aerosols | Most of the tested e-liquids induced significant levels of oxidative stress, while lipid peroxidation was less common. | [54] |
Immunodetection of protein carbonylation | Human periodontal ligament fibroblasts | E-cigarette vapors with flavorings | E-cig aerosols and flavorings caused carbonyl stress. | [138] |
Other agents | ||||
Intracellular GSH levels Generation of H2O2 in the cell culture medium | Human gingival fibroblasts | Theaflavin mixture purified from black tea | A pro-oxidant activity of the theaflavin mixture was observed, with a reduction in GSH levels (but this action was more intense for malignant cells). | [53] |
Intracellular ROS levels | Human gingival fibroblast | ZnO nanoparticles | High concentrations significantly increased ROS levels, while low concentrations had no influence. | [57] |
Intracellular ROS levels | Human gingival fibroblasts | Curcumin | Curcumin determined the dose-dependent production of ROS up to 15 μM, where it reached a plateau. | [85] |
Intracellular ROS levels | Normal human oral fibroblasts | Areca nut | Cytokine secretion increased and triggered the generation of ROS. | [152] |
Investigated Parameter | Type of Cells | Tested Agent | Results | Reference |
---|---|---|---|---|
Dental materials | ||||
Gene expressions of IL1β, IL6, IL8, IL10, TNFα, and VEGF | Human gingival stroma fibroblasts | Dental adhesives | The adhesives upregulated the gene expression of IL1β, IL6, IL8, and VEGF, but did not affect the IL-10 and TNFα expression. | [33] |
1L-6 and 1L-8 levels | Human gingival fibroblasts | Denture base acrylic resins | An increased secretion of cytokines was observed. | [158] |
IL-1b, IL-6, IL-10, and TNF-α levels | Human dental pulp cells | Dental adhesives (the influence of HEMA and solvent concentrations) | HEMA 20% + ethanol significantly increased cytokine release (IL-6, IL-10, and TNF-α) after a 24 h exposure. | [159] |
IL-6 and PGE2 levels | Human gingival fibroblasts and human oral mucosal keratinocytes | Three-dimensional-printed oral appliances | The PGE2 concentration in the gingival fibroblast cultures was reduced after exposure to some printable resins. | [155] |
Nicotine, ENDS and tobacco products | ||||
PGE2 and COX-2 levels IL 8 level | Human periodontal ligament fibroblasts | E-cigarette vapors with flavoring | E-cig aerosols and flavorings increased pro-inflammatory cytokine release and COX-2 levels. | [138] |
Gene expressions of IL-1α, IL-1β, and IL-6 | Human epithelial oral cells | E-cigarette liquid aerosols and traditional cigarette smoke | E-cig aerosol and traditional cigarette smoke increased the expression of interleukins (higher for traditional cigarette smoke). | [39] |
Gene expressions of IL6, IL8, and TNFα | Human gingival fibroblasts | Flavored nicotine pouches and CRP1.1 (a smokeless tobacco reference product—snus) | Only the expression of the gene IL6 was upregulated by CRP1.1. | [160] |
Other agents | ||||
Production of IL-1β, IL-6, IL-8, IL-15, and TNF-α | Human gingival fibroblasts | Ethanolic extract of propolis and CAPE | CAPE inhibited the production of TNF-α and IL-6, caused by the LPS and IFN-α stimulation. The propolis extract did not significantly influence the concentrations of IL-1β and TNF-α. | [161] |
PGE2 levels Gene expression of COX-2, IL-6, MMP-2, MMP-8, MIP-1α, and SDF-1 | Human gingival fibroblasts | Ecklonia cava extract | The extract decreased the expression of pro-inflammatory enzymes and chemokines and reduced PGE2 production. | [162] |
PGE2, NO, IL-6, and IL-8 levels | Human gingival fibroblasts | LPS from Porphyromonas gingivalis and asiatic acid | Asiatic acid reduced LPS-induced production of PGE2, IL-6, and IL-8 in a concentration-dependent manner. | [163] |
Production of IL-1β and IL-6 | Human gingival fibroblasts | Bamboo salt | Bamboo salt exhibited anti-inflammatory activities and inhibited the release of IL-1β and IL-6. | [164] |
Production of IL-6 and IL-8 | Human gingival fibroblasts | Ozone ointment | The ozone ointment inhibited the production of IL-6 and IL-8, induced by LPS. | [165] |
Concentrations of TNF-α, IL-6, IL-8, MCP-1, and PGE2 | Human gingival fibroblasts | Synthetic azulene- related compounds | Benzo[b]cyclohepta[e][1,4] thiazine was the most potent inhibitor of IL-8 production. | [166] |
Type of Organ-on-a-Chip | Investigated Parameter | Tested Agent | Results | Reference |
---|---|---|---|---|
Tooth-on-a-chip | Cytotoxicity, morphology, proliferation, and metabolic activity | Dental materials (HEMA, phosphoric acid, Adper-Scotchbond) | All dental materials had cytotoxic effects and affected morphology and metabolic activity. | [207] |
Cell viability and morphology | Silver diamine fluoride | Silver diamine fluoride toxicity against dental pulp stem cells depended on the thickness of the dentin. | [203] | |
Pulp-dentin-on-a-chip | Cell viability, morphology, pH, and release of TGFβ | Calcium silicate cements | Of the three tested compounds, one compound affected cell proliferation and morphology, while the other two promoted proliferation and the release of TGFβ. | [211] |
Oral-mucosa-on-a-chip | Cell viability, morphology, and organization | HEMA | HEMA reduced cell viability and disrupted the mucosal layer organization. | [212] |
Cytotoxicity and morphology | HEMA | HEMA reduced mucosal cell viability. | [213] | |
Gingiva-on-a-chip | Cell viability and cytotoxicity | Oral-care formulations (mouthwashes) | The mucosal irritation potential was slightly higher for alcohol-based mouthwashes. | [204] |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2025 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Jităreanu, A.; Agoroaei, L.; Caba, I.-C.; Cojocaru, F.-D.; 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. https://doi.org/10.3390/toxics13030195
Jităreanu A, Agoroaei L, Caba I-C, Cojocaru F-D, 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(3):195. https://doi.org/10.3390/toxics13030195
Chicago/Turabian StyleJităreanu, Alexandra, Luminița Agoroaei, Ioana-Cezara Caba, Florina-Daniela Cojocaru, Liliana Vereștiuc, Mădălina Vieriu, and Ioana Mârțu. 2025. "The Evolution of In Vitro Toxicity Assessment Methods for Oral Cavity Tissues—From 2D Cell Cultures to Organ-on-a-Chip" Toxics 13, no. 3: 195. https://doi.org/10.3390/toxics13030195
APA StyleJităreanu, A., Agoroaei, L., Caba, I.-C., Cojocaru, F.-D., Vereștiuc, L., Vieriu, M., & Mârțu, I. (2025). The Evolution of In Vitro Toxicity Assessment Methods for Oral Cavity Tissues—From 2D Cell Cultures to Organ-on-a-Chip. Toxics, 13(3), 195. https://doi.org/10.3390/toxics13030195