RESUMEN
OBJECTIVE: To evaluate the efficacy and cytotoxicity of experimental 6% and 35% hydrogen peroxide gels (HP6 or HP35) incorporated with titanium dioxide nanoparticles (NP) co-doped with nitrogen and fluorine and irradiated with a violet LED light (LT). METHODS: Bovine enamel-dentin disks adapted to artificial pulp chambers were randomly assigned to bleaching (n = 8/group): NC (negative control), NP, HP6, HP6 + LT, HP6 + NP, HP6 + NP + LT, HP35, HP35 + LT, HP35 + NP, HP35 + NP + LT, and commercial HP35 (COM). Color (ΔE00) and whiteness index (ΔWID) changes were measured before and 14 days after bleaching. The extracts (culture medium + diffused gel components) collected after the first session were applied to odontoblast-like MDPC-23 cells, which were assessed concerning their viability, oxidative stress, and morphology. The amount of HP diffused through the disks was determined. Data were analyzed by generalized linear models or Kruskal Wallis Tests (α = 5%). RESULTS: HP6 + NP + LT exhibited ΔE00 and ΔWID higher than HP6 (p < 0.05) and similar to all HP35 groups. HP6 + NP + LT showed the lowest HP diffusion, and the highest cell viability (%) among bleached groups, preserving cell morphology and number of living cells similar to NC and NP. HP6 + LT, HP6 + NP, and HP6 + NP + LT exhibited the lowest cell oxidative stress among bleached groups (p < 0.05). HP35, HP35 + LT, and HP35 (COM) displayed the lowest cell viability. CONCLUSION: HP6 achieved significantly higher color and whiteness index changes when incorporated with nanoparticles and light-irradiated and caused lower cytotoxicity than HP35 gels. The nanoparticles significantly increased cell viability and reduced the hydrogen peroxide diffusion and oxidative stress, regardless of HP concentration. CLINICAL SIGNIFICANCE: Incorporation of co-doped titanium dioxide nanoparticles combined with violet irradiation within the HP6 gel could promote a higher perceivable and acceptable efficacy than HP6 alone, potentially reaching the optimal esthetic outcomes rendered by HP35. This approach also holds the promise of reducing cytotoxic damages and, consequently, tooth sensitivity.
Asunto(s)
Supervivencia Celular , Geles , Peróxido de Hidrógeno , Nanopartículas , Titanio , Blanqueadores Dentales , Blanqueamiento de Dientes , Peróxido de Hidrógeno/farmacología , Peróxido de Hidrógeno/toxicidad , Blanqueamiento de Dientes/métodos , Titanio/química , Titanio/toxicidad , Animales , Bovinos , Blanqueadores Dentales/toxicidad , Blanqueadores Dentales/farmacología , Supervivencia Celular/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Técnicas In Vitro , Odontoblastos/efectos de los fármacos , Esmalte Dental/efectos de los fármacos , Distribución Aleatoria , Dentina/efectos de los fármacosRESUMEN
PURPOSE: To determine and compare the cytotoxicity, odontoblast-like differentiation, shear bond strength (SBS) and Vickers microhardness of four commercial light-cured orthodontic adhesives. METHODS: The orthodontic resins selected were Transbond XT - GI, Transbond Plus Color Change - GII (both from 3M Unitek), Enlight - GIII and Blugloo - GIV (both from Ormco). Samples were prepared, and leached monomers were obtained. Cytotoxicity was evaluated using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and deposited calcium was analyzed using Alizarin red staining (ARS). SBS and the adhesive remnant index (ARI) were determined using 120 human premolars. The Vickers microhardness test was performed on the resin discs. RESULTS: All adhesives showed moderate to severe cytotoxicity (21-37%) and promoted similar formation of calcium deposits. A SBS of 6-8 MPa was achieved only by Blugloo (7.1 ± 2.4 MPa), and Enlight showed the lowest Vickers hardness score (40 ± 2.5 HV). Transbond Plus Color Change (score 0 = 42.9%) and Blugloo (score 0 = 46.4%) showed better ARI scores than Transbond XT (score 0 = 7.1%) and Enlight (score 0 = 3.6%). CONCLUSION: On the basis of the properties evaluated, Blugloo seems to be the best option.
Asunto(s)
Diferenciación Celular , Resinas Compuestas , Odontoblastos , Resistencia al Corte , Humanos , Odontoblastos/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Resinas Compuestas/toxicidad , Resinas Compuestas/química , Ensayo de Materiales , Cementos Dentales/toxicidad , Cementos Dentales/química , Dureza , Diente PremolarRESUMEN
OBJECTIVE: The molecular regulation of odontoblasts in dentin formation remains largely uncharacterized. Using neohesperidin (NEO), a well-documented osteoblast regulator, we investigated whether and how NEO participates in odontoblast regulation through longitudinal treatments using various doses of NEO. DESIGN: Mouse dental papilla cell-23 (MDPC-23) served as a model for odontoblasts. MDPC-23 were treated with various doses of NEO (0, 1, 5, 10, 15, 20 µmol/L). Proliferation was assessed using the Cell counting kit-8 assay. Survival/apoptosis was assayed by live/dead ratio. Migration capability was assessed using scratch healing and Transwell migration assays. Mineralization was assessed using alkaline phosphatase staining and alizarin red staining. The expression levels of four key genes (Runx2, osteocalcin [OCN], ß-catenin, and bone morphogenetic protein [BMP]-2) representing NEO-induced differentiation of MDPC-23 were measured by quantitative reverse transcription polymerase chain reaction. RESULTS: The proliferation trajectories of MDPC-23 treated with the five doses of NEO demonstrated similar curves, with a rapid increase in the 10 µmol/L NEO condition after 48 h of treatment. Similar dose-dependent trajectories were observed for survival/apoptosis. All four key genes representing odontogenic differentiation were upregulated in MDPC-23 induced by NEO treatments at two optimal doses (5 µmol/L and 10 µmol/L). Optimal migration and mobility trajectories were observed in MDPC-23 treated with 10 µmol/L NEO. Optimal mineralization was observed in MDPC-23 treated with 5 µmol/L NEO. CONCLUSION: NEO can subtly regulate odontoblast proliferation, differentiation, migration, and mineralization in vitro. NEO at 5-10 µmol/L offers a safe and effective perspective for clinical promotion of dentin bridge formation in teenagers.
Asunto(s)
Apoptosis , Diferenciación Celular , Movimiento Celular , Proliferación Celular , Papila Dental , Hesperidina , Odontoblastos , Animales , Ratones , Proliferación Celular/efectos de los fármacos , Hesperidina/farmacología , Hesperidina/análogos & derivados , Diferenciación Celular/efectos de los fármacos , Odontoblastos/efectos de los fármacos , Papila Dental/citología , Papila Dental/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Apoptosis/efectos de los fármacos , Osteocalcina/metabolismo , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Técnicas In Vitro , Proteína Morfogenética Ósea 2/farmacología , Supervivencia Celular/efectos de los fármacos , beta Catenina/metabolismo , Fosfatasa Alcalina/metabolismo , Células Cultivadas , Reacción en Cadena en Tiempo Real de la PolimerasaRESUMEN
Epigenetic change has been found to play an important role in cell differentiation and regulation and the dental pulp stem cell in tissue engineering is gaining attention due to the ability of cells to differentiate into odontoblast and other cells. This study evaluated the influence of poly L- lactic acid with hydroxyapatite-coated with polyaniline scaffold (PLLA/HA/PANI) on dental pulp stem cell (DPSC) proliferation and differentiation. After scaffold preparation and DPSCs seeding, the cells proliferation and differentiation were evaluated by immunocytochemistry assay and cell viability was measured by cytotoxicity / MTT assay. The results showed (PLLA/HA/PANI) scaffold facilitates DPSC proliferation and differentiation with gene expression. This finding underscores the promise of this biomaterial combination as a scaffold for dental tissue regeneration and application.
Asunto(s)
Materiales Biocompatibles , Diferenciación Celular , Proliferación Celular , Pulpa Dental , Durapatita , Odontoblastos , Osteoblastos , Células Madre , Andamios del Tejido , Pulpa Dental/citología , Humanos , Diferenciación Celular/efectos de los fármacos , Odontoblastos/citología , Odontoblastos/efectos de los fármacos , Odontoblastos/metabolismo , Andamios del Tejido/química , Células Madre/citología , Células Madre/metabolismo , Células Madre/efectos de los fármacos , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Proliferación Celular/efectos de los fármacos , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología , Durapatita/química , Durapatita/farmacología , Compuestos de Anilina/farmacología , Compuestos de Anilina/química , Poliésteres/química , Poliésteres/farmacología , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Ingeniería de Tejidos/métodosRESUMEN
OBJECTIVES: To investigate the transdentinal effects of surface reaction-type pre-reacted glass-ionomer (S-PRG) fillers on odontoblast-like cells. METHODS: An eluate of S-PRG fillers was obtained by dissolving the particles in distilled water (1:1 m/v). Dentin discs with similar permeability were mounted into artificial pulp chambers and MDPC-23 cells were seeded on their pulpal surface. The occlusal surface was treated with (n = 10): ultrapure water (negative control - NC), hydrogen peroxide (positive control - PC), S-PRG eluate exposure for 1 min (S-PRG 1 min), or S-PRG filler eluate exposure for 30 min (S-PRG 30 min). After 24 h, cell viability (alamarBlue) and morphology (SEM) were evaluated. The extract obtained from transdentinal diffusion was applied to MDPC-23 pre-cultured in plates for another 24 h to evaluate viability (alamarBlue, 1, 3, and 7 days), gene expression of Col1a1, Alpl, Dspp, and Dmp1 (RT-qPCR, 1 and 7 days), and mineralization (Alizarin Red, 7 days). Data were analyzed with ANOVA (α = 5 %). RESULTS: While S-PRG 1 min did not differ from NC, S-PRG 30 min reduced 17.9 % viability of cells from discs. S-PRG treatments resulted in low cell detaching from dentin, and the remaining cells exhibited typical morphology or minor cytoplasmic contraction. S-PRG 30 min slightly increased cell viability (6 %) 1 day after contact with the extract. S-PRG treatments upregulated the expression of the investigated genes, especially after 1 day. S-PRG 30 min stimulated mineralization activity by 39.7 %. CONCLUSIONS: S-PRG filler eluate does not cause transdentinal cytotoxicity on odontoblast-like cells, and long-term exposure can stimulate their dentinogenic-related mineralization activity. SIGNIFICANCE: The transdentinal elution of ions from S-PRG fillers is not expected to be harmful to the dental pulp and may exert bioactive effects by inducing dentin matrix deposition through the metabolism of underlying odontoblasts.
Asunto(s)
Supervivencia Celular , Dentina , Odontoblastos , Odontoblastos/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Dentina/efectos de los fármacos , Cementos de Ionómero Vítreo/farmacología , Cementos de Ionómero Vítreo/química , Cementos de Ionómero Vítreo/toxicidad , Animales , Microscopía Electrónica de Rastreo , Ensayo de Materiales , Propiedades de Superficie , Ratones , Células Cultivadas , Expresión Génica/efectos de los fármacos , Resinas Acrílicas , Dióxido de SilicioRESUMEN
OBJECTIVES: This study aimed to investigate the regulatory mechanisms governing dental mesenchymal cell commitment during tooth development, focusing on odontoblast differentiation and the role of epigenetic regulation in this process. METHODS: We performed single-cell RNA sequencing (scRNA-seq) of dental cells from embryonic day 14.5 (E14.5) mice to understand the heterogeneity of developing tooth germ cells. Computational analyses including gene regulatory network (GRN) assessment were conducted. We validated our findings using immunohistochemistry (IHC) and in vitro loss-of-function analyses using the DNA methyltransferase 1 (DNMT1) inhibitor Gsk-3484862 in primary dental mesenchymal cells (DMCs) isolated from E14.5 mouse tooth germs. Bulk RNA-seq of Gsk-3484862-treated DMCs was performed to identify potential downstream targets of DNMT1. RESULTS: scRNA-seq analysis revealed diverse cell populations within the tooth germs, including epithelial, mesenchymal, immune, and muscle cells. Using single-cell regulatory network inference and clustering (SCENIC), we identified Dnmt1 as a key regulator of early odontoblast development. IHC analysis showed the ubiquitous expression of DNMT1 in the dental papilla and epithelium. Bulk RNA-seq of cultured DMCs showed that Gsk-3484862 treatment upregulated odontoblast-related genes, whereas genes associated with cell division and the cell cycle were downregulated. Integrated analysis of bulk RNA-seq data with scRNA-seq SCENIC profiles was used to identify the potential Dnmt1 target genes. CONCLUSIONS: Dnmt1 may negatively affect odontoblast commitment and differentiation during tooth development. These findings contribute to a better understanding of the molecular mechanisms underlying tooth development and future development of hard-tissue regenerative therapies.
Asunto(s)
Diferenciación Celular , ADN (Citosina-5-)-Metiltransferasa 1 , Papila Dental , Odontoblastos , Análisis de la Célula Individual , Germen Dentario , Animales , Ratones , ADN (Citosina-5-)-Metiltransferasa 1/genética , ADN (Citosina-5-)-Metiltransferasa 1/metabolismo , Germen Dentario/metabolismo , Germen Dentario/citología , Germen Dentario/embriología , Papila Dental/citología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Odontoblastos/citología , Odontoblastos/metabolismo , Odontoblastos/efectos de los fármacos , Análisis de Secuencia de ARN/métodos , Odontogénesis/genética , Odontogénesis/efectos de los fármacos , Transcriptoma , Inmunohistoquímica , Redes Reguladoras de Genes/efectos de los fármacosRESUMEN
This study aimed to evaluate the physiological role of NAMPT associated with MDPC-23 odontoblast cell proliferation. Cell viability was measured using the (DAPI) staining, caspase activation analysis and immunoblotting were performed. Visfatin promoted MDPC-23 odontoblast cell growth in a dose-dependent manner. Furthermore, the up-regulation of Visfatin promoted odontogenic differentiation and accelerated mineralization through an increase in representative odontoblastic biomarkers in MDPC-23 cells. However, FK-866 cell growth in a dose-dependent manner induced nuclear condensation and fragmentation. FK-866-treated cells showed H&E staining and increased apoptosis compared to control cells. The expression of anti-apoptotic factors components of the mitochondria-dependent intrinsic apoptotic pathway significantly decreased following FK-866 treatment. The expression of pro-apoptotic increased upon FK-866 treatment. In addition, FK-866 activated caspase-3 and PARP to induce cell death. In addition, after treating FK-866 for 72 h, the 3/7 activity of MDPC-23 cells increased in a concentration-dependent manner, and the IHC results also confirmed that Caspase-3 increased in a concentration-dependent. Therefore, the presence or absence of NAMPT expression in dentin cells was closely related to cell proliferation and formation of extracellular substrates.
Asunto(s)
Apoptosis , Proliferación Celular , Nicotinamida Fosforribosiltransferasa , Odontoblastos , Nicotinamida Fosforribosiltransferasa/metabolismo , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Odontoblastos/efectos de los fármacos , Odontoblastos/citología , Odontoblastos/metabolismo , Animales , Ratones , Línea Celular , Citocinas/metabolismo , Caspasa 3/metabolismo , Diferenciación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Acrilamidas/farmacología , Odontogénesis/efectos de los fármacosRESUMEN
AIMS: This study aimed to investigate the anti-inflammatory and odontoblastic effects of cerium-containing mesoporous bioactive glass nanoparticles (Ce-MBGNs) on dental pulp cells as novel pulp-capping agents. METHODOLOGY: Ce-MBGNs were synthesized using a post-impregnation strategy based on the antioxidant properties of Ce ions and proposed the first use of Ce-MBGNs for pulp-capping application. The biocompatibility of Ce-MBGNs was analysed using the CCK-8 assay and apoptosis detection. Additionally, the reactive oxygen species (ROS) scavenging ability of Ce-MBGNs was measured using the 2,7-Dichlorofuorescin Diacetate (DCFH-DA) probe. The anti-inflammatory effect of Ce-MBGNs on THP-1 cells was further investigated using flow cytometry and quantitative real-time polymerase chain reaction (RT-qPCR). Moreover, the effect of Ce-MBGNs on the odontoblastic differentiation of the dental pulp cells (DPCs) was assessed by combined scratch assays, RT-qPCR, western blotting, immunocytochemistry, Alizarin Red S staining and tissue-nonspecific alkaline phosphatase staining. Analytically, the secretions of tumour necrosis factor-α (TNF-α) and interleukin-1ß (IL-1ß) were detected with enzyme-linked immunosorbent assay (ELISA). RESULTS: Ce-MBGNs were confirmed to effectively scavenge ROS in THP-1-derived macrophages and DPCs. Flow cytometry and RT-qPCR assays revealed that Ce-MBGNs significantly inhibited the M1 polarization of macrophages (Mφ). Furthermore, the protein levels of TNF-α and IL-1ß were downregulated in THP-1-derived macrophages after stimulation with Ce-MBGNs. With a step-forward virtue of promoting the odontoblastic differentiation of DPCs, we further confirmed that Ce-MBGNs could regulate the formation of a conductive immune microenvironment with respect to tissue repair in DPCs, which was mediated by macrophages. CONCLUSIONS: Ce-MBGNs protected cells from self-produced oxidative damage and exhibited excellent immunomodulatory and odontoblastic differentiation effects on DPCs. As a pulp-capping agent, this novel biomaterial can exert anti-inflammatory effects and promote restorative dentine regeneration in clinical treatment. We believe that this study will stimulate further correlative research on the development of advanced pulp-capping agents.
Asunto(s)
Antiinflamatorios , Cerio , Pulpa Dental , Nanopartículas , Pulpa Dental/citología , Pulpa Dental/efectos de los fármacos , Cerio/farmacología , Humanos , Antiinflamatorios/farmacología , Especies Reactivas de Oxígeno/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Cerámica/farmacología , Diferenciación Celular/efectos de los fármacos , Vidrio , Odontoblastos/efectos de los fármacos , Regeneración/efectos de los fármacos , Células THP-1 , Materiales de Recubrimiento Pulpar y Pulpectomía/farmacología , Interleucina-1beta/metabolismo , Apoptosis/efectos de los fármacos , Porosidad , Células CultivadasRESUMEN
It remains an obstacle to induce the regeneration of hard dentin tissue in clinical settings. To overcome this, a P(VDF-TrFE) piezoelectric film with 2 wt% SrCl2 addition is designed. The biofilm shows a high flexibility, a harmonious biocompatibility, and a large piezoelectric d33 coefficient of 14 pC N-1, all contributing to building an electric microenvironment that favor the recruitment of dental pulp stem cells (DPSCs) and their differentiation into odontoblasts during normal chewing, speaking, etc. On the other hand, the strontium ions can be gradually released from the film, thus promoting DPSC odonto-differentiation. In vivo experiments also demonstrate that the film induces the release of dentin minerals and regeneration of dentin tissue. In the large animal dentin defect models, this piezoelectric film induces in situ dentin tissue formation effectively over a period of three months. This study illustrates a therapeutic potential of the piezoelectric film to improve dentin tissue repair in clinical settings.
Asunto(s)
Biopelículas , Pulpa Dental , Dentina , Regeneración , Células Madre , Estroncio , Dentina/química , Biopelículas/efectos de los fármacos , Pulpa Dental/citología , Estroncio/química , Estroncio/farmacología , Animales , Humanos , Regeneración/efectos de los fármacos , Células Madre/citología , Células Madre/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Odontoblastos/citología , Odontoblastos/efectos de los fármacos , Odontoblastos/metabolismo , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacologíaRESUMEN
This study was designed to investigate the effects of Sake Lees extracts (SLE, Sake Kasu) on the functional activity of odontoblastic cells and tooth pulp of the rats. For in vitro studies, a rat clonal odontoblast-like cell line, KN-3 cells were cultured. SLE significantly decreased KN-3 cell proliferation, but showed no significant cytotoxicity. SLE effects on several protein productions of KN-3 cells were compared with PBS. SLE and PBS increased alkaline phosphatase (ALP), dentin sialoprotein (DSP), and osterix in a day-course dependent manner, while SLE increased the induction of ALP on day 9-21 and DSP on day 15-21. SLE also increased Runx2 expression on day 3 and 9 compared to PBS. Alizarin Red stainings revealed that SLE showed a subtle increase in mineralization of KN-3 cells on day 15 and 21. A histological investigation was conducted to assess if SLE induced reparative dentin formation after direct capping at the exposed tooth pulp in rats, suggesting that SLE could increase the reparative dentin formation more than PBS. These findings suggest that Sake Lees could have functional roles in the alterations of odontoblastic activity, which might influence the physiology of the tooth pulp.
Asunto(s)
Odontoblastos , Oryza , Extractos Vegetales , Animales , Diferenciación Celular , Línea Celular , Pulpa Dental , Proteínas de la Matriz Extracelular/metabolismo , Odontoblastos/efectos de los fármacos , Odontoblastos/metabolismo , Oryza/química , Extractos Vegetales/farmacología , RatasRESUMEN
This study aimed to evaluate the in vitro effect of the novel bioactive adhesive monomer CMET, a calcium salt of 4-methacryloxyethyl trimellitate acid (4-MET), on human dental pulp stem cells (hDPSCs) and its capacity to induce tertiary dentin formation in a rat pulp injury model. Aqueous solutions of four tested materials [4-MET, CMET, Ca(OH)2, and mineral trioxide aggregate (MTA)] were added to the culture medium upon confluence, and solvent (dH2O) was used as a control. Cell proliferation was assessed using the Cell Counting Kit-8 assay, and cell differentiation was evaluated by real-time quantitative reverse transcription-polymerase chain reaction. The mineralization-inducing capacity was evaluated using alizarin red S staining and an alkaline phosphatase activity assay. For an in vivo experiment, a mechanical pulp exposure model was prepared on Wistar rats; damaged pulp was capped with Ca(OH)2 or CMET. Cavities were sealed with composite resin, and specimens were assessed after 14 and 28 days. The in vitro results showed that CMET exhibited the lowest cytotoxicity and highest odontogenic differentiation capacity among all tested materials. The favorable outcome on cell mineralization after treatment with CMET involved p38 and c-Jun N-terminal kinases signaling. The nuclear factor kappa B pathway was involved in the CMET-induced mRNA expression of odontogenic markers. Similar to Ca(OH)2, CMET produced a continuous hard tissue bridge at the pulp exposure site, but treatment with only CMET produced a regular dentinal tubule pattern. The findings suggest that (1) the evaluated novel bioactive adhesive monomer provides favorable biocompatibility and odontogenic induction capacity and that (2) CMET might be a very promising adjunctive for pulp-capping materials.
Asunto(s)
Pulpa Dental/citología , Dentina/citología , Metacrilatos/farmacología , Odontoblastos/citología , Odontogénesis , Regeneración , Células Madre/citología , Ácidos Tricarboxílicos/farmacología , Adhesivos , Animales , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Pulpa Dental/efectos de los fármacos , Pulpa Dental/metabolismo , Dentina/efectos de los fármacos , Dentina/metabolismo , Masculino , Odontoblastos/efectos de los fármacos , Odontoblastos/metabolismo , Ratas , Ratas Wistar , Transducción de Señal , Células Madre/efectos de los fármacos , Células Madre/metabolismoRESUMEN
The role of inflammatory mediators in dental pulp is unique. The local environment of pulp responds to any changes in the physiology that are highly fundamental, like odontoblast cell differentiation and other secretory activity. The aim of this review is to assess the role of cathelicidins based on their capacity to heal wounds, their immunomodulatory potential, and their ability to stimulate cytokine production and stimulate immune-inflammatory response in pulp and periapex. Accessible electronic databases were searched to find studies reporting the role of cathelicidins in pulpal inflammation and regeneration published between September 2010 and September 2020. The search was performed using the following databases: Medline, Scopus, Web of Science, SciELO and PubMed. The electronic search was performed using the combination of keywords "cathelicidins" and "dental pulp inflammation". On the basis of previous studies, it can be inferred that LL-37 plays an important role in odontoblastic cell differentiation and stimulation of antimicrobial peptides. Furthermore, based on these outcomes, it can be concluded that LL-37 plays an important role in reparative dentin formation and provides signaling for defense by activating the innate immune system.
Asunto(s)
Catelicidinas/uso terapéutico , Pulpa Dental/efectos de los fármacos , Inflamación/tratamiento farmacológico , Odontoblastos/citología , Cicatrización de Heridas/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Pulpa Dental/citología , Pulpa Dental/inmunología , Pulpa Dental/metabolismo , Humanos , Inmunomodulación , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Odontoblastos/efectos de los fármacos , Odontoblastos/inmunología , Odontoblastos/metabolismoRESUMEN
Dental papilla cells (DPCs), precursors of odontoblasts, are considered promising seed cells for tissue engineering. Emerging evidence suggests that melatonin promotes odontoblastic differentiation of DPCs and affects tooth development, although the precise mechanisms remain unknown. Retinoid acid receptor-related orphan receptor α (RORα) is a nuclear receptor for melatonin that plays a critical role in cell differentiation and embryonic development. This study aimed to explore the role of RORα in odontoblastic differentiation and determine whether melatonin exerts its pro-odontogenic effect via RORα. Herein, we observed that RORα was expressed in DPCs and was significantly increased during odontoblastic differentiation in vitro and in vivo. The overexpression of RORα upregulated the expression of odontogenic markers, alkaline phosphatase (ALP) activity and mineralized nodules formation (p < 0.05). In contrast, odontoblastic differentiation of DPCs was suppressed by RORα knockdown. Moreover, we found that melatonin elevated the expression of odontogenic markers, which was accompanied by the upregulation of RORα (p < 0.001). Utilising small interfering RNA, we further demonstrated that RORα inhibition attenuated melatonin-induced odontogenic gene expression, ALP activity and matrix mineralisation (p < 0.01). Collectively, these results provide the first evidence that RORα can promote odontoblastic differentiation of DPCs and mediate the pro-odontogenic effect of melatonin.
Asunto(s)
Diferenciación Celular , Papila Dental/citología , Melatonina/farmacología , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Odontoblastos/citología , Odontoblastos/metabolismo , Odontogénesis , Animales , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Odontoblastos/efectos de los fármacos , Odontogénesis/efectos de los fármacos , Ratas Sprague-Dawley , Regulación hacia Arriba/efectos de los fármacosRESUMEN
Incomplete polymerization or biodegradation of dental resin materials results in the release of resin monomers such as triethylene glycol dimethacrylate (TEGDMA), causing severe injury of dental pulp cells. To date, there has been no efficient treatment option for this complication, in part due to the lack of understanding of the mechanism underlying these phenomena. Here, for the first time, we found that notoginsenoside R1 (NR1), a bioactive ingredient extracted from Panax notoginseng, exerted an obvious protective effect on TEGDMA-induced mitochondrial apoptosis in the preodontoblast mDPC6T cell line. In terms of the mechanism of action, NR1 enhanced the level of phosphorylated Akt (protein kinase B), resulting in the activation of a transcriptional factor, nuclear factor erythroid 2-related factor 2 (Nrf2), and eventually upregulating cellular ability to resist TEGDMA-related toxicity. Inhibiting the Akt/Nrf2 pathway by pharmaceutical inhibitors significantly decreased NR1-mediated cellular antioxidant properties and aggravated mitochondrial oxidative damage in TEGDMA-treated cells. Interestingly, NR1 also promoted mitophagy, which was identified as the potential downstream of the Akt/Nrf2 pathway. Blocking the Akt/Nrf2 pathway inhibited mitophagy and abolished the protection of NR1 on cells exposed to TEGDMA. In conclusion, these findings reveal that the activation of Akt/Nrf2 pathway-mediated mitophagy by NR1 might be a promising approach for preventing resin monomer-induced dental pulp injury.
Asunto(s)
Antioxidantes/farmacología , Apoptosis/efectos de los fármacos , Ginsenósidos/farmacología , Mitocondrias/efectos de los fármacos , Mitofagia/efectos de los fármacos , Factor 2 Relacionado con NF-E2/metabolismo , Odontoblastos/efectos de los fármacos , Polietilenglicoles/toxicidad , Ácidos Polimetacrílicos/toxicidad , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Línea Celular , Activación Enzimática , Ratones , Mitocondrias/enzimología , Mitocondrias/patología , Odontoblastos/enzimología , Odontoblastos/patología , Estrés Oxidativo/efectos de los fármacos , Fosforilación , Transducción de SeñalRESUMEN
AIM: To identify odontogenesis-promoting compounds and examine the molecular mechanism underlying enhanced odontoblast differentiation and tooth formation. METHODOLOGY: Five different nymphaeols, nymphaeol B (NB), isonymphaeol B (INB), nymphaeol A (NA), 3'-geranyl-naringenin (GN) and nymphaeol C (NC) were isolated from the fruit of Macaranga tanarius. The cytotoxic effect of nymphaeols on human DPSCs was observed using a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. The effect of nymphaeols on odontoblast differentiation was analysed with Alizarin Red S staining and odontoblast marker expression was assessed using real-time polymerase chain reaction and Western blot analysis. The molecular mechanism was investigated with Western blot analysis. In order to examine the effect of INB on dentine formation in the developing tooth germ, INB-soaked beads were placed under the tooth bud explants in the collagen gel; thereafter, the tooth bud explant-bead complexes were implanted into the sub-renal capsules for 3 weeks. Tooth root formation was analysed using micro-computed tomography and histological analysis. Data are presented as mean ± standard error (SEM) values of three independent experiments, and results are compared using a two-tailed Student's t-test. The data were considered to have statistical significance when the P-value was less than 0.05. RESULTS: Three of the compounds, NB, INB, and GN, did not exert a cytotoxic effect on human DPSCs. However, INB was most effective in promoting the deposition of calcium minerals in vitro (P < 0.001) and induced the expression of odontogenic marker genes (P < 0.05). Moreover, this compound strongly induced the phosphorylation of mitogen-activated protein (MAP) kinases and protein kinase B (AKT) (P < 0.05). The inhibition of p38 MAP, c-Jun N-terminal kinase (JNK), and AKT substantially suppressed the INB-induced odontoblast differentiation (P < 0.001). In addition, isonymphaeol B significantly induced the formation of dentine and elongation of the tooth root in vivo (P < 0.05). CONCLUSIONS: Prenylflavonoids, including INB, exerted stimulatory effects on odontoblast differentiation and tooth root and dentine formation via the MAP kinase and AKT signalling pathways. These results suggest that nymphaeols could stimulate the repair processes for dentine defects or injuries.
Asunto(s)
Diferenciación Celular/efectos de los fármacos , Euphorbiaceae/química , Flavonoides/farmacología , Odontoblastos/efectos de los fármacos , Células Madre/efectos de los fármacos , Células Cultivadas , Pulpa Dental/citología , Humanos , Proteínas Quinasas Activadas por Mitógenos , Proteínas Proto-Oncogénicas c-akt , Transducción de Señal , Raíz del Diente , Microtomografía por Rayos XRESUMEN
Functional peptides derived from the active domains of odontogenesis-related proteins have been reported to promote dental hard tissue regeneration. The purpose of this study was to evaluate the effects of an artificially synthesized peptide, TVH-19, on odontoblast differentiation and tertiary dentin formation in indirect pulp capping (IPC) using in vitro and in vivo experiments. TVH-19 did not exhibit any effect on the proliferation of human dental pulp cells (hDPCs) but significantly promoted cell migration, compared with the control (p < 0.05). TVH-19-treated hDPCs showed significantly higher alkaline phosphatase (ALP) activity and stronger alizarin red staining (ARS) reactivity than the control group (p < 0.05). TVH-19 also upregulated the mRNA and protein expression levels of odontogenic genes. After generating IPC in rats, the samples of teeth were studied using micro-computed tomography (Micro-CT), hematoxylin & eosin (HE) staining, and immunohistochemical staining to investigate the functions of TVH-19. The in vivo results showed that TVH-19 induced the formation of tertiary dentin, and reduced inflammation and apoptosis, as evident from the downregulated expression of interleukin 6 (IL-6) and cleaved-Caspase-3 (CL-CASP3). Overall, the results of our study suggest that TVH-19 induces differentiation of hDPCs, promotes tertiary dentin formation, relieves inflammation, and reduces apoptosis, indicating the potential applications of TVH-19 in IPC.
Asunto(s)
Pulpa Dental/citología , Pulpa Dental/efectos de los fármacos , Dentina/metabolismo , Péptidos/farmacología , Diferenciación Celular/efectos de los fármacos , Línea Celular , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Pulpa Dental/metabolismo , Humanos , Odontoblastos/citología , Odontoblastos/efectos de los fármacos , Odontoblastos/metabolismo , Péptidos/química , Calcificación de Dientes/efectos de los fármacosRESUMEN
Resin-based dental materials consist of filler particles and different monomers that are light cured in situ to re-establish dental function and aesthetics. Due to the degree of conversion of adhesive polymers, the monomers triethyleneglycol dimethacrylate (TEGDMA) and 2-hydroxyethyl methacrylate (HEMA) are released in relatively high amounts and are susceptible to degradation, acting as bioactive compounds and affecting cell and tissues. This study aimed to assess the effect of HEMA and TEGDMA exposure on metabolic activity, membrane integrity, and cell survival of human odontoblast-like cell (hOLCs). Exposure to resin monomers for 24 h induced major changes in cell membrane integrity, metabolic activity, and survival, which were measured by the calcein method and lactate dehydrogenase release. Increased and early reactive oxygen species (ROS) production was observed leading to degradative oxidation of membrane lipids identified as malondialdehyde production. Severe alteration in mitochondria occurred due to transmembrane mitochondrial potential collapse, possibly inducing activation of apoptotic cell death. hOLCs exposure to resin monomers modified the cell redox potential, with consequences on membrane permeability and integrity, including mitochondrial function. Lipid peroxidation appears to be a key phenomenon for the membrane structures oxidation after HEMA and TEGDMA exposure, leading to cell death and cytotoxicity. hOLCs respond early by differential induction of adaptive mechanisms to maintain cell homeostasis. Modulation of oxidative stress-induced response involves the regulation of genes that encode for antioxidant proteins such as catalase and heme oxygenase-1; regulation that functions as a critical protection mechanism against oxidative cell damage induced by HEMA and TEGDMA. Ascorbic acid as an antioxidant substance mitigates the oxidative damage associated with exposure to monomers.
Asunto(s)
Metacrilatos/efectos adversos , Odontoblastos/citología , Estrés Oxidativo/efectos de los fármacos , Polietilenglicoles/efectos adversos , Ácidos Polimetacrílicos/efectos adversos , Resinas Sintéticas/química , Apoptosis/efectos de los fármacos , Catalasa/metabolismo , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Hemo-Oxigenasa 1/metabolismo , Humanos , Mitocondrias/efectos de los fármacos , Odontoblastos/efectos de los fármacos , Odontoblastos/metabolismo , Especies Reactivas de Oxígeno/metabolismoRESUMEN
As traditional root canal obturation leads to the loss of the biological activity of the tooth, it is necessary to develop a material that promotes the regeneration of dental tissue. However, this remains a challenging task. Our study aims to construct a mineralized material to support the proliferation and differentiation of dental pulp stem cells (DPSCs), and to explore a new strategy for the treatment of pulp tissue necrosis. Mineralized keratin (M-keratin), defined as keratin that has been mineralized in simulated body fluid, was first harvested to construct the root canal filling material. Characterizations indicated that new substances or components were formed on the surface of keratin particles after mineralization, and the morphology of the keratin was changed. M-keratin promoted the growth, proliferation, and differentiation of DPSCs. After cultivation with M-keratin, DPSCs exhibited more extracellular matrix proteins interacting with the culture interface, the number of these cells increased significantly, and the 3-[4,5-dimethylthiazol-2-yl-]-2,5-diphenyltetrazolium bromide values of cells in the experimental group also increased. Meanwhile, signs that the DPSCs began to differentiate into odontoblasts were observed or detected by alizarin red S staining, ELISA, RNA-Seq, and western blot. We hope that this study will contribute to the development of a new material that promotes the regeneration of dental tissue as well as providing new ideas and strategies for the treatment of dental pulp disease.
Asunto(s)
Microambiente Celular/efectos de los fármacos , Queratinas/farmacología , Odontoblastos/efectos de los fármacos , Animales , Materiales Biomiméticos/química , Materiales Biomiméticos/farmacología , Líquidos Corporales/química , Calcificación Fisiológica/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Implantes Dentales , Pulpa Dental/citología , Pulpa Dental/fisiología , Humanos , Queratinas/química , Nanoestructuras/química , Odontoblastos/citología , Odontoblastos/fisiología , Ratas , Células Madre/efectos de los fármacos , Células Madre/fisiologíaRESUMEN
Increased expression of the transient receptor potential ankyrin 1 (TRPA1) channel has been detected in carious tooth pulp, suggesting involvement of TRPA1 in defense or repair of this tissue after exogenous noxious stimuli. This study aimed to elucidate the induction mechanism in response to lipopolysaccharide (LPS) stimulation and the function of TRPA1 in dental pulp cells. Stimulation of human dental pulp cells with LPS up-regulated TRPA1 expression, as demonstrated by quantitative RT-PCR and Western blotting. LPS stimulation also promoted nitric oxide (NO) synthesis and p38/mitogen-activated protein kinase (MAPK) phosphorylation. NOR5, an NO donor, up-regulated TRPA1 expression, whereas 1400W, an inhibitor of inducible nitric oxide synthase, and SB202190, a p38/MAPK inhibitor, down-regulated LPS-induced TRPA1 expression. Moreover, JT010, a TRPA1 agonist, increased the intracellular calcium concentration and extracellular signal-regulated kinase 1/2 phosphorylation, and up-regulated alkaline phosphatase mRNA in human dental pulp cells. Icilin-a TRPA1 agonist-up-regulated secreted phosphoprotein 1 mRNA expression and promoted mineralized nodule formation in mouse dental papilla cells. In vivo expression of TRPA1 was detected in odontoblasts along the tertiary dentin of carious teeth. In conclusion, this study demonstrated that LPS stimulation induced TRPA1 via the NO-p38 MAPK signaling pathway and TRPA1 agonists promoted differentiation or mineralization of dental pulp cells.
Asunto(s)
Diferenciación Celular/efectos de los fármacos , Pulpa Dental/citología , Odontoblastos/efectos de los fármacos , Canal Catiónico TRPA1/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Ancirinas/efectos de los fármacos , Ancirinas/genética , Ancirinas/metabolismo , Pulpa Dental/efectos de los fármacos , Proteínas de la Matriz Extracelular/efectos de los fármacos , Proteínas de la Matriz Extracelular/metabolismo , Humanos , Lipopolisacáridos/farmacología , Odontoblastos/citología , Transducción de Señal/efectos de los fármacos , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Canal Catiónico TRPA1/metabolismoRESUMEN
One of the main goals of dentistry is the natural preservation of the tooth structure following damage. This is particularly implicated in deep dental cavities affecting dentin and pulp, where odontoblast survival is jeopardized. This activates pulp stem cells and differentiation of new odontoblast-like cells, accompanied by increased Wnt signaling. Our group has shown that delivery of small molecule inhibitors of GSK3 stimulates Wnt/ß-catenin signaling in the tooth cavity with pulp exposure and results in effective promotion of dentin repair. Small molecules are a good therapeutic option due to their ability to pass across cell membranes and reach target. Here, we investigate a range of non-GSK3 target small molecules that are currently used for treatment of various medical conditions based on other kinase inhibitory properties. We analyzed the ability of these drugs to stimulate Wnt signaling activity by off-target inhibition of GSK3. Our results show that a c-Met inhibitor, has the ability to stimulate Wnt/ß-catenin pathway in dental pulp cells in vitro at low concentrations. This work is an example of drug repurposing for dentistry and suggests a candidate drug to be tested in vivo for natural dentin repair. This approach bypasses the high level of economical and time investment that are usually required in novel drug discoveries.