Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 198
Filter
1.
Arch Oral Biol ; 162: 105964, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38582010

ABSTRACT

OBJECTIVE: This study aimed to explore the effects of small extracellular vesicles derived from lipopolysaccharide-preconditioned dental follicle cells (L-D-sEV) on periodontal ligament cells from periodontitis affected teeth (p-PDLCs) in vitro and experimental periodontitis in mice. DESIGN: In vitro, the biological function of p-PDLCs and the underlying molecular mechanism were investigated by flow cytometry, Western blot, and quantitative real-time PCR (qRT-PCR) analysis. Eighteen-eight-week-old male C57BL/6 mice were randomly divided into three groups: control (Con), periodontitis (Peri), and L-D-sEV groups. Mice periodontitis model was induced by placing the 5-0 silk thread (around the maxillary second molar) and P.gingivalis (1 ×107 CFUs per mouse). In vivo, the alveolar bone loss, osteoclast activity, and macrophage polarization were measured by micro-computed tomography and histological analysis. RESULTS: In vitro, the RANKL/OPG ratio and phosphorylation of JNK and P38 protein levels of p-PDLCs were significantly decreased after L-D-sEV administration. Besides, flow cytometry and qRT-PCR analysis showed that L-D-sEV reduced apoptosis of p-PDLCs, down-regulated apoptosis-related genes Caspase-3 and BCL-2-Associated X expression, and up-regulated B-cell lymphoma-2 gene levels. In vivo, L-D-sEV administration significantly reduced alveolar bone loss, inhibited osteoclast activity, and induced M2 polarization. The histological analysis showed that iNOS/CD206, RANKL/OPG, p-JNK/JNK, and p-P38/P38 ratios were significantly lower in the L-D-sEV group than in the Peri group. CONCLUSIONS: L-D-sEV administration alleviated alveolar bone loss by mediating RANKL/OPG-related osteoclast activity and M2 macrophage polarization, alleviating p-PDLCs apoptosis and proliferation via the JNK and P38 pathways.


Subject(s)
Alveolar Bone Loss , Periodontitis , Mice , Male , Animals , Alveolar Bone Loss/pathology , Lipopolysaccharides/pharmacology , X-Ray Microtomography , Dental Sac/metabolism , Mice, Inbred C57BL , Periodontitis/metabolism , Apoptosis , Disease Models, Animal
2.
ACS Biomater Sci Eng ; 10(5): 3173-3187, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38605468

ABSTRACT

The application of bioengineering techniques for achieving bone regeneration in the oral environment is an increasingly prominent field. However, the clinical use of synthetic materials carries certain risks. The liquid phase of concentrated growth factor (LPCGF), as a biologically derived material, exhibits superior biocompatibility. In this study, LPCGF was employed as a tissue engineering scaffold, hosting dental follicle cells (DFCs) to facilitate bone regeneration. Both in vivo and in vitro experimental results demonstrate that this platform significantly enhances the expression of osteogenic markers in DFCs, such as alkaline phosphatase (ALP), runt-related transcription factor 2 (Runx2), and type I collagen (Col1a1). Simultaneously, it reduces the expression of inflammation-related genes, particularly interleukin-6 (IL-6) and interleukin-8 (IL-8), thereby alleviating the negative impact of the inflammatory microenvironment on DFCs. Further investigation into potential mechanisms reveals that this process is regulated over time by the WNT pathway. Our research results demonstrate that LPCGF, with its favorable physical characteristics, holds great potential as a scaffold. It can effectively carry DFCs, thereby providing an optimal initial environment for bone regeneration. Furthermore, LPCGF endeavors to closely mimic the mechanisms of bone healing post-trauma to facilitate bone formation. This offers new perspectives and insights into bone regeneration engineering.


Subject(s)
Bone Regeneration , Dental Sac , Intercellular Signaling Peptides and Proteins , Tissue Scaffolds , Bone Regeneration/drug effects , Dental Sac/cytology , Dental Sac/metabolism , Tissue Scaffolds/chemistry , Animals , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/pharmacology , Stem Cells/metabolism , Stem Cells/cytology , Osteogenesis , Humans , Tissue Engineering/methods
3.
Int J Mol Sci ; 24(22)2023 Nov 09.
Article in English | MEDLINE | ID: mdl-38003310

ABSTRACT

N6-methyladenosine (m6A) is the most abundant RNA modification, regulating gene expression in physiological processes. However, its effect on the osteogenic differentiation of dental follicle stem cells (DFSCs) remains unknown. Here, m6A demethylases, the fat mass and obesity-associated protein (FTO), and alkB homolog 5 (ALKBH5) were overexpressed in DFSCs, followed by osteogenesis assay and transcriptome sequencing to explore potential mechanisms. The overexpression of FTO or ALKBH5 inhibited the osteogenesis of DFSCs, evidenced by the fact that RUNX2 independently decreased calcium deposition and by the downregulation of the osteogenic genes OCN and OPN. MiRNA profiling revealed that miR-7974 was the top differentially regulated gene, and the overexpression of m6A demethylases significantly accelerated miR-7974 degradation in DFSCs. The miR-7974 inhibitor decreased the osteogenesis of DFSCs, and its mimic attenuated the inhibitory effects of FTO overexpression. Bioinformatic prediction and RNA sequencing analysis suggested that FK506-binding protein 15 (FKBP15) was the most likely target downstream of miR-7974. The overexpression of FKBP15 significantly inhibited the osteogenesis of DFSCs via the restriction of actin cytoskeleton organization. This study provided a data resource of differentially expressed miRNA and mRNA after the overexpression of m6A demethylases in DFSCs. We unmasked the RUNX2-independent effects of m6A demethylase, miR-7974, and FKBP15 on the osteogenesis of DFSCs. Moreover, the FTO/miR-7974/FKBP15 axis and its effects on actin cytoskeleton organization were identified in DFSCs.


Subject(s)
MicroRNAs , Osteogenesis , Core Binding Factor Alpha 1 Subunit/metabolism , Dental Sac/metabolism , Cells, Cultured , Cell Differentiation/genetics , MicroRNAs/metabolism , Stem Cells/metabolism
4.
Eur J Oral Sci ; 131(5-6): e12952, 2023.
Article in English | MEDLINE | ID: mdl-37664892

ABSTRACT

Dental follicle cells (DFCs) are osteogenic progenitor cells and are well suited for molecular studies of differentiation of alveolar osteoblasts. A recent study examined the metabolism in DFCs during osteogenic differentiation and showed that energy metabolism is increased after 14 days of differentiation (mid phase). However, previous studies have examined proteomes at early (2 h, 24 h) or very late (28 days) stages of differentiation, but not during the phase of increased metabolic activity. In this study, we examined the phosphoproteome at the mid phase (14 days) of osteogenic differentiation. Analysis of DFC phosphoproteomes showed that during this phase of osteogenic differentiation, proteins that are part of signal transduction are significantly regulated. Proteins involved in the regulation of the cytoskeleton and apoptosis were also increased in expression. As osteogenic differentiation induced oxidative stress and apoptosis in DFCs, the oxidative stress defense protein, catalase, was also upregulated during osteogenic differentiation, which supports the biomineralization of DFCs. In summary, this study revealed that during the middle phase (14 days) of osteogenic differentiation, processes in DFCs related to the control of cell organization, apoptosis, and oxidative stress are regulated.


Subject(s)
Osteogenesis , Proteome , Humans , Osteogenesis/physiology , Dental Sac/metabolism , Cell Differentiation/physiology , Stem Cells , Cells, Cultured
5.
Front Biosci (Landmark Ed) ; 28(5): 104, 2023 05 25.
Article in English | MEDLINE | ID: mdl-37258464

ABSTRACT

BACKGROUND: Dental follicle cells (DFCs) are promising candidates for tissue engineering. However, the molecular mechanisms that regulate the biological characteristics of DFCs are still unclear. Transient receptor potential melastatin 7 (TRPM7) is a Ca2+- and Mg2+-permeable cation channel. The aim of this study was to determine the impact of TRPM7 on the proliferation, migration and osteogenic differentiation of DFCs. METHODS: PCR, Western blotting, Immunocytochemical staining and Patch clamp methods were used to identify the gene and protein expression of TRPM7 in DFCs. DFCs were infected with lentiviruses that expressed either TRPM7 specific shRNA or scrambled non-effective shRNA to investigate its functional role. Cell proliferation and migration were assessed using Cell Counting Kit-8 assays and transwell cell culture chambers separately. Cell osteogenic differentiation were determined by ALP assay kit and Alizarin Red staining. RESULTS: Gene and protein expression of TRPM7 were detected in DFCs, but not of TRPM6, which is a closely related channel with similar function. In the absence of Mg2+, typical whole cell TRPM7-like currents were recorded by patch clamp. These were inhibited by low concentrations of 2-APB, but activated by high concentrations of 2-APB. Functional studies demonstrated that suppression of TRPM7 expression inhibited the proliferation and migration of DFCs, and promoted their osteogenic differentiation. Furthermore, Mg2+ deficiency mimicked the effects of TRPM7 knockdown in terms of osteogenic differentiation of DFCs. CONCLUSIONS: These results demonstrate that TRPM7 is involved in regulating the proliferation, migration and osteogenic differentiation of DFCs.


Subject(s)
Osteogenesis , TRPM Cation Channels , Humans , Osteogenesis/genetics , Magnesium/pharmacology , Magnesium/metabolism , TRPM Cation Channels/genetics , Dental Sac/metabolism , Cell Differentiation/genetics , Cell Proliferation/physiology , Cells, Cultured , RNA, Small Interfering/metabolism , Protein Serine-Threonine Kinases/metabolism
6.
Exp Gerontol ; 172: 112071, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36563529

ABSTRACT

Dental Follicle Cells (DFCs) are somatic stem cells with a limited lifespan, but little is known about a possible mechanism of cellular senescence. Previous studies have shown that cellular senescence is associated with increased demand of glycolsis or the "glycolytic metabotype", which can be induced by activation of 5' adenosine monophosphate-activated protein kinase (AMPK), and decreased autophagy. This study examined the role of AMPK in inducing senescence in DFCs. During the induction of cellular senescence, AMPK activity was impaired, suggesting a negative impact on senescence induction. In line with this assumption, cellular senescence was induced upon inhibition of AMPK with a specific siRNA. In addition, after this inhibition, autophagy was also inhibited. Moreover, specific inhibition of autophagy promoted cellular senescence. However, inducers of AMPK such as metformin or AICAR surprisingly increased senescence in DFCs. Interestingly, autophagy was impaired after long-term induction of AMPK with AICAR and metformin. Moreover, activation of AMPK induces the consumption of glucose but decreases NAD/NADH ratio in DFCs that suggest not only "glycolytic metabotype" of DFCs but also Mitochondrial Dysfunction Associated Senescence (MiDAS). Both changes are highly associated with the induction of cellular senescence. Hence, both AMPK activation and inhibition promote the induction of cellular senecence of DFCs.


Subject(s)
AMP-Activated Protein Kinases , Metformin , Humans , AMP-Activated Protein Kinases/metabolism , Dental Sac/metabolism , Cellular Senescence , Metformin/pharmacology , Phosphorylation , Autophagy
7.
Stem Cell Res Ther ; 13(1): 466, 2022 09 08.
Article in English | MEDLINE | ID: mdl-36076278

ABSTRACT

BACKGROUND: Dental follicle stem cells (DFSCs) show mesenchymal stem cell properties with the potential for alveolar bone regeneration. Stem cell properties can be impaired by reactive oxygen species (ROS), prompting us to examine the importance of scavenging ROS for stem cell-based tissue regeneration. This study aimed to investigate the effect and mechanism of N-acetylcysteine (NAC), a promising antioxidant, on the properties of DFSCs and DFSC-based alveolar bone regeneration. METHODS: DFSCs were cultured in media supplemented with different concentrations of NAC (0-10 mM). Cytologic experiments, RNA-sequencing and antioxidant assays were performed in vitro in human DFSCs (hDFSCs). Rat maxillary first molar extraction models were constructed, histological and radiological examinations were performed at day 7 post-surgery to investigate alveolar bone regeneration in tooth extraction sockets after local transplantation of NAC, rat DFSCs (rDFSCs) or NAC-treated rDFSCs. RESULTS: 5 mM NAC-treated hDFSCs exhibited better proliferation, less senescent rate, higher stem cell-specific marker and immune-related factor expression with the strongest osteogenic differentiation; other concentrations were also beneficial for maintaining stem cell properties. RNA-sequencing identified 803 differentially expressed genes between hDFSCs with and without 5 mM NAC. "Developmental process (GO:0032502)" was prominent, bioinformatic analysis of 394 involved genes revealed functional and pathway enrichment of ossification and PI3K/AKT pathway, respectively. Furthermore, after NAC treatment, the reduction of ROS levels (ROS, superoxide, hydrogen peroxide), the induction of antioxidant levels (glutathione, catalase, superoxide dismutase), the upregulation of PI3K/AKT signaling (PI3K-p110, PI3K-p85, AKT, phosphorylated-PI3K-p85, phosphorylated-AKT) and the rebound of ROS level upon PI3K/AKT inhibition were showed. Local transplantation of NAC, rDFSCs or NAC-treated rDFSCs was safe and promoted oral socket bone formation after tooth extraction, with application of NAC-treated rDFSCs possessing the best effect. CONCLUSIONS: The proper concentration of NAC enhances DFSC properties, especially osteogenesis, via PI3K/AKT/ROS signaling, and offers clinical potential for stem cell-based alveolar bone regeneration.


Subject(s)
Acetylcysteine , Osteogenesis , Acetylcysteine/metabolism , Acetylcysteine/pharmacology , Animals , Antioxidants/metabolism , Antioxidants/pharmacology , Cell Differentiation/physiology , Cells, Cultured , Dental Sac/metabolism , Humans , Osteogenesis/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA/metabolism , Rats , Reactive Oxygen Species/metabolism , Stem Cells/metabolism
8.
Stem Cell Res Ther ; 13(1): 486, 2022 09 30.
Article in English | MEDLINE | ID: mdl-36175952

ABSTRACT

Dental follicles are necessary for tooth eruption, surround the enamel organ and dental papilla, and regulate both the formation and resorption of alveolar bone. Dental follicle progenitor cells (DFPCs), which are stem cells found in dental follicles, differentiate into different kinds of cells that are necessary for tooth formation and eruption. Runt-related transcription factor 2 (Runx2) is a transcription factor that is essential for osteoblasts and osteoclasts differentiation, as well as bone remodeling. Mutation of Runx2 causing cleidocranial dysplasia negatively affects osteogenesis and the osteoclastic ability of dental follicles, resulting in tooth eruption difficulties. Among a variety of cells and molecules, Nel-like molecule type 1 (Nell-1) plays an important role in neural crest-derived tissues and is strongly expressed in dental follicles. Nell-1 was originally identified in pathologically fused and fusing sutures of patients with unilateral coronal synostosis, and it plays indispensable roles in bone remodeling, including roles in osteoblast differentiation, bone formation and regeneration, craniofacial skeleton development, and the differentiation of many kinds of stem cells. Runx2 was proven to directly target the Nell-1 gene and regulate its expression. These studies suggested that Runx2/Nell-1 axis may play an important role in the process of tooth eruption by affecting DFPCs. Studies on short and long regulatory noncoding RNAs have revealed the complexity of RNA-mediated regulation of gene expression at the posttranscriptional level. This ceRNA network participates in the regulation of Runx2 and Nell-1 gene expression in a complex way. However, non-study indicated the potential connection between Runx2 and Nell-1, and further researches are still needed.


Subject(s)
Calcium-Binding Proteins , Core Binding Factor Alpha 1 Subunit , Tooth Eruption , Bone Remodeling/genetics , Calcium-Binding Proteins/genetics , Cell Differentiation/genetics , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Dental Sac/metabolism , Humans , Osteogenesis/genetics , RNA , Stem Cells/metabolism , Tooth Eruption/genetics , Transcription Factors/genetics
9.
Int J Mol Sci ; 23(11)2022 May 25.
Article in English | MEDLINE | ID: mdl-35682637

ABSTRACT

Human dental follicle cells (DFCs) as periodontal progenitor cells are used for studies and research in regenerative medicine and not only in dentistry. Even if innovative regenerative therapies in medicine are often considered the main research area for dental stem cells, these cells are also very useful in basic research and here, for example, for the elucidation of molecular processes in the differentiation into mineralizing cells. This article summarizes the molecular mechanisms driving osteogenic differentiation of DFCs. The positive feedback loop of bone morphogenetic protein (BMP) 2 and homeobox protein DLX3 and a signaling pathway associated with protein kinase B (AKT) and protein kinase C (PKC) are presented and further insights related to other signaling pathways such as the WNT signaling pathway are explained. Subsequently, some works are presented that have investigated epigenetic modifications and non-coding ncRNAs and their connection with the osteogenic differentiation of DFCs. In addition, studies are presented that have shown the influence of extracellular matrix molecules or fundamental biological processes such as cellular senescence on osteogenic differentiation. The putative role of factors associated with inflammatory processes, such as interleukin 8, in osteogenic differentiation is also briefly discussed. This article summarizes the most important insights into the mechanisms of osteogenic differentiation in DFCs and is intended to be a small help in the direction of new research projects in this area.


Subject(s)
Dental Sac , Osteogenesis , Cell Differentiation/genetics , Cells, Cultured , Dental Sac/metabolism , Homeodomain Proteins/metabolism , Humans , Osteogenesis/genetics , Wnt Signaling Pathway
10.
Arch Oral Biol ; 141: 105484, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35749976

ABSTRACT

OBJECTIVE: The aim was to provide a comprehensive review of the current knowledge of the multiple roles of Runt-related transcription factor-2 (RUNX2) in regulating tooth eruption, focusing on the molecular mechanisms regarding tooth eruption mediated by RUNX2. DESIGN: Relevant literatures in PubMed, Medline, and Scopus database were searched, and a narrative review was performed. The multiple roles of RUNX2 in regulating tooth eruption was reviewed and discussed. RESULTS: Aberrant RUNX2 expression leads to disturbed or failed tooth eruption. Tooth eruption involves both the process of bone formation and bone resorption. RUNX2 promotes osteogenesis around the radicular portion of the dental follicle that provides the biological force for tooth eruption through inducing the expression of osteogenesis-related genes in dental follicle cells/osteoblasts. On the other hand, through indirect and direct pathways, RUNX2 regulates osteoclastogenesis and the formation of the eruption pathway. CONCLUSION: RUNX2 exerts a pivotal and complex influence in regulating tooth eruption. This review provides a better understanding of the function of RUNX2 in tooth eruption, which is beneficial to illuminate the precise molecular mechanism of osteogenesis and bone resorption, aiding the development of effective therapy for the failure of tooth eruption.


Subject(s)
Bone Resorption , Osteogenesis , Bone Resorption/metabolism , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Dental Sac/metabolism , Humans , Osteoclasts/metabolism , Osteogenesis/physiology , Tooth Eruption/physiology
11.
Int J Nanomedicine ; 17: 799-819, 2022.
Article in English | MEDLINE | ID: mdl-35228798

ABSTRACT

PURPOSE: Lipopolysaccharide (LPS) pretreatment can enhance the therapeutic effect of dental follicle stem cells-derived small extracellular vesicles (DFC-sEV) for periodontitis, and this study aimed to investigate the underlying mechanisms and clinical application Of LPS-preconditioned DFC-sEV in periodontitis. METHODS: The protein spectrum of DFC-sEV before and after LPS pretreatment was determined by liquid chromatography-tandem mass spectrometry and bioinformatic analysis. Their effects on inflammatory periodontal ligament stem cells (PDLSCs) and macrophages were investigated for cell proliferation, migration, type 2 macrophage (M2) polarization, and intracellular reactive oxygen species (ROS) levels separately. In addition, the regulation of ROS/Jun amino-terminal kinases (JNK) and ROS/extracellular signal-related kinases (ERK) signaling by LPS-preconditioned DFC-sEV was also studied to reveal the antioxidant mechanism. In vivo, two kinds of DFC-sEV loaded with 0.2% hyaluronic acid (HA) gel were applied for canine periodontitis to evaluate the therapeutic potential. RESULTS: The proteomic analysis showed that thirty-eight proteins were differentially expressed in LPS-preconditioned DFC-sEV, and interestingly, the highly expressed proteins were mainly involved in antioxidant and enzyme-regulating activities. In addition to promoting PDLSCs and macrophage proliferation, LPS-preconditioned DFC-sEV inhibited intracellular ROS as an antioxidant. It reduced the RANKL/OPG ratio of PDLSCs by inhibiting ROS/JNK signaling under inflammatory conditions and promoted macrophages to polarize toward the M2 phenotype via ROS/ERK signaling. Furthermore, LPS-preconditioned DFC-sEV loaded with the HA injectable system could sustainably release sEV and enhance the therapeutic efficacy for periodontitis in canines. CONCLUSION: LPS-preconditioned DFC-sEV could be effectively used as an auxiliary method for periodontitis treatment via antioxidant effects in a subgingival environment, and loading it with HA is feasible and effective for clinical applications.


Subject(s)
Extracellular Vesicles , Periodontitis , Antioxidants/metabolism , Antioxidants/pharmacology , Dental Sac/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Extracellular Vesicles/metabolism , Humans , Lipopolysaccharides/pharmacology , Periodontitis/therapy , Proteomics , Reactive Oxygen Species/metabolism , Stem Cells
12.
Cell Mol Life Sci ; 79(3): 153, 2022 Feb 26.
Article in English | MEDLINE | ID: mdl-35217915

ABSTRACT

Insight into human tooth epithelial stem cells and their biology is sparse. Tissue-derived organoid models typically replicate the tissue's epithelial stem cell compartment. Here, we developed a first-in-time epithelial organoid model starting from human tooth. Dental follicle (DF) tissue, isolated from unerupted wisdom teeth, efficiently generated epithelial organoids that were long-term expandable. The organoids displayed a tooth epithelial stemness phenotype similar to the DF's epithelial cell rests of Malassez (ERM), a compartment containing dental epithelial stem cells. Single-cell transcriptomics reinforced this organoid-ERM congruence, and uncovered novel, mouse-mirroring stem cell features. Exposure of the organoids to epidermal growth factor induced transient proliferation and eventual epithelial-mesenchymal transition, highly mimicking events taking place in the ERM in vivo. Moreover, the ERM stemness organoids were able to unfold an ameloblast differentiation process, further enhanced by transforming growth factor-ß (TGFß) and abrogated by TGFß receptor inhibition, thereby reproducing TGFß's known key position in amelogenesis. Interestingly, by creating a mesenchymal-epithelial composite organoid (assembloid) model, we demonstrated that the presence of dental mesenchymal cells (i.e. pulp stem cells) triggered ameloblast differentiation in the epithelial stem cells, thus replicating the known importance of mesenchyme-epithelium interaction in tooth development and amelogenesis. Also here, differentiation was abrogated by TGFß receptor inhibition. Together, we developed novel organoid models empowering the exploration of human tooth epithelial stem cell biology and function as well as their interplay with dental mesenchyme, all at present only poorly defined in humans. Moreover, the new models may pave the way to future tooth-regenerative perspectives.


Subject(s)
Dental Sac/metabolism , Organoids/metabolism , Ameloblasts/cytology , Ameloblasts/metabolism , Cell Differentiation , Cells, Cultured , Dental Sac/cytology , Epidermal Growth Factor/pharmacology , Epithelial Cells/cytology , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition/drug effects , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Humans , Organoids/cytology , Organoids/pathology , Phenotype , Receptor, Transforming Growth Factor-beta Type I/antagonists & inhibitors , Receptor, Transforming Growth Factor-beta Type I/metabolism , STAT2 Transcription Factor/genetics , STAT2 Transcription Factor/metabolism , Single-Cell Analysis , Stem Cells/cytology , Stem Cells/metabolism , Transcriptome , Transforming Growth Factor beta/metabolism
13.
Clin Oral Investig ; 26(3): 2817-2825, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34988693

ABSTRACT

OBJECTIVES: Dental follicle (DF) is made up of mesenchymal cells and fibers surrounding the enamel organ of a developing tooth. It has been shown that cystic and neoplastic lesions can develop from the pericoronal follicles of impacted third molars (ITMs). But the molecular transformation of DF tissues has not yet been uncovered and remains elusive. Accordingly, in the present study, we aimed to investigate the differential expression of lncRNA genes in DF tissues associated with asymptomatic impacted mandibular third molars (IMTMs) that do not show pathological pericoronal radiolucency in radiographic examination. MATERIAL AND METHODS: A total of 30 patients with unilateral mesioangular IMTMs were enrolled for the study. The expressions of lncRNA genes were determined in the DF and healthy gingival tissues obtained from study patients. For the determination of lncRNA expression levels, RNA was isolated from the obtained tissues, converted to cDNA samples, and analyzed by quantitative real-time PCR method. RESULTS: As a result, we found that the gene expression of MEG3 was increased about 10-fold in DF tissues compared to healthy gingival tissues (p < 0.0001). In addition, NORAD expression was found to be upregulated 4.2-fold (p = 0.0002) in DF tissues. Also, expression level of MALAT1 was found to be decreased 1.24-fold (p = 0.584) and TP73-AS1 increased 2.6-fold (p = 0.093) in DF tissues compared to healthy gingival tissues. CONCLUSIONS: Consequently, present findings suggest that differentially expressed lncRNAs in DFs might be associated with the various levels of cellular events including osteogenic differentiation, DNA damage, and the transformation into odontogenic pathology. CLINICAL RELEVANCE: Expression levels of MEG3 and NORAD lncRNA molecules may guide clinicians in the evaluation of asymptomatic ITM dental follicles that cannot be determined radiologically and during extraction of these teeth for prophylactic purposes.


Subject(s)
RNA, Long Noncoding , Tooth, Impacted , Dental Sac/metabolism , Humans , Molar, Third/pathology , Osteogenesis , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Tooth, Impacted/diagnostic imaging , Tooth, Impacted/genetics
14.
ACS Appl Mater Interfaces ; 13(48): 58152-58161, 2021 Dec 08.
Article in English | MEDLINE | ID: mdl-34808061

ABSTRACT

This study experimentally substantiates that the micromechanical compatibility between cell and substrate is essential for cells to achieve energetically favorable mechanotransduction that directs phenotypic transitions. The argument for this compatibility is based on a thermodynamic model that suggests that the response of cells to their substrate mechanical environment is a consequence of the interchange between forms of energy governing the cell-substrate interaction. Experimental validation for the model has been carried out by investigating the osteogenic differentiation of dental follicle stem cells (DFSCs) seeded on electrospun fibrous scaffolds. Electrospinning of blends containing polycaprolactone (PCL) and silk fibroin (SF) with varying composition of cellulose nanocrystals (CNCs) resulted in three-dimensional (3D) fibrous scaffolds with bimodal distribution of fiber diameter, which provides both macroscopically stiff and microscopically compliant scaffolds for cells without affecting the surface chemical functionality of scaffolds. Atomic force microscopy (AFM) with a colloidal probe and single-cell force spectroscopy were used to characterize cell stiffness and scaffold stiffness on the cellular level, as well as cell-scaffold adhesive interaction (chemical functionality). This study has successfully varied scaffold mechanical properties without affecting their surface chemistry. In vitro tests indicate that the micromechanical compatibility between cells and scaffolds has been significantly correlated with mechanosensitive gene expression markers and osteogenic differentiation markers of DFSCs. The agreement between experimental observations and the thermodynamic model affirms that the cellular response to the mechanical environment, though biological in nature, follows the laws of the energy interchange to achieve its self-regulating behavior. More importantly, this study provides systematic evidence, through extensive and rigorous experimental studies, for the first time that rationalizes that micromechanical compatibility is indeed important to the efficacy of regenerative medicine.


Subject(s)
Biocompatible Materials/metabolism , Dental Sac/metabolism , Stem Cells/metabolism , Tissue Scaffolds/chemistry , Animals , Biocompatible Materials/chemistry , Cell Differentiation , Dental Sac/chemistry , Materials Testing , Osteogenesis , Phenotype , Rats , Stem Cells/chemistry , Thermodynamics
15.
Front Endocrinol (Lausanne) ; 12: 730913, 2021.
Article in English | MEDLINE | ID: mdl-34456880

ABSTRACT

Most cells use calcium (Ca2+) as a second messenger to convey signals that affect a multitude of biological processes. The ability of Ca2+ to bind to proteins to alter their charge and conformation is essential to achieve its signaling role. Cytosolic Ca2+ (cCa2+) concentration is maintained low at ~100 nM so that the impact of elevations in cCa2+ is readily sensed and transduced by cells. However, such elevations in cCa2+ must be transient to prevent detrimental effects. Cells have developed a variety of systems to rapidly clear the excess of cCa2+ including Ca2+ pumps, exchangers and sequestering Ca2+ within intracellular organelles. This Ca2+ signaling toolkit is evolutionarily adapted so that each cell, tissue, and organ can fulfill its biological function optimally. One of the most specialized cells in mammals are the enamel forming cells, the ameloblasts, which also handle large quantities of Ca2+. The end goal of ameloblasts is to synthesize, secrete and mineralize a unique proteinaceous matrix without the benefit of remodeling or repair mechanisms. Ca2+ uptake into ameloblasts is mainly regulated by the store operated Ca2+ entry (SOCE) before it is transported across the polarized ameloblasts to reach the insulated enamel space. Here we review the ameloblasts Ca2+ signaling toolkit and address how the common electronegative non-metal fluoride can alter its function, potentially addressing the biology of dental fluorosis.


Subject(s)
Ameloblasts/metabolism , Calcification, Physiologic/drug effects , Calcium/metabolism , Dental Sac/metabolism , Epithelial Cells/metabolism , Fluorides/pharmacology , Ameloblasts/drug effects , Animals , Dental Sac/drug effects , Epithelial Cells/drug effects , Humans
16.
In Vitro Cell Dev Biol Anim ; 57(6): 620-630, 2021 Jun.
Article in English | MEDLINE | ID: mdl-34212339

ABSTRACT

Human dental follicle cells (HDFCs) are an ideal cell source of stem cells for dental tissue repair and regeneration and they have great potential for regenerative medicine applications. However, the conventional monolayer culture usually reduces cell proliferation and differentiation potential due to the continuous passage during in vitro expansion. In this study, primary HDFC spheroids were generated on 1% agarose, and the HDFCs spontaneously formed cell spheroids in the agarose-coated dishes. Compared with monolayer culture, the spheroid-derived HDFCs exhibited increased proliferative ability for later passage HDFCs as analysed by Cell Counting Kit-8 (CCK-8). The transcription-quantitative polymerase chain reaction (qRT-PCR), western blot and immunofluorescence assay showed that the expression of stemness marker genes Sox2, Oct4 and Nanog was increased significantly in the HDFC spheroids. Furthermore, we found that the odontogenic differentiation capability of HDFCs was significantly improved by spheroid culture in the agarose-coated dishes. On the other hand, the osteogenic differentiation capability was weakened compared with monolayer culture. Our results suggest that spheroid formation of HDFCs in agarose-coated dishes partially restores the proliferative ability of HDFCs at later passages, enhances their stemness and improves odontogenic differentiation capability in vitro. Therefore, spheroid formation of HDFCs has great therapeutic potential for stem cell clinical therapy.


Subject(s)
Cell Culture Techniques , Dental Sac/growth & development , Odontogenesis/drug effects , Spheroids, Cellular/drug effects , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Proliferation/drug effects , Dental Sac/cytology , Dental Sac/metabolism , Humans , Odontogenesis/genetics , Sepharose/pharmacology , Spheroids, Cellular/cytology , Stem Cells/drug effects
17.
Belo Horizonte; s.n; 2021. 99 p. ilus.
Thesis in Portuguese | BBO - Dentistry | ID: biblio-1344019

ABSTRACT

Envelhecer não é uma escolha; é nosso destino. O declínio funcional dependente do tempo que afeta a maioria dos organismos vivos está vinculado às alterações em variados processos celulares, incluindo senescência celular, alterações epigenéticas, instabilidade genômica, exaustão de células-tronco, entre outras. As alterações morfológicas relacionadas à idade nos folículos dentários associados a dentes não-erupcionados têm sido investigadas há décadas, principalmente motivado pelo fato de que cistos e tumores podem surgir em associação a dentes inclusos e/ou impactados. Quanto mais entendemos a fisiologia dos folículos dentários, mais nos tornamos capazes de contextualizar eventos biológicos que podem estar associados à ocorrência de lesões odontogênicas cuja incidência aumenta com a idade. Assim, nosso objetivo foi avaliar as alterações metabólicas relacionadas à idade em amostras de folículos dentários associados à terceiros molares inferiores inclusos/impactados de indivíduos adolescentes e adultos. Uma amostra de conveniência de folículos dentários fixados em formalina e embebidos em parafina de indivíduos adolescentes (<16 anos, n= 13) e adultos (>26 anos, n= 7) foi selecionada. As amostras foram preparadas e submetidas à cromatografia líquida de alta eficiência acoplada a espectrometria de massa (HPLC-MS) em análise metabolômica untargeted. Análises uni- e multivariadas foram conduzidas, e a previsão de vias alteradas foi realizada pelas abordagens mummichog e GSEA. Folículos dentários de indivíduos adolescentes e adultos mostraram diferenças nas vias relacionadas à biossíntese do hormônio esteróide C21, biossíntese dos ácidos biliares, metabolismo da galactose, biossíntese de androgênio e estrogênio, metabolismo do amido e sacarose e metabolismo do lipoato. Nossos achados sugerem que, assim como outros tecidos humanos, os folículos dentários associados a dentes não erupcionados apresentam, durante o envelhecimento, alterações em nível metabólico, o que pode abrir caminho para novos estudos sobre biologia, fisiologia e patologia oral.


Aging is not a matter of choice; it is our fate. The 'time-dependent functional decline that affects most living organisms' is coupled with several alterations in cellular processes, including cell senescence, epigenetic alterations, genomic instability, stem cell exhaustion, amongst others. Age-related morphological changes in dental follicles have been investigated for decades, mainly motivated by the fact that cysts and tumors may arise in association with a unerupted and/or impacted teeth. The more we understand dental follicles' physiology, the more we become able to contextualize biological events that can be associated with the occurrence of odontogenic lesions which incidence increases with age. Thus, our objective was to assess age-related changes in metabolic pathways of dental follicles associated with unerupted/impacted mandibular third molars from young and adult individuals. For this purpose, a convenience sample of formalin-fixed paraffin-embedded dental follicles from young (<16 y.o., n = 13) and adult (>26 y.o., n = 7) individuals was selected. Samples were analyzed by high-performance liquid chromatography-mass spectrometry (HPLC-MS)-based untargeted metabolomics. Multivariate and univariate analyses were conducted, and the prediction of altered pathways was performed by mummichog and GSEA approaches. Dental follicles from young and older individuals showed differences in pathways related to C21-steroid hormone biosynthesis, bile acid biosynthesis, galactose metabolism, androgen and estrogen biosynthesis, starch and sucrose metabolism and lipoate metabolism. Our findings support that similar to other human tissues, dental follicles associated with unerupted tooth show alterations at a metabolic level with aging, which can pave the way for further studies on oral pathology, oral biology and physiology.


Subject(s)
Pathology, Oral , Tooth, Unerupted , Aging , Dental Sac/metabolism , Dental Physiological Phenomena , Mass Spectrometry , Chromatography, Liquid , Metabolomics
18.
Sci Rep ; 10(1): 17567, 2020 10 16.
Article in English | MEDLINE | ID: mdl-33067558

ABSTRACT

Ameloblastomas are epithelial odontogenic tumours that, although benign, are locally invasive and may exhibit aggressive behaviour. In the tumour microenvironment, the concentration of oxygen is reduced, which leads to intratumoral hypoxia. Under hypoxia, the crosstalk between the HIF-1α, MMP-2, VEGF, and VEGFR-2 proteins has been associated with hypoxia-induced angiogenesis, leading to tumour progression and increased invasiveness. This work showcases 24 ameloblastoma cases, 10 calcifying odontogenic cysts, and 9 dental follicles, used to investigate the expression of these proteins by immunohistochemistry. The anti-HIF-1α, anti-MMP-2, anti-VEGF, and anti-VEGFR-2 primary antibodies are used in this work. The results have been expressed by the mean grey value after immunostaining in images acquired with an objective of 40×. The ameloblastoma samples showed higher immunoexpression of HIF-1α, MMP-2, VEGF, and VEGFR-2 when compared to the dental follicles and calcifying odontogenic cysts. Ameloblastomas show a higher degree of expression of proteins associated with intratumoral hypoxia and proangiogenic proteins, which indicates the possible role of these proteins in the biological behaviour of this tumour.


Subject(s)
Ameloblastoma/metabolism , Ameloblastoma/pathology , Hypoxia , Neovascularization, Pathologic , Odontogenic Tumors/metabolism , Odontogenic Tumors/pathology , Biomarkers/metabolism , Dental Sac/metabolism , Disease Progression , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunohistochemistry , Matrix Metalloproteinase 2/metabolism , Neoplasm Invasiveness , Prognosis , Tumor Microenvironment , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
19.
Int J Oral Sci ; 12(1): 20, 2020 06 30.
Article in English | MEDLINE | ID: mdl-32606293

ABSTRACT

As a member of the AFF (AF4/FMR2) family, AFF4 is a transcription elongation factor that is a component of the super elongation complex. AFF4 serves as a scaffolding protein that connects transcription factors and promotes gene transcription through elongation and chromatin remodelling. Here, we investigated the effect of AFF4 on human dental follicle cells (DFCs) in osteogenic differentiation. In this study, we found that small interfering RNA-mediated depletion of AFF4 resulted in decreased alkaline phosphatase (ALP) activity and impaired mineralization. In addition, the expression of osteogenic-related genes (DLX5, SP7, RUNX2 and BGLAP) was significantly downregulated. In contrast, lentivirus-mediated overexpression of AFF4 significantly enhanced the osteogenic potential of human DFCs. Mechanistically, we found that both the mRNA and protein levels of ALKBH1, a critical regulator of epigenetics, changed in accordance with AFF4 expression levels. Overexpression of ALKBH1 in AFF4-depleted DFCs partially rescued the impairment of osteogenic differentiation. Our data indicated that AFF4 promoted the osteogenic differentiation of DFCs by upregulating the transcription of ALKBH1.


Subject(s)
Dental Sac/metabolism , Osteogenesis/genetics , Transcription Factors/metabolism , Transcriptional Elongation Factors/metabolism , Biomarkers/metabolism , Cell Differentiation , Cells, Cultured , Dental Sac/drug effects , Gene Expression Regulation , Humans , Repressor Proteins , Transcription Factors/genetics
20.
Histochem Cell Biol ; 154(4): 397-403, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32710187

ABSTRACT

Dental follicle cells (DFCs) are progenitor cells for mineralizing cells such as alveolar osteoblasts, but little is known about the mechanisms of the differentiation. Interestingly, different cell lines sometimes have different potentials to differentiate into mineralizing cells. In this study, we compared two different DFC lines, with one cell line (DFC_B) showing a high alkaline phosphatase (ALP) activity in long-term cultures with standard medium and a reliable mineralizing potential. However, the other cell line DFC_A shows low ALP activity in standard medium and almost no mineralization. Known osteogenic markers such as RUNX2 were similarly expressed in both cell lines. However, the proosteogenic signaling pathway of the bone morphogenetic protein (BMP) is induced in DFC_B, and the parathyroid hormone-related protein (PTHrP), which is involved in tooth root development, was also expressed more strongly. Previous studies have shown that the secreted PTHrP negatively regulate the transition from pre-osteoblastic progenitors to osteoblasts, but we showed that an inhibition of PTHrP gene expression reduced the ALP activity and the BMP-signaling pathway. In addition, endogenously expressed PTHrP is located in the cell nucleus. In contrast, supplementation of PTHrP or an inhibitor for the PTHrP receptor did not affect the ALP activity of DFC_B. In conclusion, our data suggest that a high endogenous expression of PTHrP in DFCs supports the induction of osteogenic differentiation via an intracrine mode.


Subject(s)
Dental Sac/metabolism , Osteogenesis/genetics , Parathyroid Hormone-Related Protein/genetics , Cell Differentiation/genetics , Cells, Cultured , Dental Sac/cytology , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Humans , Parathyroid Hormone-Related Protein/analysis , Parathyroid Hormone-Related Protein/metabolism , Staining and Labeling
SELECTION OF CITATIONS
SEARCH DETAIL
...