Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 88
Filter
1.
J Mol Histol ; 52(1): 63-75, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33141361

ABSTRACT

Much information is currently available for molecules in early odontogenesis, but there is limited knowledge regarding terminal cytodifferentiation of ameloblasts and odontoblasts for the determination of normal crown morphology. The present differential display PCR (DD-PCR) revealed that insulin-like growth factor-binding protein 5 (IGFBP5) was differentially expressed in molar tooth germs between the cap (before crown mineralization) and root formation (after crown mineralization) stages. Real-time PCR confirmed that the expression levels of IGFBP1-4 were not significantly changed but those of IGFBP5-7 were upregulated in a time-dependent manner. Immunoreactivities for IGFBP5-7 were hardly seen in molar germs at the cap/early bell stage and protective-stage ameloblasts at the root formation stage. However, the reactivity was strong in odontoblasts and maturation-stage ameloblasts, which are morphologically and functionally characterized by wide intercellular space and active enamel matrix mineralization. The localization of each IGFBP was temporospatial. IGFBP5 was localized in the nuclei of fully differentiated odontoblasts and ameloblasts, while IGFBP6 was localized in the apical cytoplasm of ameloblasts and odontoblasts with dentinal tubules, and IGFBP7 was mainly found in the whole cytoplasm of odontoblasts and the intercellular space of ameloblasts. IGFBP silencing using specific siRNAs upregulated representative genes for dentinogenesis and amelogenesis, such as DMP1 and amelogenin, respectively, and augmented the differentiation media-induced mineralization, which was confirmed by alizarin red s and alkaline phosphatase staining. These results suggest that IGFBP5-7 may play independent and redundant regulatory roles in late-stage odontogenesis by modulating the functional differentiation of ameloblasts and odontoblasts.


Subject(s)
Insulin-Like Growth Factor Binding Proteins/metabolism , Odontogenesis , Tooth Calcification , Amelogenesis/genetics , Animals , Dental Enamel/metabolism , Dentin/metabolism , Gene Expression Regulation , Insulin-Like Growth Factor Binding Proteins/genetics , Molar/metabolism , Odontoblasts/metabolism , Odontogenesis/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Sprague-Dawley , Tooth Calcification/genetics , Tooth Germ/metabolism , Up-Regulation/genetics
2.
J World Fed Orthod ; 9(3): 101-105, 2020 09.
Article in English | MEDLINE | ID: mdl-32943154

ABSTRACT

OBJECTIVES: The purpose of this study was to determine if shear bond resistance of orthodontic brackets bonded to enamel is associated with genes implicated in the enamel mineralization process. METHODS: Ninety-two permanent, caries-free premolars extracted for orthodontic purposes and their associated saliva samples were obtained. Eighteen single nucleotide polymorphisms (SNPs) were studied for association with shear bond resistance. The genes of interest in this study were those previously associated with dental caries by our group. All tooth samples were bonded on the buccal surface with metallic lower lateral brackets, and then subjected to physical debonding. The force required to debond the bracket was recorded in Newtons (N) and converted to a shear bond resistance value in Megapascals (N/mm2). The data were analyzed for statistical significance as compared with the mean shear bond resistance value via PLINK whole genome analysis software. RESULTS: Associations were found between the SNPs for tuftelin (rs7526319, P = 0.004) and amelogenin (rs17878486, P = 0.04) and a higher shear bond resistance. CONCLUSION: The collected data support the proposed hypothesis that genes involved in the mineralization process affect the bonding of orthodontic brackets, and such an association is of value for the field of orthodontics, particularly in evaluating the efficacy of enamel-resin bond strength for patients receiving treatment.


Subject(s)
Dental Enamel/physiology , Orthodontic Brackets/adverse effects , Polymorphism, Single Nucleotide , Tooth Calcification/genetics , Adolescent , Cross-Sectional Studies , Female , Humans , Male , Risk Factors , Shear Strength , Tooth Demineralization/genetics
3.
Biochem Biophys Res Commun ; 501(1): 206-211, 2018 06 18.
Article in English | MEDLINE | ID: mdl-29709481

ABSTRACT

FAM83H was identified as the major causative gene for autosomal dominant hypocalcified amelogenesis imperfect (ADHCAI). The pathogenic mechanism of FAM83H in ADHCAI remains elusive. The present study aims to investigate the effect of Fam83h mutation on the mineralization of mouse ameloblast cell line LS8 and to explore the possible pathogenesis of ADHCAI. Lentivirus package was performed for the plasmids with mouse Fam83h mutant cDNA (c.1186C > T, M3) and empty vector (Control) and transfected into LS8, which were divided into M3-FLAG and Control groups. Immunoprecipitation, western-blot and immunofluorescence were performed to detect the expression and subcellular localization of Fam83 h, CK1α and ß-catenin. ALP activity, ALP staining, expression of the mineralization factors were detected in two groups during mineralization induction. Expression of the mineralization factors was also detected in M3-FLAG and LS8 exposing to pyrvinium pamoate. Compared with the Control, the Fam83h mutation altered the expression and localization of Fam83 h, CK1α and ß-catenin in LS8, inhibited the mineralization and down-regulated the expression of mineralization factors in M3-FLAG. Pyrvinium pamoate, an inhibitor of the Wnt/ß-catenin signaling pathway, up-regulated expression of mineralization factors in LS8 and rescued the inhibited mineralization in M3-FLAG. The results indicated that the Fam83h mutation could inhibit the mineralization in ameloblasts by activating Wnt/ß-catenin signaling pathway.


Subject(s)
Ameloblasts/metabolism , Proteins/genetics , Proteins/metabolism , Amelogenesis/genetics , Amelogenesis/physiology , Amelogenesis Imperfecta/etiology , Amelogenesis Imperfecta/genetics , Amelogenesis Imperfecta/metabolism , Animals , Casein Kinase I/genetics , Casein Kinase I/metabolism , Cell Line , Humans , Mice , Mutant Proteins/genetics , Mutant Proteins/metabolism , Mutation , Tooth Calcification/genetics , Tooth Calcification/physiology , Transfection , Wnt Signaling Pathway , beta Catenin/metabolism
4.
J Dent Res ; 97(2): 209-217, 2018 02.
Article in English | MEDLINE | ID: mdl-28892649

ABSTRACT

The importance of phosphate (Pi) as an essential component of hydroxyapatite crystals suggests a key role for membrane proteins controlling Pi uptake during mineralization in the tooth. To clarify the involvement of the currently known Pi transporters (Slc17a1, Slc34a1, Slc34a2, Slc34a3, Slc20a1, Slc20a2, and Xpr1) during tooth development and mineralization, we determined their spatiotemporal expression in murine tooth germs from embryonic day 14.5 to postnatal day 15 and in human dental samples from Nolla stages 6 to 9. Using real-time polymerase chain reaction, in situ hybridization, immunohistochemistry, and X-gal staining, we showed that the expression of Slc17a1, Slc34a1, and Slc34a3 in tooth germs from C57BL/6 mice were very low. In contrast, Slc34a2, Slc20a1, Slc20a2, and Xpr1 were highly expressed, mostly during the postnatal stages. The expression of Slc20a2 was 2- to 10-fold higher than the other transporters. Comparable results were obtained in human tooth germs. In mice, Slc34a2 and Slc20a1 were predominantly expressed in ameloblasts but not odontoblasts, while Slc20a2 was detected neither in ameloblasts nor in odontoblasts. Rather, Slc20a2 was highly expressed in the stratum intermedium and the subodontoblastic cell layer. Although Slc20a2 knockout mice did not show enamel defects, mutant mice showed a disrupted dentin mineralization, displaying unmerged calcospherites at the mineralization front. This latter phenotypical finding raises the possibility that Slc20a2 may play an indirect role in regulating the extracellular Pi availability for mineralizing cells rather than a direct role in mediating Pi transport through mineralizing plasma cell membranes. By documenting the spatiotemporal expression of Pi transporters in the tooth, our data support the possibility that the currently known Pi transporters may be dispensable for the initiation of dental mineralization and may rather be involved later during the tooth mineralization scheme.


Subject(s)
Phosphate Transport Proteins/metabolism , Tooth Calcification/genetics , Animals , Female , France , Humans , Immunohistochemistry , In Situ Hybridization , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Real-Time Polymerase Chain Reaction , Tooth Germ/embryology , Tooth Germ/metabolism , X-Ray Microtomography , Xenotropic and Polytropic Retrovirus Receptor
5.
Eur J Oral Sci ; 125(1): 8-17, 2017 02.
Article in English | MEDLINE | ID: mdl-28084688

ABSTRACT

Latent-transforming growth factor beta-binding protein 3 (LTBP-3) is important for craniofacial morphogenesis and hard tissue mineralization, as it is essential for activation of transforming growth factor-ß (TGF-ß). To investigate the role of LTBP-3 in tooth formation we performed micro-computed tomography (micro-CT), histology, and scanning electron microscopy analyses of adult Ltbp3-/- mice. The Ltbp3-/- mutants presented with unique craniofacial malformations and reductions in enamel formation that began at the matrix formation stage. Organization of maturation-stage ameloblasts was severely disrupted. The lateral side of the incisor was affected most. Reduced enamel mineralization, modification of the enamel prism pattern, and enamel nodules were observed throughout the incisors, as revealed by scanning electron microscopy. Molar roots had internal irregular bulbous-like formations. The cementum thickness was reduced, and microscopic dentinal tubules showed minor nanostructural changes. Thus, LTBP-3 is required for ameloblast differentiation and for the formation of decussating enamel prisms, to prevent enamel nodule formation, and for proper root morphogenesis. Also, and consistent with the role of TGF-ß signaling during mineralization, almost all craniofacial bone components were affected in Ltbp3-/- mice, especially those involving the upper jaw and snout. This mouse model demonstrates phenotypic overlap with Verloes Bourguignon syndrome, also caused by mutation of LTBP3, which is hallmarked by craniofacial anomalies and amelogenesis imperfecta phenotypes.


Subject(s)
Amelogenesis/genetics , Dental Enamel/abnormalities , Latent TGF-beta Binding Proteins/genetics , Ameloblasts/metabolism , Amelogenesis Imperfecta/genetics , Animals , Dental Enamel/ultrastructure , Genotype , Male , Mice , Mice, Mutant Strains , Microscopy, Electron, Scanning , Mutation , Osteochondrodysplasias/genetics , Phenotype , Tooth Calcification/genetics , Transforming Growth Factor beta/genetics , X-Ray Microtomography
6.
Acta Odontol Scand ; 75(2): 144-153, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28030993

ABSTRACT

OBJECTIVES: Incremental markings in dental enamel suggest that the circadian clock may influence the molecular underpinnings orchestrating enamel formation. The aim of this study was to investigate whether the genes and microRNAs (miRNAs) oscillate in a circadian pattern during tooth and enamel development. MATERIAL AND METHODS: Comparative gene and miRNA expression profiling of the first mandibular molar tooth germ isolated at different time-points during the light and night period was performed using microarrays and validated using real-time RT-PCR. Bioinformatic analysis was carried out using Ingenuity Pathway Analysis (IPA), and TargetScan software was used in order to identify computationally predicted miRNA-mRNA target relationships. RESULTS: In total, 439 genes and 32 miRNAs exhibited significantly different (p < 0.05) levels of expression in the light phase compared with the night phase tooth germs. Genes involved in enamel formation, i.e. Amelx, Ambn, Amtn, and Odam, oscillated in a circadian pattern. Furthermore, the circadian clock genes, in particular Clock and Bmal1, oscillated in mouse molar tooth germ during 24-h intervals. The expression of Clock and Bmal1 was inversely correlated with the expression of miR-182 and miR-141, respectively. CONCLUSIONS: MiRNAs, including miR-182 and miR-141, are involved in the control of peripheral circadian rhythms in the developing tooth by regulating the expression of genes coding for circadian transcription factors such as CLOCK and BMAL1. Regulation of circadian rhythms may be important for enamel phenotype, and the morphology of dental enamel may vary between individuals due to differences in circadian profiles.


Subject(s)
Circadian Rhythm , Gene Expression Regulation, Developmental , Molar/growth & development , Tooth Calcification/genetics , Tooth Germ/growth & development , Amelogenesis , Animals , Dental Enamel/growth & development , Mice , MicroRNAs , Molar/chemistry , Odontogenesis/physiology , RNA, Messenger/analysis
7.
J Endod ; 42(5): 752-9, 2016 May.
Article in English | MEDLINE | ID: mdl-27020968

ABSTRACT

INTRODUCTION: This study determined the gene expression profiles of the human coronal pulp (CP) and apical pulp complex (APC) with the aim of explaining differences in their functions. METHODS: Total RNA was isolated from the CP and APC, and gene expression was analyzed using complementary DNA microarray technology. Gene ontology analysis was used to classify the biological function. Quantitative reverse-transcription polymerase chain reaction and immunohistochemical staining were performed to verify microarray data. RESULTS: In the microarray analyses, expression increases of at least 2-fold were present in 125 genes in the APC and 139 genes in the CP out of a total of 33,297 genes. Gene ontology class processes found more genes related to immune responses, cell growth and maintenance, and cell adhesion in the APC, whereas transport and neurogenesis genes predominated in the CP. Quantitative reverse-transcription polymerase chain reaction and immunohistochemical staining confirmed the microarray results, with DMP1, CALB1, and GABRB1 strongly expressed in the CP, whereas SMOC2, SHH, BARX1, CX3CR1, SPP1, COL XII, and LAMC2 were strongly expressed in the APC. CONCLUSIONS: The expression levels of genes related to dentin mineralization, neurogenesis, and neurotransmission are higher in the CP in human immature teeth, whereas those of immune-related and tooth development-related genes are higher in the APC.


Subject(s)
Dental Pulp/growth & development , Gene Expression , Odontogenesis/genetics , Tooth Apex/growth & development , Adolescent , CX3C Chemokine Receptor 1 , Calbindin 1/genetics , Calcium-Binding Proteins/genetics , Cell Adhesion/genetics , Child , Child, Preschool , Collagen Type XII/genetics , Dental Pulp/anatomy & histology , Dental Pulp/cytology , Dental Pulp/diagnostic imaging , Extracellular Matrix Proteins/genetics , Female , Gene Expression Profiling , Hedgehog Proteins/genetics , Homeodomain Proteins/genetics , Humans , Immunohistochemistry , Laminin/genetics , Male , Microarray Analysis/methods , Neurogenesis/genetics , Osteopontin/genetics , Phosphoproteins/genetics , RNA/analysis , Real-Time Polymerase Chain Reaction/methods , Receptors, Chemokine/genetics , Receptors, GABA-A/genetics , Republic of Korea , Synaptic Transmission/genetics , Tooth Apex/anatomy & histology , Tooth Apex/cytology , Tooth Apex/diagnostic imaging , Tooth Calcification/genetics , Transcription Factors/genetics , Young Adult
8.
PLoS One ; 10(5): e0127871, 2015.
Article in English | MEDLINE | ID: mdl-26010981

ABSTRACT

The molar tooth of the crayfish Cherax quadricarinatus is part of the mandible, and is covered by a layer of apatite (calcium phosphate). This tooth sheds and is regenerated during each molting cycle together with the rest of the exoskeleton. We discovered that molar calcification occurs at the pre-molt stage, unlike calcification of the rest of the new exoskeleton. We further identified a novel molar protein from C. quadricarinatus and cloned its transcript from the molar-forming epithelium. We termed this protein Cq-M13. The temporal level of transcription of Cq-M13 in an NGS library of molar-forming epithelium at different molt stages coincides with the assembly and mineralization pattern of the molar tooth. The predicted protein was found to be related to the pro-resilin family of cuticular proteins. Functionally, in vivo silencing of the transcript caused molt cycle delay and a recombinant version of the protein was found to bind chitin and exhibited elastic properties.


Subject(s)
Arthropod Proteins/metabolism , Astacoidea/metabolism , Chitin/metabolism , Molting/physiology , Tooth/metabolism , Amino Acid Sequence , Animals , Arthropod Proteins/genetics , Astacoidea/growth & development , Cloning, Molecular , Elasticity , Epithelium/metabolism , Gene Expression , Molecular Sequence Data , Molting/genetics , Phylogeny , Protein Binding , Tooth/growth & development , Tooth Calcification/genetics , Tooth Calcification/physiology
9.
J Dent Res ; 94(6): 836-42, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25792521

ABSTRACT

Amelogenins are proteins formed by alternative splicing of the amelogenin gene, and are essential for tooth enamel formation. However, the unique functions of various alternatively spliced amelogenins in enamel formation are not well understood. In this study, we determined the spatiotemporal location of amelogenins derived from transcripts containing exon4 (AMG+4) in the enamel matrix, and the relative binding of recombinant AMG+4 to hydroxyapatite (HAP). Immunohistochemistry and mass spectrometry analyses showed that AMG+4 proteins were secreted into the enamel matrix at the early maturation stage. A stage-specific increase in the synthesis of AMG+4 was further supported by our observation that in mice overexpressing leucine-rich amelogenin peptide (TgLRAP), in which ameloblasts differentiate earlier, AMG+4 transcripts were also upregulated earlier. In vitro binding studies, supported by in silico modeling of protein binding to calcium and phosphate, showed that more recombinant AMG+4 bound to hydroxyapatite (HAP) as compared with recombinant AMG-4. The temporal and spatial localization of amelogenins containing exon4 peptide, and their functional differences in HAP binding, suggests that the unique properties of amelogenins containing exon4 cause a specific enhancement of biomineralization related to stabilization of early-formed HAP at the maturation stage.


Subject(s)
Amelogenin/genetics , Exons/genetics , Tooth Calcification/genetics , Alternative Splicing/genetics , Ameloblasts/physiology , Amelogenesis/genetics , Animals , Calcium/metabolism , Cell Differentiation/physiology , Dental Enamel Proteins/genetics , Durapatite/metabolism , Female , Mass Spectrometry , Mice , Phosphates/metabolism , Protein Binding , Rats , Rats, Wistar , Recombinant Proteins , Time Factors , Up-Regulation
10.
J Dent Res ; 94(5): 697-705, 2015 May.
Article in English | MEDLINE | ID: mdl-25715379

ABSTRACT

Amelotin (AMTN) is a relatively recently discovered enamel protein that is predominantly expressed by ameloblasts during the maturation stage of amelogenesis and is present at lower levels in the junctional epithelium of erupted teeth. Previous studies have suggested a function of this protein in enamel mineralization and cell attachment. Genetic mouse models have been instrumental in defining the role of many enamel-related proteins, but a genetic mouse model lacking the Amtn gene has not been reported. Here, we describe the generation of amelotin-deficient mice and the analysis of their enamel phenotype in comparison with that of wild-type animals. Ablation of AMTN expression resulted in mechanically inferior enamel of mandibular incisors that showed chipping and fractures at the incisal edge. Enamel mineralization was delayed, resulting in hypomineralized inner enamel and structural defects in the outer enamel. Erupted enamel close to the gingival margin showed increased surface roughness. The expression levels of the enamel matrix proteins AMEL, AMBN, ENAM, and ODAM and the enamel proteases MMP-20 and KLK-4 were not significantly altered, although the expression of KLK-4 was delayed. The morphology of ameloblasts showing prominent Tomes' processes during the secretory stage was not altered, and there was no indication of disruption of cell structures or activities, but a residual layer, presumably consisting of organic material, remained at the enamel surface close to the gingival margin. The integrity of the dentogingival attachment at the junctional epithelium appeared unaffected by AMTN deficiency. These observations indicate that AMTN plays a subtle yet critical role in enamel biomineralization, particularly during the establishment of the outer and surface enamel layers. This role appears to be largely independent of other enamel proteins.


Subject(s)
Dental Enamel Hypoplasia/genetics , Dental Enamel Proteins/genetics , Ameloblasts/pathology , Amelogenesis/genetics , Amelogenin/analysis , Animals , Cell Adhesion/physiology , Dental Enamel/ultrastructure , Dental Enamel Hypoplasia/pathology , Dental Enamel Proteins/analysis , Epithelial Attachment/pathology , Gingiva/pathology , Intracellular Signaling Peptides and Proteins , Kallikreins/analysis , Matrix Metalloproteinase 20/analysis , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Transgenic , Microscopy, Electron, Transmission , Phenotype , Proteins/analysis , Tooth Calcification/genetics
11.
J Exp Zool B Mol Dev Evol ; 322(6): 390-402, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25243252

ABSTRACT

The coelacanth is the basal-most extant sarcopterygian that has teeth and tooth-like structures, comprising bone, dentin, and enamel or enameloid. Formation of these tissues involves many members of the secretory calcium-binding protein (SCPP) family. In tetrapods, acidic-residue-rich SCPPs are used in mineralization of bone and dentin, whereas Pro/Gln-rich SCPPs participate in enamel formation. Teleosts also employ many SCPPs for tissue mineralization. Nevertheless, the repertoire of SCPPs is largely different in teleosts and tetrapods; hence, filling this gap would be critical to elucidate early evolution of mineralized tissues in osteichthyans. In the present study, we searched for SCPP genes in the coelacanth genome and identified 11, of which two have clear orthologs in both tetrapods and teleosts, seven only in tetrapods, and two in neither of them. Given the divergence times of these vertebrate lineages, our discovery of this many SCPP genes shared between the coelacanth and tetrapods, but not with teleosts, suggests a complicated evolutionary scheme of SCPP genes in early osteichthyans. Our investigation also revealed both conserved and derived characteristics of SCPPs in the coelacanth and other vertebrates. Notably, acidic SCPPs independently evolved various acidic repeats in different lineages, while maintaining high acidity, presumably important for interactions with calcium. Furthermore, the three Pro/Gln-rich SCPP genes, required for mineralizing enamel matrix and confirmed only in tetrapods, were all identified in the coelacanth, strongly suggesting that enamel is equivalent in the coelacanth and tetrapods. This finding corroborates the previous proposition that true enamel evolved much earlier than the origin of tetrapods.


Subject(s)
Calcium-Binding Proteins/genetics , Evolution, Molecular , Fishes/genetics , Amelogenesis/genetics , Animals , Biological Evolution , Calcification, Physiologic/genetics , Dental Enamel Proteins/genetics , Dentin/chemistry , Phylogeny , Tooth Calcification/genetics , Vertebrates/genetics
12.
Connect Tissue Res ; 55 Suppl 1: 57-61, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25158182

ABSTRACT

Singleton-Merten syndrome (SMS) is a rare disease with a phenotype of dental dysplasia. Currently, the underlying mechanism of this disease is unknown. In order to investigate the functional mechanism of the SMS tooth phenotypes, we isolated dental pulp tissue and established SMS primary pulp cells. These cells exhibited normal morphology and could be maintained in culture. Their ability to express alkaline phosphatase and mineralize was confirmed by in vitro staining. A comparative osteogenesis polymerase chain reaction array analysis was performed revealing 22 genes up-regulated and 8 genes down-regulated greater than 2-fold in SMS versus unaffected pulp cells. Down-regulated genes included ALP, IGF2, TGFBR2 and COL1A1. Collagen type I was reduced in SMS cells as shown by Western blot analysis. Furthermore, matrix metallopeptidase 13 was found to be dramatically increased in SMS pulp cells. Our findings suggest that dentin mineralization is dysregulated in SMS and may contribute to the root phenotype found in this disease.


Subject(s)
Aortic Diseases/genetics , Dental Enamel Hypoplasia/genetics , Dental Pulp/cytology , Metacarpus/abnormalities , Muscular Diseases/genetics , Odontodysplasia/genetics , Osteogenesis/genetics , Osteoporosis/genetics , Tooth Calcification/genetics , Vascular Calcification/genetics , Cell Differentiation/physiology , Cells, Cultured , Extracellular Matrix Proteins/genetics , Humans , Metacarpus/cytology
13.
Gene ; 538(2): 266-72, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24487055

ABSTRACT

MicroRNAs (miRNAs) play an essential role in regulating cell differentiation either by inhibiting mRNA translation or by inducing its degradation. However, the role of miRNAs in odontoblastic cell differentaion is largely unknown. In the present study, we demonstrate that the expression of miR-27 was significantly increased during MDPC-23 odontoblastic cell differentiation. Furthermore, the up-regulation of miR-27 promotes the differentiation of MDPC-23 odontoblastic cells and accelerates mineralization without cell proliferation. In addition, our results of target gene prediction revealed that the mRNA of adenomatous polyposis coli (APC) associated with Wnt/ß-catenin signaling pathway has miR-27 binding site in the its 3' UTR and is suppressed by miR-27. Subsequentially, the down-regulated APC by miR-27 triggered the activation of Wnt/ß-catenin signaling through accumulation of ß-catenin in the nucleus. Our data suggest that miR-27 promotes MDPC-23 odontoblastic cell differentiation by targeting APC and activating Wnt/ß-catenin signaling. Therefore, miR-27 might be considered a critical candidate as an odontoblastic differentiation molecular target for the development of miRNA based therapeutic agents in the dental medicine.


Subject(s)
Adenomatous Polyposis Coli Protein/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Odontoblasts/cytology , Odontoblasts/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Line , Gene Expression , Mice , Odontogenesis/genetics , Odontogenesis/physiology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tooth Calcification/genetics , Tooth Calcification/physiology
14.
J Dent Res ; 93(1): 96-102, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24170372

ABSTRACT

Enamel fluorosis is an irreversible structural enamel defect following exposure to supraoptimal levels of fluoride during amelogenesis. We hypothesized that fluorosis is associated with excess release of protons during formation of hypermineralized lines in the mineralizing enamel matrix. We tested this concept by analyzing fluorotic enamel defects in wild-type mice and mice deficient in anion exchanger-2a,b (Ae2a,b), a transmembrane protein in maturation ameloblasts that exchanges extracellular Cl(-) for bicarbonate. Defects were more pronounced in fluorotic Ae2a,b (-/-) mice than in fluorotic heterozygous or wild-type mice. Phenotypes included a hypermineralized surface, extensive subsurface hypomineralization, and multiple hypermineralized lines in deeper enamel. Mineral content decreased in all fluoride-exposed and Ae2a,b(-/-) mice and was strongly correlated with Cl(-). Exposure of enamel surfaces underlying maturation-stage ameloblasts to pH indicator dyes suggested the presence of diffusion barriers in fluorotic enamel. These results support the concept that fluoride stimulates hypermineralization at the mineralization front. This causes increased release of protons, which ameloblasts respond to by secreting more bicarbonates at the expense of Cl(-) levels in enamel. The fluoride-induced hypermineralized lines may form barriers that impede diffusion of proteins and mineral ions into the subsurface layers, thereby delaying biomineralization and causing retention of enamel matrix proteins.


Subject(s)
Chloride-Bicarbonate Antiporters/drug effects , Fluorides/adverse effects , Fluorosis, Dental/etiology , Ameloblasts/drug effects , Ameloblasts/pathology , Amelogenesis/drug effects , Amelogenesis/genetics , Animals , Bicarbonates/analysis , Chloride-Bicarbonate Antiporters/analysis , Chloride-Bicarbonate Antiporters/genetics , Chlorides/analysis , Coloring Agents , Dental Enamel/chemistry , Dental Enamel/drug effects , Dental Enamel/pathology , Dental Enamel Proteins/analysis , Diffusion , Female , Fluorosis, Dental/genetics , Fluorosis, Dental/pathology , Heterozygote , Homozygote , Hydrogen-Ion Concentration , Indicators and Reagents , Mice , Mice, Knockout , Minerals/analysis , Phenotype , Rats , Rats, Wistar , Tooth Calcification/drug effects , Tooth Calcification/genetics
15.
Eur J Oral Sci ; 121(4): 303-12, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23841781

ABSTRACT

Expression of clusterin (Clu) in the murine first molar tooth germ was markedly increased at postnatal developmental stages. The time-course of expression of this gene paralleled those of other genes encoding proteins involved during the secretory phase of odontogenesis, as described previously. Immunohistochemical studies of clusterin in murine molar tooth germs suggested this protein to be located in outer enamel epithelium, regressing enamel organ, secretory ameloblasts, and the dental epithelium connecting the tooth to the oral epithelium at an early eruptive stage. Immunolabelling of transforming growth factor beta-1 (TGF-ß1) revealed it to be located close to clusterin. The levels of expression of Clu and Tgfb1 were markedly decreased following in-vivo transfection with anti-miR-214. In contrast, the expression of several genes associated with regulation of growth and development were increased by this treatment. We suggest that clusterin has functions during secretory odontogenesis and the early eruptive phase. Bioinformatic analysis after treatment with anti-miR-214 suggested that, whilst cellular activities associated with tooth mineralization and eruption were inhibited, activities associated with an alternative developmental activity (i.e. biosynthesis of contractile proteins) appeared to be stimulated. These changes probably occur through regulation mediated by a common cluster of transcription factors and support suggestions that microRNAs (miRNAs) are highly significant as regulators of differentiation during odontogenesis.


Subject(s)
Clusterin/genetics , Gene Expression Regulation, Developmental , MicroRNAs/antagonists & inhibitors , Molar/growth & development , Odontogenesis/genetics , Tooth Calcification/genetics , Tooth Germ/growth & development , Transforming Growth Factor beta1/genetics , Animals , Animals, Newborn , Clusterin/analysis , Gene Expression Profiling , Immunohistochemistry , Mice , Molar/embryology , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Tooth Germ/embryology , Tooth Germ/metabolism , Transfection , Transforming Growth Factor beta1/analysis
16.
PLoS One ; 8(5): e62645, 2013.
Article in English | MEDLINE | ID: mdl-23675415

ABSTRACT

The aim of the present study was to investigate the effect of Sonic hedgehog (Shh) on human dental pulp cells (hDPCs) and the potential of complexes with Shh gene modified hDPCs and porous calcium phosphate cement (CPC) for mineralized tissue formation. hDPCs were cultured and transfected with adenoviral mediated human Shh gene (AdShh). Overexpression of Shh and cell proliferation was tested by real-time PCR analysis, western blotting analysis, and MTT analysis, respectively. The odontoblastic differentiation was assessed by alkaline phosphatase (ALP) activity and real-time PCR analysis on markers of Patched-1 (Ptc-1), Smoothened (Smo), Gli 1, Gli 2, Gli 3, osteocalcin (OCN), dentin matrix protein-1 (DMP-1), and dentin sialophosphoprotein (DSPP). Finally, AdShh-transfected hDPCs were combined with porous CPC and placed subcutaneously in nude mice for 8 and 12 weeks, while AdEGFP-transfected and untransfected hDPCs were treated as control groups. Results indicated that Shh could promote proliferation and odontoblastic differentiation of hDPCs, while Shh/Gli 1 signaling pathway played a key role in this process. Importantly, more mineralized tissue formation was observed in combination with AdShh transfected hDPCs and porous CPC, moreover, the mineralized tissue exhibited dentin-like features such as structures similar to dentin-pulp complex and the positive staining for DSPP protein similar to the tooth tissue. These results suggested that the constructs with AdShh-transfected hDPCs and porous CPC might be a better alternative for dental tissue regeneration.


Subject(s)
Dental Cementum/metabolism , Dental Pulp/cytology , Dental Pulp/metabolism , Dentin/metabolism , Hedgehog Proteins/genetics , Adenoviridae/genetics , Animals , Calcium Phosphates/chemistry , Cell Adhesion/drug effects , Cell Adhesion/genetics , Cell Differentiation/genetics , Cell Proliferation/drug effects , Dental Cementum/chemistry , Dental Cementum/ultrastructure , Gene Expression , Genetic Vectors/genetics , Guided Tissue Regeneration , Hedgehog Proteins/metabolism , Humans , Mice , Odontoblasts/cytology , Odontoblasts/drug effects , Time Factors , Tooth Calcification/drug effects , Tooth Calcification/genetics , Transduction, Genetic , Transfection , Veratrum Alkaloids/pharmacology
17.
J Dent Res ; 92(8): 721-7, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23694930

ABSTRACT

Phosphatases are involved in bone and tooth mineralization, but their mechanisms of action are not completely understood. Tissue-nonspecific alkaline phosphatase (TNAP, ALPL) regulates inhibitory extracellular pyrophosphate through its pyrophosphatase activity to control mineral propagation in the matrix; mice without TNAP lack acellular cementum, and have mineralization defects in dentin, enamel, and bone. PHOSPHO1 is a phosphatase found within membrane-bounded matrix vesicles in mineralized tissues, and double ablation of Alpl and Phospho1 in mice leads to a complete absence of skeletal mineralization. Here, we describe mineralization abnormalities in the teeth of Phospho1(-/-) mice, and in compound knockout mice lacking Phospho1 and one allele of Alpl (Phospho1(-/-);Alpl(+/-) ). In wild-type mice, PHOSPHO1 and TNAP co-localized to odontoblasts at early stages of dentinogenesis, coincident with the early mineralization of mantle dentin. In Phospho1 knockout mice, radiography, micro-computed tomography, histology, and transmission electron microscopy all demonstrated mineralization abnormalities of incisor dentin, with the most remarkable findings being reduced overall mineralization coincident with decreased matrix vesicle mineralization in the Phospho1(-/-) mice, and the almost complete absence of matrix vesicles in the Phospho1(-/-);Alpl(+/-) mice, whose incisors showed a further reduction in mineralization. Results from this study support prominent non-redundant roles for both PHOSPHO1 and TNAP in dentin mineralization.


Subject(s)
Alkaline Phosphatase/genetics , Dentin/enzymology , Phosphoric Monoester Hydrolases/genetics , Tooth Calcification/genetics , Alleles , Alveolar Process/enzymology , Ameloblasts/enzymology , Animals , Apatites/analysis , Calcification, Physiologic/genetics , Dentinogenesis/genetics , Enamel Organ/enzymology , Extracellular Matrix/enzymology , Immunohistochemistry , Incisor/enzymology , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Mice, Mutant Strains , Microscopy, Electron, Transmission , Molar/enzymology , Odontoblasts/enzymology , Osteoblasts/enzymology , Radiographic Image Enhancement , Tooth Germ/enzymology , X-Ray Microtomography
18.
J Endod ; 39(5): 612-8, 2013 May.
Article in English | MEDLINE | ID: mdl-23611378

ABSTRACT

INTRODUCTION: The molecular mechanisms behind odontoblast differentiation remain obscure. Lymphoid enhancer-binding factor 1 (Lef1) is a transcription factor that mediates Wnt signaling and has been suggested to regulate dentin sialophosphoprotein (Dspp) expression in vitro. This study aimed to clarify their precise relationship in the process of odontoblast differentiation in vivo. METHODS: The detailed spatiotemporal expression patterns of Lef1 and Dspp together with other known and putative odontoblast differentiation markers such as P21 and heat-shock protein 25 (Hsp25) were examined by in situ hybridization and immunohistochemistry on paraffin sections of rat incisors and developing molars at postnatal days 1-100. To observe odontoblast regeneration following tooth injury, a cavity was prepared on the upper first molar of 10-week-old rats and the expressions of Lef1 and Dspp were investigated. RESULTS: Following undifferentiated state expressing none of these examined markers, preodontoblasts begun to express P21, Lef1 and Hsp25 according to their progress of differentiation, although Dspp was undetectable. Immature odontoblasts commenced transcribing Dspp simultaneously with dentin calcification. Lef1, Dspp and Hsp25 were co-expressed in mature odontoblasts. In contrast to continuously growing incisors, Lef1, Dspp and P21 were down-regulated in the resting odontoblasts in molars when primary dentin formation was completed. Remarkably, Lef1 expression also preceded Dspp expression in newly differentiated odontoblast-like cells during the pulpal healing process after tooth injury. CONCLUSIONS: Lef1 expression precedes Dspp expression without exception in both primary and reparative dentinogeneses. Our results suggest that Lef1 might play a key role in odontoblast differentiation through regulating Dspp expression.


Subject(s)
Extracellular Matrix Proteins/genetics , Lymphoid Enhancer-Binding Factor 1/genetics , Odontoblasts/physiology , Phosphoproteins/genetics , Sialoglycoproteins/genetics , Animals , Cell Differentiation/genetics , Cyclin-Dependent Kinase Inhibitor p21/genetics , Dentin/growth & development , Dentinogenesis/genetics , HSP27 Heat-Shock Proteins/genetics , Incisor/growth & development , Molar/cytology , Molar/growth & development , Molar/injuries , Protein Kinase Inhibitors/analysis , Rats , Rats, Wistar , Regeneration/genetics , Tooth Calcification/genetics , Transcription, Genetic/genetics , Wnt Signaling Pathway/genetics
19.
Eur J Oral Sci ; 121(2): 76-85, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23489896

ABSTRACT

Dentin sialophosphoprotein (DSPP) is a large precursor protein that is proteolytically processed into a NH2 -terminal fragment [composed of dentin sialoprotein (DSP) and a proteoglycan form (DSP-PG)] and a COOH-terminal fragment [dentin phosphoprotein (DPP)]. In vitro studies indicate that DPP is a strong initiator and regulator of hydroxyapatite crystal formation and growth, but the role(s) of the NH2 -terminal fragment of DSPP (i.e., DSP and DSP-PG) in dentinogenesis remain unclear. This study focuses on the function of the NH2 -terminal fragment of DSPP in dentinogenesis. Here, transgenic (Tg) mouse lines expressing the NH2 -terminal fragment of DSPP driven by a 3.6-kb type I collagen promoter (Col 1a1) were generated and cross-bred with Dspp null mice to obtain mice that express the transgene but lack the endogenous Dspp (Dspp KO/DSP Tg). We found that dentin from the Dspp KO/DSP Tg mice was much thinner, more poorly mineralized, and remarkably disorganized compared with dentin from the Dspp KO mice. The fact that Dspp KO/DSP Tg mice exhibited more severe dentin defects than did the Dspp null mice indicates that the NH2 -terminal fragment of DSPP may inhibit dentin mineralization or may serve as an antagonist against the accelerating action of DPP and serve to prevent predentin from being mineralized too rapidly during dentinogenesis.


Subject(s)
Dentin/metabolism , Dentinogenesis/physiology , Extracellular Matrix Proteins/metabolism , Phosphoproteins/metabolism , Sialoglycoproteins/metabolism , Tooth Calcification/physiology , Animals , Dentin/chemistry , Dentinogenesis/genetics , Extracellular Matrix Proteins/chemistry , Extracellular Matrix Proteins/genetics , Immunohistochemistry , Mice , Mice, Knockout , Microscopy, Electron, Scanning , Phosphoproteins/chemistry , Phosphoproteins/genetics , RNA, Messenger , Real-Time Polymerase Chain Reaction , Sialoglycoproteins/chemistry , Sialoglycoproteins/genetics , Tooth Calcification/genetics , X-Ray Microtomography
20.
J Dent Res ; 92(4): 358-64, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23428435

ABSTRACT

Sphingomyelin phosphodiesterase 3 (Smpd3) encodes a membrane-bound enzyme that cleaves sphingomyelin to generate several bioactive metabolites. A recessive mutation called fragilitas ossium (fro) in the Smpd3 gene leads to impaired mineralization of bone and tooth extracellular matrix (ECM) in fro/fro mice. In teeth from fro/fro mice at various neonatal ages, radiography and light and electron microscopy showed delayed mantle dentin mineralization and a consequent delay in enamel formation as compared with that in control +/fro mice. These tooth abnormalities progressively improved with time. Immunohistochemistry showed expression of SMPD3 by dentin-forming odontoblasts. SMPD3 deficiency, however, did not affect the differentiation of these cells, as shown by osterix and dentin sialophosphoprotein expression. Using a transgenic mouse rescue model (fro/fro; Col1a1-Smpd3) in which Smpd3 expression is driven by a murine Col1a1 promoter fragment active in osteoblasts and odontoblasts, we demonstrate a complete correction of the tooth mineralization delays. In conclusion, analysis of these data demonstrates that Smpd3 expression in odontoblasts is required for tooth mineralization.


Subject(s)
Dental Enamel/enzymology , Osteogenesis Imperfecta/enzymology , Sphingomyelin Phosphodiesterase/metabolism , Tooth Calcification/physiology , Animals , Dentin/enzymology , Disease Models, Animal , Extracellular Matrix/enzymology , Mice , Mice, Mutant Strains , Mice, Transgenic , Odontoblasts/enzymology , Osteogenesis Imperfecta/genetics , Sphingomyelin Phosphodiesterase/genetics , Tooth Calcification/genetics
SELECTION OF CITATIONS
SEARCH DETAIL