Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 100
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Dent Res ; 103(6): 662-671, 2024 06.
Article in English | MEDLINE | ID: mdl-38716742

ABSTRACT

Amelogenesis imperfecta (AI) is a diverse group of inherited diseases featured by various presentations of enamel malformations that are caused by disturbances at different stages of enamel formation. While hypoplastic AI suggests a thickness defect of enamel resulting from aberrations during the secretory stage of amelogenesis, hypomaturation AI indicates a deficiency of enamel mineralization and hardness established at the maturation stage. Mutations in ENAM, which encodes the largest enamel matrix protein, enamelin, have been demonstrated to cause generalized or local hypoplastic AI. Here, we characterized 2 AI families with disparate hypoplastic and hypomaturation enamel defects and identified 2 distinct indel mutations at the same location of ENAM, c588+1del and c.588+1dup. Minigene splicing assays demonstrated that they caused frameshifts and truncation of ENAM proteins, p.Asn197Ilefs*81 and p.Asn197Glufs*25, respectively. In situ hybridization of Enam on mouse mandibular incisors confirmed its restricted expression in secretory stage ameloblasts and suggested an indirect pathogenic mechanism underlying hypomaturation AI. In silico analyses indicated that these 2 truncated ENAMs might form amyloid structures and cause protein aggregation with themselves and with wild-type protein through the added aberrant region at their C-termini. Consistently, protein secretion assays demonstrated that the truncated proteins cannot be properly secreted and impede secretion of wild-type ENAM. Moreover, compared to the wild-type, overexpression of the mutant proteins significantly increased endoplasmic reticulum stress and upregulated the expression of unfolded protein response (UPR)-related genes and TNFRSF10B, a UPR-controlled proapoptotic gene. Caspase, terminal deoxynucleotidyl transferase UTP nick-end labeling (TUNEL), and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) assays further revealed that both truncated proteins, especially p.Asn197Ilefs*81, induced cell apoptosis and decreased cell survival, suggesting that the 2 ENAM mutations cause AI through ameloblast cell pathology and death rather than through a simple loss of function. This study demonstrates that an ENAM mutation can lead to generalized hypomaturation enamel defects and suggests proteinopathy as a potential pathogenesis for ENAM-associated AI.


Subject(s)
Amelogenesis Imperfecta , Animals , Female , Humans , Male , Mice , Ameloblasts/pathology , Amelogenesis Imperfecta/genetics , Apoptosis/genetics , Dental Enamel Proteins/genetics , Extracellular Matrix Proteins , In Situ Hybridization , Mutation , Pedigree
2.
J Dent Res ; 101(1): 37-45, 2022 01.
Article in English | MEDLINE | ID: mdl-34036831

ABSTRACT

Amelogenesis imperfecta (AI) is an innate disorder that affects the formation and mineralization of the tooth enamel. When diagnosed with AI, one's teeth can be hypoplastic (thin enamel), hypomature (normal enamel thickness but discolored and softer than normal enamel), hypocalcified (normal enamel thickness but extremely weak), or mixed conditions of the above. Numerous studies have revealed the genes that are involved in causing AI. Recently, ACP4 (acid phosphatase 4) was newly found as a gene causing hypoplastic AI, and it was suggested that mutant forms of ACP4 might affect access to the catalytic core or the ability to form a homodimer. In this study, a Korean and a Turkish family with hypoplastic AI were recruited, and their exome sequences were analyzed. Biallelic mutations were revealed in ACP4: paternal (NM_033068: c.419C>T, p.(Pro140Leu)) and maternal (c.262C>A, p.(Arg88Ser)) mutations in family 1 and a paternal (c.713C>T, p.(Ser238Leu)) mutation and de novo (c.350A>G, p.(Gln117Arg)) mutation in the maternal allele in family 2. Mutations were analyzed by cloning, mutagenesis, immunofluorescence, immunoprecipitation, and acid phosphatase activity test. Comparison between the wild-type and mutant ACP4s showed a decreased amount of protein expression from the mutant forms, a decreased ability to form a homodimer, and a decreased acid phosphatase activity level. We believe that these findings will not only expand the mutational spectrum of ACP4 but also increase our understanding of the mechanism of ACP4 function during normal and pathologic amelogenesis.


Subject(s)
Acid Phosphatase/genetics , Amelogenesis Imperfecta , Tooth , Amelogenesis Imperfecta/genetics , Dental Enamel , Humans , Mutation/genetics , Pedigree
3.
Cells Tissues Organs ; 194(2-4): 188-93, 2011.
Article in English | MEDLINE | ID: mdl-21576914

ABSTRACT

N-terminal and C-terminal (CT) domains of amelogenin have been shown to be essential for proper enamel formation. Recent studies have also suggested that although the C-terminus plays an apparent role in protein-mineral interactions, other amelogenin structural domains are involved. The objective was to explore the role of the amelogenin N-terminus in the regulation of calcium phosphate formation in vitro. Spontaneous mineralization studies were carried out using the phosphorylated (+P) and nonphosphorylated (-P) N-terminus of the leucine-rich amelogenin peptide (LRAP) that lacks the hydrophilic CT domain. Mineralization progress was monitored via changes in solution pH. Mineral phases formed were characterized using TEM, selected area electron diffraction, and FT-IR. In controls, amorphous calcium phosphate was initially formed and subsequently transformed to randomly oriented hydroxyapatite (HA) plate-like crystals. In contrast to the control, LRAP(+P)-CT stabilized ACP formation for >1 day, while LRAP(-P)-CT accelerated the transformation of ACP to HA but had little effect on crystal shape or orientation. In conclusion, the N-terminal domain found in LRAP, as in amelogenins, appears to have the capacity to interact with forming calcium phosphate mineral phases. Results suggest that the N-terminal domain of amelogenin may play a direct role in early stages of enamel formation.


Subject(s)
Amelogenin/chemistry , Amelogenin/metabolism , Calcium Phosphates/metabolism , Amelogenin/ultrastructure , Amino Acid Sequence , Animals , Calcification, Physiologic/physiology , Dental Enamel Proteins/chemistry , Dental Enamel Proteins/ultrastructure , Hydrogen-Ion Concentration , Molecular Sequence Data , Spectroscopy, Fourier Transform Infrared , Structure-Activity Relationship , Sus scrofa , Time Factors
4.
J Dent Res ; 100(3): 293-301, 2021 03.
Article in English | MEDLINE | ID: mdl-33034243

ABSTRACT

Autosomal dominant hypocalcified amelogenesis imperfecta (ADHCAI; OMIM #130900) is a genetic disorder exhibiting severe hardness defects and reduced fracture toughness of dental enamel. While the condition is nonsyndromic, it can be associated with other craniofacial anomalies, such as malocclusions and delayed or failed tooth eruption. Truncation mutations in FAM83H (OMIM *611927) are hitherto the sole cause of ADHCAI. With human genetic studies, Fam83h knockout and mutation-knock-in mouse models indicated that FAM83H does not serve a critical physiologic function during enamel formation and suggested a neomorphic mutation mechanism causing ADHCAI. The function of FAM83H remains obscure. FAM83H has been shown to interact with various isoforms of casein kinase 1 (CK1) and keratins and to mediate organization of keratin cytoskeletons and desmosomes. By considering FAM83H a scaffold protein to anchor CK1s, further molecular characterization of the protein could gain insight into its functions. In this study, we characterized 9 kindreds with ADHCAI and identified 3 novel FAM83H truncation mutations: p.His437*, p.Gln459*, and p.Glu610*. Some affected individuals exhibited hypoplastic phenotypes, in addition to the characteristic hypocalcification enamel defects, which have never been well documented. Failed eruption of canines or second molars in affected persons was observed in 4 of the families. The p.Glu610* mutation was located in a gap area (amino acids 470 to 625) within the zone of previously reported pathogenic variants (amino acids 287 to 694). In vitro pull-down studies with overexpressed FAM83H proteins in HEK293 cells demonstrated an interaction between FAM83H and SEC16A, a protein component of the COP II complex at endoplasmic reticulum exit sites. The interaction was mediated by the middle part (amino acids 287 to 657) of mouse FAM83H protein. Results of this study significantly extended the phenotypic and genotypic spectrums of FAM83H-associated ADHCAI and suggested a role for FAM83H in endoplasmic reticulum-to-Golgi vesicle trafficking and protein secretion (dbGaP phs001491.v1.p1).


Subject(s)
Amelogenesis Imperfecta , Amelogenesis Imperfecta/genetics , Endoplasmic Reticulum/genetics , Golgi Apparatus , HEK293 Cells , Humans , Proteins , Vesicular Transport Proteins
5.
J Periodontal Res ; 45(3): 353-60, 2010 Jun.
Article in English | MEDLINE | ID: mdl-19909398

ABSTRACT

BACKGROUND AND OBJECTIVE: Emdogain (EMD), consisting mostly of amelogenin, is used in periodontal therapy to regenerate lost connective tissue. Emdogain is applied onto periodontally affected root surfaces, where it becomes exposed to proteolytic enzymes. In this study, we aimed to find out whether gingival crevicular fluid or matrix metalloproteinases (MMPs) could degrade EMD, and whether this degradation has consequences for in vitro cell proliferation. MATERIAL AND METHODS: We studied the effects of 156 gingival crevicular fluid samples collected from subjects with different stages of periodontal disease and from healthy control subjects and the effects of MMP-1, -2, -8, -9, -13 and -14 on the degradation of EMD using EMD-embedded zymography. The effects of gingival crevicular fluid with or without EMD and the effects of amelogenin on the proliferation of cultured periodontal ligament fibroblasts were studied by cell proliferation enzyme-linked immunosorbent assay kit. RESULTS: Degradation of Emdogain induced by gingival crevicular fluid was greater in samples from all stages of periodontal diseases compared with healthy control samples. Of the MMPs studied, only MMP-2 and MMP-8 showed limited EMD-degrading activities. One hundred micrograms per millilitre of EMD increased proliferation of periodontal ligament fibroblasts on average by 24% (confidence interval 0.60-0.64) and at 200 microg/mL by 30% (confidence interval 0.62-0.68) compared with control fibroblasts (confidence interval 0.48-0.52). However, gingival crevicular fluid (10 microg/mL) together with 100 microg/mL EMD induced the proliferation only by 6% (confidence interval 0.51-0.55) and with 200 microg/mL EMD by 12% (confidence interval 0.54-0.58). Amelogenin at 200 microg/mL decreased the proliferation of periodontal ligament fibroblasts by 54% (confidence interval 0.22-0.25). CONCLUSION: We suggest that diseased gingival crevicular fluid containing various proteases leads to degradation of EMD and decreased proliferation of periodontal ligament fibroblasts.


Subject(s)
Dental Enamel Proteins/metabolism , Fibroblasts/drug effects , Gingival Crevicular Fluid/metabolism , Periodontal Ligament/drug effects , Adolescent , Adult , Aggressive Periodontitis/metabolism , Alveolar Bone Loss/metabolism , Amelogenin/metabolism , Amelogenin/pharmacology , Cell Culture Techniques , Cell Proliferation/drug effects , Chronic Periodontitis/metabolism , Dental Enamel Proteins/pharmacology , Female , Fibroblasts/cytology , Gingival Crevicular Fluid/enzymology , Gingival Hemorrhage/metabolism , Gingivitis/metabolism , Humans , Male , Matrix Metalloproteinase 1/pharmacology , Matrix Metalloproteinase 13/pharmacology , Matrix Metalloproteinase 14/pharmacology , Matrix Metalloproteinase 2/pharmacology , Matrix Metalloproteinase 8/pharmacology , Matrix Metalloproteinase 9/pharmacology , Middle Aged , Periodontal Attachment Loss/metabolism , Periodontal Diseases/metabolism , Periodontal Ligament/cytology , Periodontal Pocket/metabolism , Young Adult
6.
J Dent Res ; 99(4): 410-418, 2020 04.
Article in English | MEDLINE | ID: mdl-31999931

ABSTRACT

Amelogenesis imperfecta (AI) is a collection of genetic disorders affecting the quality and/or quantity of tooth enamel. More than 20 genes are, so far, known to be responsible for this condition. In this study, we recruited 3 Turkish families with hypomaturation AI. Whole-exome sequence analyses identified disease-causing mutations in each proband, and these mutations cosegregated with the AI phenotype in all recruited members of each family. The AI-causing mutations in family 1 were a novel AMELX mutation [NM_182680.1:c.143T>C, p.(Leu48Ser)] in the proband and a novel homozygous MMP20 mutation [NM_004771.3:c.616G>A, p.(Asp206Asn)] in the mother of the proband. Previously reported compound heterozygous MMP20 mutations [NM_004771.3:c.103A>C, p.(Arg35=) and c.389C>T, p.(Thr130Ile)] caused the AI in family 2 and family 3. Minigene splicing analyses revealed that the AMELX missense mutation increased exonic definition of exon 4 and the MMP20 synonymous mutation decreased exonic definition of exon 1. These mutations would trigger an alteration of exon usage during RNA splicing, causing the enamel malformations. These results broaden our understanding of molecular genetic pathology of tooth enamel formation.


Subject(s)
Amelogenesis Imperfecta , Amelogenesis Imperfecta/genetics , Dental Enamel , Exons/genetics , Humans , Mutation , Pedigree
7.
J Dent Res ; 98(5): 541-548, 2019 05.
Article in English | MEDLINE | ID: mdl-30779877

ABSTRACT

Dental enamel malformations, or amelogenesis imperfecta (AI), can be isolated or syndromic. To improve the prospects of making a successful diagnosis by genetic testing, it is important that the full range of genes and mutations that cause AI be determined. Defects in WDR72 (WD repeat-containing protein 72; OMIM *613214) cause AI, type IIA3 (OMIM #613211), which follows an autosomal recessive pattern of inheritance. The defective enamel is normal in thickness, severely hypomineralized, orange-brown stained, and susceptible to attrition. We identified 6 families with biallelic WDR72 mutations by whole exome sequence analyses that perfectly segregated with the enamel phenotype. The novel mutations included 3 stop-gains [NM_182758.2: c.377G>A/p.(Trp126*), c.1801C>T/p.(Arg601*), c.2350A>T/p.(Arg784*)], a missense mutation [c.1265G>T/p.(Gly422Val)], and a 62,138-base pair deletion (NG_017034.2: g.35441_97578del62138) that removed WDR72 coding exons 3 through 13. A previously reported WDR72 frameshift was also observed [c.1467_1468delAT/p.(Val491Aspfs*8)]. Three of the affected patients showed decreased serum pH, consistent with a diagnosis of renal tubular acidosis. Percentiles of stature and body weight varied among 8 affected individuals but did not show a consistent trend. These studies support that WDR72 mutations cause a syndromic form of AI and improve our ability to diagnose AI caused by WDR72 defects.


Subject(s)
Acidosis , Amelogenesis Imperfecta , Proteins/immunology , Acidosis/genetics , Amelogenesis Imperfecta/genetics , Humans , Mutation , Pedigree
8.
J Dent Res ; 87(1): 56-9, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18096894

ABSTRACT

Proteolytic enzymes are necessary for the mineralization of dental enamel during development, and mutations in the kallikrein 4 (KLK4) and enamelysin (MMP20) genes cause autosomal-recessive amelogenesis imperfecta (ARAI). So far, only one KLK4 and two MMP20 mutations have been reported. We have identified an ARAI-causing point mutation (c.102G>A, g.102G>A, and p.W34X) in exon 1 of MMP20 in a proband with autosomal-recessive hypoplastic-hypomaturation amelogenesis imperfecta. The G to A transition changes the tryptophan (W) codon (TGG) at amino acid position 34 into a translation termination (X) codon (TGA). No disease-causing sequence variations were detected in KLK4. The affected enamel is thin, with mild spacing in the anterior dentition. The enamel layer is hypomineralized, does not contrast with dentin on radiographs, and tends to chip away from the underlying dentin. An intrinsic yellowish pigmentation is evident, even during eruption. The phenotype supports current ideas concerning the function of enamelysin.


Subject(s)
Amelogenesis Imperfecta/genetics , Codon, Nonsense/genetics , Matrix Metalloproteinase 20/genetics , Point Mutation/genetics , Adenine , Child , Dental Enamel/pathology , Exons/genetics , Female , Guanine , Humans , Sequence Analysis, DNA , Tryptophan/genetics
9.
J Dent Res ; 97(9): 1064-1069, 2018 08.
Article in English | MEDLINE | ID: mdl-29554435

ABSTRACT

Tooth enamel, the hardest tissue in the human body, is formed after a complex series of interactions between dental epithelial tissue and the underlying ectomesenchyme. Nonsyndromic amelogenesis imperfecta (AI) is a rare genetic disorder affecting tooth enamel without other nonoral symptoms. In this study, we identified 2 novel ENAM mutations in 2 families with hypoplastic AI by whole exome sequencing. Family 1 had a heterozygous splicing donor site mutation in intron 4, NM_031889; c.123+2T>G. Affected individuals had hypoplastic enamel with or without the characteristic horizontal hypoplastic grooves in some teeth. Family 2 had a nonsense mutation in the last exon, c.1842C>G, p.(Tyr614*), that was predicted to truncate the protein by 500 amino acids. Participating individuals had at least 1 mutant allele, while the proband had a homozygous mutation. Most interestingly, the clinical phenotype of the individuals harboring the heterozygous mutation varied from a lack of penetrance to a mild hypoplastic enamel defect. We believe that these findings will broaden our understanding of the clinical phenotype of AI caused by ENAM mutations.


Subject(s)
Amelogenesis Imperfecta/genetics , Extracellular Matrix Proteins/genetics , Mutation/genetics , Child , Consanguinity , Female , Humans , Pedigree , Phenotype , Polymerase Chain Reaction , Turkey , Exome Sequencing , Young Adult
10.
J Dent Res ; 86(5): 392-9, 2007 May.
Article in English | MEDLINE | ID: mdl-17452557

ABSTRACT

By the Shields classification, articulated over 30 years ago, inherited dentin defects are divided into 5 types: 3 types of dentinogenesis imperfecta (DGI), and 2 types of dentin dysplasia (DD). DGI type I is osteogenesis imperfecta (OI) with DGI. OI with DGI is caused, in most cases, by mutations in the 2 genes encoding type I collagen. Many genes are required to generate the enzymes that catalyze collagen's diverse post-translational modifications and its assembly into fibers, fibrils, bundles, and networks. Rare inherited diseases of bone are caused by defects in these genes, and some are occasionally found to include DGI as a feature. Appreciation of the complicated genetic etiology of DGI associated with bony defects splintered the DGI type I description into a multitude of more precisely defined entities, all with their own designations. In contrast, DD-II, DGI-II, and DGI-III, each with its own pattern of inherited defects limited to the dentition, have been found to be caused by various defects in DSPP (dentin sialophosphoprotein), a gene encoding the major non-collagenous proteins of dentin. Only DD-I, an exceedingly rare condition featuring short, blunt roots with obliterated pulp chambers, remains untouched by the revolution in genetics, and its etiology is still a mystery. A major surprise in the characterization of genes underlying inherited dentin defects is the apparent lack of roles played by the genes encoding the less-abundant non-collagenous proteins in dentin, such as dentin matrix protein 1 (DMP1), integrin-binding sialoprotein (IBSP), matrix extracellular phosphoglycoprotein (MEPE), and secreted phosphoprotein-1, or osteopontin (SPP1, OPN). This review discusses the development of the dentin extracellular matrix in the context of its evolution, and discusses the phenotypes and clinical classifications of isolated hereditary defects of tooth dentin in the context of recent genetic data respecting their genetic etiologies.


Subject(s)
Dentin Dysplasia/genetics , Dentinogenesis Imperfecta/genetics , Animals , Chromosomes, Human, Pair 4 , Collagen Type I/genetics , Dentin/abnormalities , Dentin Dysplasia/classification , Dentin Dysplasia/pathology , Dentinogenesis Imperfecta/classification , Dentinogenesis Imperfecta/pathology , Extracellular Matrix/genetics , Extracellular Matrix Proteins/genetics , Humans , Mutation , Phosphoproteins , Sialoglycoproteins
11.
J Dent Res ; 86(10): 962-7, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17890672

ABSTRACT

In developing porcine enamel, the space between enamel rods selectively binds lectins and ameloblastin (Ambn) N-terminal antibodies. We tested the hypothesis that ameloblastin N-terminal cleavage products are glycosylated. Assorted Ambn cleavage products showed positive lectin staining by peanut agglutinin (PNA), Maclura pomifera agglutinin (MPA), and Limulus polyphemus agglutinin (LPA), suggesting the presence of an O-linked glycosylation containing galactose (Gal), N-acetylgalactosamine (GalNAc), and sialic acid. Edman sequencing of the lectin-positive bands gave the Ambn N-terminal sequence: VPAFPRQPGTXGVASLXLE. The blank cycles for Pro(11) and Ser(17) confirmed that these residues are hydroxylated and phosphorylated, respectively. The O-glycosylation site was determined by Edman sequencing of pronase-digested Ambn, which gave HPPPLPXQPS, indicating that Ser(86) is the site of the O-linked glycosylation. This modification is within the 15-amino-acid segment (73-YEYSLPVHPPPLPSQ-87) deleted by splicing in the mRNA encoding the 380-amino-acid Ambn isoform. We conclude that only the N-terminal Ambn products derived from the 395-Ambn isoform are glycosylated.


Subject(s)
Dental Enamel Proteins/genetics , Dental Enamel Proteins/metabolism , Amino Acid Sequence , Animals , Blotting, Western , Glycosylation , Lectins/metabolism , Peptide Fragments/metabolism , Protein Isoforms , Protein Processing, Post-Translational , RNA Splicing , Swine
12.
J Dent Res ; 86(2): 153-7, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17251515

ABSTRACT

Ameloblastin (AMBN) cleavage products are the most abundant non-amelogenin proteins in the enamel matrix of developing teeth. AMBN N-terminal cleavage products accumulate in the sheath space between enamel rods, while AMBN C-terminal products localize within rods. We tested the hypothesis that MMP-20 is the protease that cleaves AMBN. Glycosylated recombinant porcine AMBN (rpAMBN) was expressed in human kidney 293F cells, and recombinant porcine enamelysin (rpMMP-20) was expressed in bacteria. The purified proteins were incubated together at an enzyme:substrate ratio of 1:100. N-terminal sequencing of AMBN digestion products determined that rpMMP-20 cleaved rpAMBN after Pro(2), Gln(130), Gln(139), Arg(170), and Ala(222). This shows that MMP-20 generates the 23-kDa AMBN starting at Tyr(223), as well as the 17-kDa (Val(1)-Arg(170)) and 15-kDa (Val(1)-Gln(130)) AMBN cleavage products that concentrate in the sheath space during the secretory stage. We conclude that MMP-20 processes ameloblastin in vitro and in vivo.


Subject(s)
Amelogenesis/physiology , Dental Enamel Proteins/metabolism , Matrix Metalloproteinase 20/metabolism , Amino Acid Sequence , Animals , Blotting, Western , Cell Line , Electrophoresis, Polyacrylamide Gel , Humans , Immunohistochemistry , Recombinant Proteins/metabolism , Sus scrofa
13.
J Dent Res ; 96(5): 524-530, 2017 May.
Article in English | MEDLINE | ID: mdl-28113034

ABSTRACT

We report here a novel biomimetic approach to the regeneration of human enamel. The approach combines the use of inorganic pyrophosphate (PPi) to control the onset and rate of enamel regeneration and the use of leucine-rich amelogenin peptide (LRAP), a nonphosphorylated 56-amino acid alternative splice product of amelogenin, to regulate the shape and orientation of growing enamel crystals. This study builds on our previous findings that show LRAP can effectively guide the formation of ordered arrays of needle-like hydroxyapatite (HA) crystals in vitro and on the known role mineralization inhibitors, like PPi, play in the regulation of mineralized tissue formation. Acid-etched enamel surfaces of extracted human molars, cut perpendicular or parallel to the direction of the enamel rods, were exposed to a PPi-stabilized supersaturated calcium phosphate (CaP) solution containing 0 to 0.06 mg/mL LRAP for 20 h. In the absence of LRAP, PPi inhibition was reversed by the presence of etched enamel surfaces and led to the formation of large, randomly distributed plate-like HA crystals that were weakly attached, regardless of rod orientation. In the presence of 0.04 mg/mL LRAP, however, densely packed mineral layers, comprising bundles of small needle-like HA crystals, formed on etched surfaces that were cut perpendicular to the enamel rods. These crystals were strongly attached, and their arrangement reflected to a significant degree the underlying enamel prism pattern. In contrast, under the same conditions with LRAP, little to no crystal formation was found on enamel surfaces that were cut parallel to the direction of the enamel rods. These results suggest that LRAP preferentially interacts with ab surfaces of mature enamel crystals, inhibiting their directional growth, thus selectively promoting linear growth along the c-axis of enamel crystals. The present findings demonstrate a potential for the development of a new approach to regenerate enamel structure and properties.


Subject(s)
Amelogenesis/drug effects , Biomimetics , Dental Enamel Proteins/pharmacology , Acid Etching, Dental , Animals , Calcium Phosphates/pharmacology , Crystallization , Humans , Hydrogen-Ion Concentration , In Vitro Techniques , Microscopy, Electron, Scanning , Surface Properties , Swine
14.
J Dent Res ; 85(10): 894-9, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16998127

ABSTRACT

Amelogenin RNA transcripts undergo extensive alternative splicing, and MMP-20 processes the isoforms following their secretion. Since amelogenins have been ascribed cell-signaling activities, we asked if a lack of proteolytic processing by MMP-20 affects amelogenin signaling and consequently alters amelogenin splice site selection. RT-PCR analyses of amelogenin mRNA between control and Mmp20(-/-)mice revealed no differences in the splicing pattern. We characterized 3 previously unidentified amelogenin alternatively spliced transcripts and demonstrated that exon-8-encoded amelogenin isoforms are processed by MMP-20. Transcripts with exon 8 were expressed approximately five-fold less than those with exon 7. Analyses of the mouse and rat amelogenin gene structures confirmed that exon 8 arose in a duplication of exons 4 through 5, with translocation of the copy downstream of exon 7. No downstream genomic sequences homologous to exons 4-5 were present in the bovine or human amelogenin genes, suggesting that this translocation occurred only in rodents.


Subject(s)
Alternative Splicing/physiology , Amelogenin/metabolism , Gene Expression Regulation, Developmental/physiology , Matrix Metalloproteinase 20/metabolism , RNA, Messenger/metabolism , Alternative Splicing/genetics , Amelogenin/genetics , Animals , Base Sequence , Dental Enamel/enzymology , Dental Enamel/metabolism , Exons/physiology , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Matrix Metalloproteinase 20/genetics , Mice , Mice, Knockout , Molar/enzymology , Molar/metabolism , Molecular Sequence Data , Protein Isoforms , Signal Transduction/genetics
15.
J Dent Res ; 85(3): 267-71, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16498076

ABSTRACT

Can kindreds with tooth agenesis caused by MSX1 or PAX9 mutations be distinguished by their phenotypes? We have identified an MSX1second bicuspids and mandibular central incisors. The dominant phenotype is apparently due to haploinsufficiency. We analyzed patterns of partial tooth agenesis in seven kindreds with defined MSX1 mutations and ten kindreds with defined PAX9 mutations. The probability of missing a particular type of tooth is always bilaterally symmetrical, but differences exist between the maxilla and mandible. MSX1-associated oligodontia typically includes missing maxillary and mandibular second bicuspids and maxillary first bicuspids. The most distinguishing feature of MSX1-associated oligodontia is the frequent (75%) absence of maxillary first bicuspids, while the most distinguishing feature of PAX9-associated oligodontia is the frequent (> 80%) absence of the maxillary and mandibular second molars.


Subject(s)
Anodontia/genetics , Bicuspid/abnormalities , MSX1 Transcription Factor/genetics , Chi-Square Distribution , Child , Child, Preschool , DNA Mutational Analysis , Female , Frameshift Mutation , Genes, Dominant , Humans , Male , Molar/abnormalities , PAX9 Transcription Factor/genetics , Pedigree
16.
J Dent Res ; 85(4): 329-33, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16567553

ABSTRACT

Dentinogenesis imperfecta (DGI) and dentin dysplasia (DD) are allelic disorders that primarily affect the formation of tooth dentin. Both conditions are autosomal-dominant and can be caused by mutations in the dentin sialophosphoprotein gene (DSPP, 4q21.3). We recruited 23 members of a four-generation kindred, including ten persons with dentin defects, and tested the hypothesis that these defects are linked to DSPP. The primary dentition showed amber discoloration, pulp obliteration, and severe attrition. The secondary dentition showed either pulp obliteration with bulbous crowns and gray discoloration or thistle-tube pulp configurations, normal crowns, and mild gray discoloration. Haplotype analyses showed no recombination between three 4q21-q24 markers and the disease locus. Mutational analyses identified no coding or intron junction sequence variations associated with affection status in DMP1, MEPE, or the DSP portion of DSPP. The defects in the permanent dentition were typically mild and consistent with a diagnosis of DD-II, but some dental features associated with DGI-II were also present. We conclude that DD-II and DGI-II are milder and more severe forms, respectively, of the same disease.


Subject(s)
Chromosomes, Human, Pair 4/genetics , Dentin Dysplasia/genetics , Dentin/physiopathology , Dentinogenesis Imperfecta/genetics , Extracellular Matrix Proteins/genetics , Adult , Aged , DNA Mutational Analysis , Dentin Dysplasia/classification , Dentin Dysplasia/physiopathology , Dentinogenesis Imperfecta/classification , Dentinogenesis Imperfecta/physiopathology , Dentition, Permanent , Female , Genetic Linkage , Glycoproteins/genetics , Humans , Male , Middle Aged , Pedigree , Phenotype , Phosphoproteins/genetics , Severity of Illness Index , Sialoglycoproteins/genetics
17.
J Dent Res ; 95(13): 1511-1517, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27558264

ABSTRACT

Recent studies have shown that native phosphorylated full-length porcine amelogenin (P173) and its predominant cleavage product (P148) can inhibit spontaneous calcium phosphate formation in vitro by stabilizing an amorphous calcium phosphate (ACP) precursor phase. Since full-length amelogenin undergoes proteolysis by matrix metalloproteinase 20 (MMP20, enamelysin) soon after secretion, the present study was conducted to assess the effect of amelogenin proteolysis on calcium phosphate formation. Calcium and phosphate were sequentially added to protein solutions without and with added MMP20 (ratio = 200:1) under physiological-like conditions of ionic strength (163 mM) in 50 mM Tris-HCl (pH 7.4) at 37 °C. Protein degradation with time was assessed by gel-electrophoresis, and mineral products formed were characterized by transmission electron microscopy (TEM). MMP20 was found to cleave P173 to primarily generate P148, along with P162, P46-148, and P63/64-148. In sharp contrast, MMP20 did not cleave P148. In addition, the formation of well-aligned bundles of enamel-like hydroxyapatite (HA) crystals was promoted in the presence of P173 with added MMP20, while only ACP particles were seen in the absence of MMP20. Although P148 was found to have a somewhat lower capacity to stabilize ACP and prevent HA formation compared with P173 in the absence of MMP20, essentially no HA formation was observed in the presence of somewhat higher concentrations of P148 regardless of MMP20 addition, due to the lack of observed protein proteolysis. Present findings suggest that ACP transformation to ordered arrays of enamel crystals may be regulated in part by the proteolysis of full-length native amelogenin, while the predominant amelogenin degradation product in developing enamel (e.g., P148) primarily serves to prevent uncontrolled mineral formation during the secretory stage of amelogenesis.


Subject(s)
Amelogenesis/physiology , Matrix Metalloproteinase 20/metabolism , Proteolysis , Amelogenin , Animals , Calcium Phosphates , Electrophoresis, Polyacrylamide Gel , Hydrogen-Ion Concentration , In Vitro Techniques , Microscopy, Electron, Transmission , Phosphorylation , Solutions , Swine
18.
J Dent Res ; 84(2): 144-8, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15668331

ABSTRACT

Amelogenin is the major enamel matrix component in developing teeth. In eutherian mammals, amelogenin is expressed from the X chromosome only, or from both the X and Y chromosomes. Two classes of porcine amelogenin cDNA clones have been characterized, but the chromosomal localization of the gene(s) encoding them is unknown. To determine if there are sex-based differences in the expression of porcine amelogenin, we paired PCR primers for exons 1a, 1b, 7a, and 7b, and amplified enamel organ-derived cDNA separately from porcine males and females. The results show that exons 1a/2a and 7a are always together and can be amplified from both males (XY) and females (XX). Exons 1b/2b and 7b are also always paired, but can be amplified only from females. We conclude that porcine amelogenin is expressed from separate genes on the X and Y chromosomes, and not, as previously proposed, from a single gene with two promoters.


Subject(s)
Dental Enamel Proteins/genetics , Enamel Organ/metabolism , Tooth Germ/metabolism , X Chromosome/metabolism , Y Chromosome/metabolism , Amelogenin , Animals , Chromosome Mapping , Dental Enamel Proteins/metabolism , Exons/genetics , Female , Incisor , Male , Mandible , RNA/analysis , Sex Factors , Swine , X Chromosome/genetics , Y Chromosome/genetics
19.
J Dent Res ; 84(3): 278-82, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15723871

ABSTRACT

To date, 4 unique enamelin gene (ENAM) defects have been identified in kindreds with amelogenesis imperfecta. To improve our understanding of the roles of enamelin in normal enamel formation, and to gain information related to possible genotype/phenotype correlations, we have identified 2 ENAM mutations in kindreds with hypoplastic ADAI, 1 novel (g.4806A>C, IVS6-2A>C) and 1 previously identified (g.8344delG), and have characterized the resulting enamel phenotypes. The IVS6-2A>C mutation caused a severe enamel phenotype in the proband, exhibiting horizontal grooves of severely hypoplastic enamel. The affected mother had several shallow hypoplastic horizontal grooves in the lower anterior teeth. In the case of the g.8344delG mutation, the phenotype was generalized hypoplastic enamel with shallow horizontal grooves in the middle 1/3 of the anterior teeth. In general, mutations in the human enamelin gene cause hypoplastic enamel, often with horizontal grooves, but the severity of the enamel defects is variable, even among individuals with the same mutation.


Subject(s)
Amelogenesis Imperfecta/genetics , Dental Enamel Proteins/genetics , Genes, Dominant/genetics , Mutation/genetics , Adult , Child , Dental Enamel/abnormalities , Dental Enamel/pathology , Dental Enamel Hypoplasia/genetics , Exons/genetics , Female , Humans , Introns/genetics , Male , Phenotype
20.
Gene ; 251(1): 1-8, 2000 Jun 13.
Article in English | MEDLINE | ID: mdl-10863090

ABSTRACT

The human PRSS17 (serine protease 17) gene, which is located on chromosome 19q in a cluster of genes encoding serine proteases, has been variously designated enamel matrix serine proteinase 1 (EMSP1), prostase, KLK4, and KLK-L1. We have cloned and characterized the mouse and human PRSS17 genes. Both have six exons and five introns. The mouse PRSS17 gene sequence is 10134bp; the human sequence is 7115bp. Computer analysis of the mouse PRSS17 gene sequence upstream of the translation initiation codon identified two potential transcription initiation sites, at nucleotides 2878 and 2336. The first nucleotide of the reported mouse PRSS17 cDNA sequence corresponds to position 2352 on the gene, only 16 bases downstream from one of the putative transcription initiation sites. Repetitive DNA sequences from the MSR1 family are found in both the mouse and human PRSS17 genes. Additionally, the human PRSS17 gene contains Tigger2, MER8, and Alu repetitive sequences. Phylogenetic analyses of human and rodent proteases suggest that the PRSS17 protein is not a member of the kallikrein family of serine proteases but that the PRSS17 gene may have originated prior to the divergence of the kallikrein and trypsin families of proteases. To better characterize the timing of PRSS17 expression in developing teeth, we performed in-situ hybridization on postnatal day 3 developing mouse mandibular incisors. PRSS17 mRNA was not detected in secretory stage ameloblasts but could be detected in odontoblasts, while transition-stage and maturation-stage ameloblasts were strongly positive. This pattern supports a role for the PRSS17 protein in the degradation of enamel proteins.


Subject(s)
Incisor/metabolism , Kallikreins , Serine Endopeptidases/genetics , Amino Acid Sequence , Animals , Base Sequence , DNA/chemistry , DNA/genetics , Exons , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , Genes/genetics , Humans , In Situ Hybridization , Incisor/growth & development , Introns , Mice , Molecular Sequence Data , Phylogeny , Promoter Regions, Genetic , Repetitive Sequences, Nucleic Acid , Sequence Alignment , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid
SELECTION OF CITATIONS
SEARCH DETAIL