Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 703
Filter
Add more filters

Publication year range
1.
PLoS Genet ; 20(7): e1011364, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39052671

ABSTRACT

How the dorsal-ventral axis of the vertebrate jaw, particularly the position of tooth initiation site, is established remains a critical and unresolved question. Tooth development starts with the formation of the dental lamina, a localized thickened strip within the maxillary and mandibular epithelium. To identify transcriptional regulatory networks (TRN) controlling the specification of dental lamina from the naïve mandibular epithelium, we utilized Laser Microdissection coupled low-input RNA-seq (LMD-RNA-seq) to profile gene expression of different domains of the mandibular epithelium along the dorsal-ventral axis. We comprehensively identified transcription factors (TFs) and signaling pathways that are differentially expressed along mandibular epithelial domains (including the dental lamina). Specifically, we found that the TFs Sox2 and Tfap2 (Tfap2a/Tfap2b) formed complimentary expression domains along the dorsal-ventral axis of the mandibular epithelium. Interestingly, both classic and novel dental lamina specific TFs-such as Pitx2, Ascl5 and Zfp536-were found to localize near the Sox2:Tfap2a/Tfap2b interface. To explore the functional significance of these domain specific TFs, we next examined loss-of-function mouse models of these domain specific TFs, including the dental lamina specific TF, Pitx2, and the ventral surface ectoderm specific TFs Tfap2a and Tfap2b. We found that disruption of domain specific TFs leads to an upregulation and expansion of the alternative domain's TRN. The importance of this cross-repression is evident by the ectopic expansion of Pitx2 and Sox2 positive dental lamina structure in Tfap2a/Tfap2b ectodermal double knockouts and the emergence of an ectopic tooth in the ventral surface ectoderm. Finally, we uncovered an unappreciated interface of mesenchymal SHH and WNT signaling pathways, at the site of tooth initiation, that were established by the epithelial domain specific TFs including Pitx2 and Tfap2a/Tfap2b. These results uncover a previously unknown molecular mechanism involving cross-repression of domain specific TFs including Pitx2 and Tfap2a/Tfap2b in patterning the dorsal-ventral axis of the mouse mandible, specifically the regulation of tooth initiation site.


Subject(s)
Gene Expression Regulation, Developmental , Homeobox Protein PITX2 , Homeodomain Proteins , Mandible , SOXB1 Transcription Factors , Transcription Factor AP-2 , Transcription Factors , Transcription Factor AP-2/metabolism , Transcription Factor AP-2/genetics , Animals , Mice , Transcription Factors/genetics , Transcription Factors/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , SOXB1 Transcription Factors/metabolism , SOXB1 Transcription Factors/genetics , Mandible/metabolism , Epithelium/metabolism , Odontogenesis/genetics , Tooth/metabolism , Tooth/growth & development , Tooth/embryology , Gene Regulatory Networks , Cell Lineage/genetics , Signal Transduction
2.
Nature ; 547(7662): 209-212, 2017 07 13.
Article in English | MEDLINE | ID: mdl-28678781

ABSTRACT

Despite the wide variety of adaptive modifications in the oral and facial regions of vertebrates, their early oropharyngeal development is considered strictly uniform. It involves sequential formation of the mouth and pharyngeal pouches, with ectoderm outlining the outer surface and endoderm the inner surface, as a rule. At the extreme anterior domain of vertebrate embryos, the ectoderm and endoderm directly juxtapose and initial development of this earliest ecto-endoderm interface, the primary mouth, typically involves ectodermal stomodeal invagination that limits the anterior expansion of the foregut endoderm. Here we present evidence that in embryos of extant non-teleost fishes, oral (stomodeal) formation is preceded by the development of prominent pre-oral gut diverticula (POGD) between the forebrain and roof of the forming mouth. Micro-computed tomography (micro-CT) imaging of bichir, sturgeon and gar embryos revealed that foregut outpocketing at the pre-oral domain begins even before the sequential formation of pharyngeal pouches. The presence of foregut-derived cells in the front of the mouth was further confirmed by in vivo experiments that allowed specific tracing of the early endodermal lining. We show that POGD in sturgeons contribute to the orofacial surface of their larvae, comprising oral teeth, lips, and sensory barbels. To our knowledge, this is the first thorough evidence for endodermal origin of external craniofacial structures in any vertebrate. In bichir and gar embryos, POGD form prominent cranial adhesive organs that are characteristic of the ancient bauplan of free-living chordate larvae. POGD hence seem arguably to be ancestral for all ray-finned fishes, and their topology, pharyngeal-like morphogenesis and gene expression suggest that they are evolutionarily related to the foregut-derived diverticula of early chordate and hemichordate embryos. The formation of POGD might thus represent an ancestral developmental module with deep deuterostome origins.


Subject(s)
Digestive System/embryology , Endoderm/embryology , Fishes/anatomy & histology , Fishes/embryology , Maxillofacial Development , Mouth/embryology , Animals , Fishes/classification , Fishes/genetics , Gene Expression Regulation, Developmental , Larva/genetics , Larva/growth & development , Maxillofacial Development/genetics , Phylogeny , Skull/embryology , Tooth/embryology , X-Ray Microtomography
3.
Development ; 146(3)2019 02 08.
Article in English | MEDLINE | ID: mdl-30737241

ABSTRACT

While many vertebrates have multiple sets of teeth over their lifetime, some, like humans, have just a single set of replacement teeth (diphydonty), while others, like mice, manage with a single set (monophydonty). This diversity raises both evolutionary questions - how did different tooth replacement strategies evolve? - and developmental ones - what mechanisms prevent replacement teeth in animals that have lost them? A new paper in this issue of Development tackles these questions with a molecular analysis of mouse tooth development. We caught up with first author Elena Popa and her supervisor Abigail Tucker, Professor of Development and Evolution at King's College London, to find out more about the work.


Subject(s)
Embryology , Evolution, Molecular , Tooth/embryology , Animals , Antibodies, Monoclonal , Antibodies, Monoclonal, Humanized , Embryology/instrumentation , Embryology/methods , Embryology/trends , Humans , Portraits as Topic , Tooth/cytology
4.
Development ; 146(3)2019 02 08.
Article in English | MEDLINE | ID: mdl-30658984

ABSTRACT

Most mammals have two sets of teeth (diphyodont) - a deciduous dentition replaced by a permanent dentition; however, the mouse possesses only one tooth generation (monophyodont). In diphyodonts, the replacement tooth forms on the lingual side of the first tooth from the successional dental lamina. This lamina expresses the stem/progenitor marker Sox2 and has activated Wnt/ß-catenin signalling at its tip. Although the mouse does not replace its teeth, a transient rudimentary successional dental lamina (RSDL) still forms during development. The mouse RSDL houses Sox2-positive cells, but no Wnt/ß-catenin signalling. Here, we show that stabilising Wnt/ß-catenin signalling in the RSDL in the mouse leads to proliferation of the RSDL and formation of lingually positioned teeth. Although Sox2 has been shown to repress Wnt activity, overexpression of Wnts leads to a downregulation of Sox2, suggesting a negative-feedback loop in the tooth. In the mouse, the first tooth represses the formation of the replacement, and isolation of the RSDL is sufficient to induce formation of a new tooth germ. Our data highlight key mechanisms that may have influenced the evolution of replacement teeth.This article has an associated 'The people behind the papers' interview.


Subject(s)
Cell Proliferation/physiology , SOXB1 Transcription Factors/metabolism , Tooth Germ/embryology , Tooth/embryology , Wnt Signaling Pathway/physiology , Animals , Mice , Mice, Transgenic , SOXB1 Transcription Factors/genetics , Swine , Swine, Miniature , Tooth/cytology , Tooth Germ/cytology
5.
PLoS Biol ; 17(2): e3000064, 2019 02.
Article in English | MEDLINE | ID: mdl-30730874

ABSTRACT

When patterns are set during embryogenesis, it is expected that they are straightly established rather than subsequently modified. The patterning of the three mouse molars is, however, far from straight, likely as a result of mouse evolutionary history. The first-formed tooth signaling centers, called MS and R2, disappear before driving tooth formation and are thought to be vestiges of the premolars found in mouse ancestors. Moreover, the mature signaling center of the first molar (M1) is formed from the fusion of two signaling centers (R2 and early M1). Here, we report that broad activation of Edar expression precedes its spatial restriction to tooth signaling centers. This reveals a hidden two-step patterning process for tooth signaling centers, which was modeled with a single activator-inhibitor pair subject to reaction-diffusion (RD). The study of Edar expression also unveiled successive phases of signaling center formation, erasing, recovering, and fusion. Our model, in which R2 signaling center is not intrinsically defective but erased by the broad activation preceding M1 signaling center formation, predicted the surprising rescue of R2 in Edar mutant mice, where activation is reduced. The importance of this R2-M1 interaction was confirmed by ex vivo cultures showing that R2 is capable of forming a tooth. Finally, by introducing chemotaxis as a secondary process to RD, we recapitulated in silico different conditions in which R2 and M1 centers fuse or not. In conclusion, pattern formation in the mouse molar field relies on basic mechanisms whose dynamics produce embryonic patterns that are plastic objects rather than fixed end points.


Subject(s)
Body Patterning , Edar Receptor/metabolism , Models, Biological , Signal Transduction , Tooth/embryology , Tooth/metabolism , Animals , Chemotaxis , Edar Receptor/genetics , Epithelium/embryology , Epithelium/metabolism , Gene Expression Regulation, Developmental , Hair/embryology , Mice , Mice, Mutant Strains , Tooth Germ/embryology , Tooth Germ/metabolism
6.
Dev Dyn ; 250(7): 1021-1035, 2021 07.
Article in English | MEDLINE | ID: mdl-33452709

ABSTRACT

BACKGROUND: Organs that develop early in life, and are replaced by a larger version as the animal grows, often represent a miniature version of the adult organ. Teeth constituting the first functional dentition in small-sized teleost fish, such as medaka (Oryzias latipes), are examples of such miniature organs. With a dentin cone as small as the size of one human cell, or even smaller, these teeth raise the question how many dentin-producing cells (odontoblasts) are required to build such a tooth, and whether this number can be as little as one. RESULTS: Based on detailed observations with transmission electron microscopy (TEM) and TEM-based 3D-reconstructions, we show that only one mesenchymal cell qualifies as a true odontoblast. A second mesenchymal cell potentially participates in dentin formation, but only at a late stage of tooth development. Moreover, the fate of these cells appears to be specified very early during tooth development. CONCLUSIONS: Our observations indicate that in this system, one single odontoblast fulfills roles normally exerted by a large and communicating cell population. First-generation teeth in medaka thus provide an exciting model to study integration of multiple functions into a single cell.


Subject(s)
Mesenchymal Stem Cells/cytology , Odontogenesis/physiology , Tooth/embryology , Animals , Cell Count , Cell Differentiation , Cell Lineage , Computer Simulation , Embryo, Nonmammalian , Imaging, Three-Dimensional , Mesenchymal Stem Cells/physiology , Mesenchymal Stem Cells/ultrastructure , Miniaturization , Morphogenesis/physiology , Odontoblasts/cytology , Odontoblasts/physiology , Odontoblasts/ultrastructure , Oryzias/embryology , Tooth/growth & development , Tooth/ultrastructure , Tooth Eruption/physiology
7.
Dev Dyn ; 250(12): 1796-1809, 2021 12.
Article in English | MEDLINE | ID: mdl-34091971

ABSTRACT

BACKGROUND: Hand genes are required for the development of the vertebrate jaw, heart, peripheral nervous system, limb, gut, placenta, and decidua. Two Hand paralogues, Hand1 and Hand2, are present in most vertebrates, where they mediate different functions yet overlap in expression. In ray-finned fishes, Hand gene expression and function is only known for the zebrafish, which represents the rare condition of having a single Hand gene, hand2. Here we describe the developmental expression of hand1 and hand2 in the cichlid Copadichromis azureus. RESULTS: hand1 and hand2 are expressed in the cichlid heart, paired fins, pharyngeal arches, peripheral nervous system, gut, and lateral plate mesoderm with different degrees of overlap. CONCLUSIONS: Hand gene expression in the gut, peripheral nervous system, and pharyngeal arches may have already been fixed in the lobe- and ray-finned fish common ancestor. In other embryonic regions, such as paired appendages, hand2 expression was fixed, while hand1 expression diverged in lobe- and ray-finned fish lineages. In the lateral plate mesoderm and arch associated catecholaminergic cells, hand1 and hand2 swapped expression between divergent lineages. Distinct expression of cichlid hand1 and hand2 in the epicardium and myocardium of the developing heart may represent the ancestral pattern for bony fishes.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Cichlids/embryology , Embryonic Development/genetics , Animal Fins/embryology , Animal Fins/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Branchial Region/embryology , Branchial Region/metabolism , Cichlids/genetics , Cichlids/metabolism , Embryo, Nonmammalian , Gene Expression Regulation, Developmental , Heart/embryology , Intestines/embryology , Intestines/metabolism , Mesoderm/embryology , Mesoderm/metabolism , Myocardium/metabolism , Peripheral Nervous System/embryology , Peripheral Nervous System/metabolism , Sequence Homology , Skull/embryology , Skull/metabolism , Tooth/embryology , Tooth/metabolism , Zebrafish/embryology , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
8.
Development ; 145(12)2018 06 26.
Article in English | MEDLINE | ID: mdl-29777011

ABSTRACT

Facial form depends on the precise positioning of cartilage, bone, and tooth fields in the embryonic pharyngeal arches. How complex signaling information is integrated to specify these cell types remains a mystery. We find that modular expression of Forkhead domain transcription factors (Fox proteins) in the zebrafish face arises through integration of Hh, Fgf, Bmp, Edn1 and Jagged-Notch pathways. Whereas loss of C-class Fox proteins results in reduced upper facial cartilages, loss of F-class Fox proteins results in distal jaw truncations and absent midline cartilages and teeth. We show that Fox proteins are required for Sox9a to promote chondrogenic gene expression. Fox proteins are sufficient in neural crest-derived cells for cartilage development, and neural crest-specific misexpression of Fox proteins expands the cartilage domain but inhibits bone. These results support a modular role for Fox proteins in establishing the competency of progenitors to form cartilage and teeth in the face.


Subject(s)
Body Patterning , Cartilage/embryology , Cartilage/metabolism , Forkhead Transcription Factors/metabolism , Tooth/embryology , Tooth/metabolism , Animals , Body Patterning/genetics , Bone and Bones/metabolism , Branchial Region/metabolism , Cell Proliferation/genetics , Cell Survival/genetics , Chondrogenesis/genetics , Face , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Developmental , Mutation/genetics , Neural Crest/cytology , Signal Transduction , Skull/cytology , Zebrafish/embryology , Zebrafish Proteins/metabolism
9.
Subcell Biochem ; 95: 175-196, 2020.
Article in English | MEDLINE | ID: mdl-32297300

ABSTRACT

Explaining how the extensive diversity in form of vertebrate teeth arose in evolution and the mechanisms by which teeth are made during embryogenesis are intertwined questions that can merit from a better understanding of the roles of retinoic acid (RA) in tooth development. Pioneering studies in rodents showed that dietary vitamin A (VA), and eventually RA (one of the major active metabolites of VA), are required for proper tooth formation and that dentin-forming odontoblast cells seem to be especially sensitive to changes in RA levels. Later, rodent studies further indicated that RA signaling interactions with other cell-signaling pathways are an important part of RA's actions in odontogenesis. Recent investigations employing zebrafish and other teleost fish continued this work in an evolutionary context, and specifically demonstrated that RA is required for the initiation of tooth development. RA is also sufficient in certain circumstances to induce de novo tooth formation. Both effects appear to involve cranial-neural crest cells, again suggesting that RA signaling has a particular influence on odontoblast development. These teleost studies have also highlighted both evolutionary conservation and change in how RA is employed during odontogenesis in different vertebrate lineages, and thus raises the possibility that developmental changes to RA signaling has led to some of the diversity of form seen across vertebrate dentitions. Future progress in this area will come at least in part from expanding the species examined to get a better picture of how often RA signaling has changed in evolution and how this relates to the evolution of dental form.


Subject(s)
Biological Evolution , Dentition , Odontogenesis , Signal Transduction , Tooth/embryology , Tretinoin/metabolism , Zebrafish/embryology , Animals , Gene Expression Regulation, Developmental , Tooth/metabolism
10.
PLoS Genet ; 14(6): e1007449, 2018 06.
Article in English | MEDLINE | ID: mdl-29902209

ABSTRACT

Threespine stickleback fish offer a powerful system to dissect the genetic basis of morphological evolution in nature. Marine sticklebacks have repeatedly invaded and adapted to numerous freshwater environments throughout the Northern hemisphere. In response to new diets in freshwater habitats, changes in craniofacial morphology, including heritable increases in tooth number, have evolved in derived freshwater populations. Using a combination of quantitative genetics and genome resequencing, here we fine-mapped a quantitative trait locus (QTL) regulating evolved tooth gain to a cluster of ten QTL-associated single nucleotide variants, all within intron four of Bone Morphogenetic Protein 6 (Bmp6). Transgenic reporter assays revealed this intronic region contains a tooth enhancer. We induced mutations in Bmp6, revealing required roles for survival, growth, and tooth patterning. Transcriptional profiling of Bmp6 mutant dental tissues identified significant downregulation of a set of genes whose orthologs were previously shown to be expressed in quiescent mouse hair stem cells. Collectively these data support a model where mutations within a Bmp6 intronic tooth enhancer contribute to evolved tooth gain, and suggest that ancient shared genetic circuitry regulates the regeneration of diverse vertebrate epithelial appendages including mammalian hair and fish teeth.


Subject(s)
Bone Morphogenetic Protein 6/genetics , Smegmamorpha/genetics , Animals , Biological Evolution , Bone Morphogenetic Protein 6/physiology , Chromosome Mapping , Enhancer Elements, Genetic/genetics , Evolution, Molecular , Fresh Water , Gene Expression Regulation, Developmental/genetics , Genetic Linkage , Genotype , Introns/genetics , Phenotype , Polymorphism, Single Nucleotide/genetics , Quantitative Trait Loci , Tooth/embryology
11.
J Fish Biol ; 98(4): 906-918, 2021 Apr.
Article in English | MEDLINE | ID: mdl-31820456

ABSTRACT

Most extant vertebrates display a high variety of tooth and tooth-like organs (odontodes) that vary in shape, position over the body and nature of composing tissues. The development of these structures is known to involve similar genetic cascades and teeth and odontodes are believed to share a common evolutionary history. Gene expression patterns have previously been compared between mammalian and teleost tooth development but we highlight how the comparative framework was not always properly defined to deal with different tooth types or tooth developmental stages. Larger-scale comparative analyses also included cartilaginous fishes: sharks display oral teeth and dermal scales for which the gene expression during development started to be investigated in the small-spotted catshark Scyliorhinus canicula during the past decade. We report several descriptive approaches to analyse the embryonic tooth and caudal scale gene expressions in S. canicula. We compare these expressions wih the ones reported in mouse molars and teleost oral and pharyngeal teeth and highlight contributions and biases that arise from these interspecific comparisons. We finally discuss the evolutionary processes that can explain the observed intra and interspecific similarities and divergences in the genetic cascades involved in tooth and odontode development in jawed vertebrates.


Subject(s)
Biological Evolution , Elasmobranchii/classification , Odontogenesis/genetics , Vertebrates/classification , Vertebrates/genetics , Animals , Elasmobranchii/embryology , Elasmobranchii/genetics , Gene Expression Profiling , Mice , Sharks/embryology , Tooth/embryology , Vertebrates/embryology
12.
Development ; 144(15): 2824-2836, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28694256

ABSTRACT

During development and homeostasis, precise control of Wnt/ß-catenin signaling is in part achieved by secreted and membrane proteins that negatively control activity of the Wnt co-receptors Lrp5 and Lrp6. Lrp4 is related to Lrp5/6 and is implicated in modulation of Wnt/ß-catenin signaling, presumably through its ability to bind to the Wise (Sostdc1)/sclerostin (Sost) family of Wnt antagonists. To gain insights into the molecular mechanisms of Lrp4 function in modulating Wnt signaling, we performed an array of genetic analyses in murine tooth development, where Lrp4 and Wise play important roles. We provide genetic evidence that Lrp4 mediates the Wnt inhibitory function of Wise and also modulates Wnt/ß-catenin signaling independently of Wise. Chimeric receptor analyses raise the possibility that the Lrp4 extracellular domain interacts with Wnt ligands, as well as the Wnt antagonists. Diverse modes of Lrp4 function are supported by severe tooth phenotypes of mice carrying a human mutation known to abolish Lrp4 binding to Sost. Our data suggest a model whereby Lrp4 modulates Wnt/ß-catenin signaling via interaction with Wnt ligands and antagonists in a context-dependent manner.


Subject(s)
Receptors, LDL/metabolism , Tooth/embryology , Tooth/metabolism , beta Catenin/metabolism , Adaptor Proteins, Signal Transducing , Animals , Bone Morphogenetic Proteins/deficiency , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , LDL-Receptor Related Proteins , Low Density Lipoprotein Receptor-Related Protein-5/genetics , Low Density Lipoprotein Receptor-Related Protein-5/metabolism , Low Density Lipoprotein Receptor-Related Protein-6/genetics , Low Density Lipoprotein Receptor-Related Protein-6/metabolism , Mice , Mice, Mutant Strains , Receptors, LDL/deficiency , Receptors, LDL/genetics , Tooth/pathology , Wnt Signaling Pathway/genetics , Wnt Signaling Pathway/physiology , beta Catenin/genetics
13.
Nat Mater ; 18(6): 627-637, 2019 06.
Article in English | MEDLINE | ID: mdl-31114073

ABSTRACT

Cells are transplanted to regenerate an organs' parenchyma, but how transplanted parenchymal cells induce stromal regeneration is elusive. Despite the common use of a decellularized matrix, little is known as to the pivotal signals that must be restored for tissue or organ regeneration. We report that Alx3, a developmentally important gene, orchestrated adult parenchymal and stromal regeneration by directly transactivating Wnt3a and vascular endothelial growth factor. In contrast to the modest parenchyma formed by native adult progenitors, Alx3-restored cells in decellularized scaffolds not only produced vascularized stroma that involved vascular endothelial growth factor signalling, but also parenchymal dentin via the Wnt/ß-catenin pathway. In an orthotopic large-animal model following parenchyma and stroma ablation, Wnt3a-recruited endogenous cells regenerated neurovascular stroma and differentiated into parenchymal odontoblast-like cells that extended the processes into newly formed dentin with a structure-mechanical equivalency to native dentin. Thus, the Alx3-Wnt3a axis enables postnatal progenitors with a modest innate regenerative capacity to regenerate adult tissues. Depleted signals in the decellularized matrix may be reinstated by a developmentally pivotal gene or corresponding protein.


Subject(s)
Homeodomain Proteins/metabolism , Parenchymal Tissue/physiology , Tooth/cytology , Tooth/embryology , Adolescent , Animals , Female , Homeodomain Proteins/genetics , Humans , Incisor/cytology , Incisor/embryology , Mice, Inbred Strains , Molar, Third/cytology , Organ Culture Techniques , Parenchymal Tissue/cytology , Pregnancy , Promoter Regions, Genetic , Regeneration , Stromal Cells/physiology , Swine , Vascular Endothelial Growth Factor A/genetics , Wnt3A Protein/genetics , Wnt3A Protein/metabolism
14.
Biochem Soc Trans ; 48(6): 2729-2742, 2020 12 18.
Article in English | MEDLINE | ID: mdl-33155644

ABSTRACT

Teeth constitute a classical model for the study of signaling pathways and their roles in mediating interactions between cells and tissues in organ development, homeostasis and regeneration. Rodent teeth are mostly used as experimental models. Rodent molars have proved fundamental in the study of epithelial-mesenchymal interactions and embryonic organ morphogenesis, as well as to faithfully model human diseases affecting dental tissues. The continuously growing rodent incisor is an excellent tool for the investigation of the mechanisms regulating stem cells dynamics in homeostasis and regeneration. In this review, we discuss the use of teeth as a model to investigate signaling pathways, providing an overview of the many unique experimental approaches offered by this organ. We discuss how complex networks of signaling pathways modulate the various aspects of tooth biology, and the models used to obtain this knowledge. Finally, we introduce new experimental approaches that allow the study of more complex interactions, such as the crosstalk between dental tissues, innervation and vascularization.


Subject(s)
Signal Transduction , Tooth/embryology , Tooth/physiology , Animals , Cell Culture Techniques , Cell Differentiation , Epithelial-Mesenchymal Transition , Genetic Therapy , Germ Cells/metabolism , Homeostasis , Humans , Mesenchymal Stem Cells/metabolism , Mice , Models, Animal , Morphogenesis , Rats , Regeneration , Stem Cells/cytology , Tooth/metabolism
15.
Stem Cells ; 37(1): 26-32, 2019 01.
Article in English | MEDLINE | ID: mdl-30270477

ABSTRACT

Development of teeth depends on the reciprocal interactions between the surface epithelium (ectoderm) and the underlying neural crest-derived mesenchyme. These interactions are facilitated by the conserved signaling pathways, which build a complex network of signals and transcription factors. Tooth development starts at specific and predetermined loci in the oral ectoderm and is described as a morphologically distinct thickening of oral ectoderm, named dental lamina. Cells within the dental lamina invaginate into the underlying mesenchyme, generating placodes that mark the onset of individual tooth development. In the following stages of development, the tooth epithelium buds and folds transitioning through various shapes, including bud, cap, and bell shapes, which also identify the specific stages of tooth development. Although much of the molecular regulation of tooth development has been unraveled, the regulation of the initial stages of tooth development, as well as the cellular mechanisms that govern tooth development remain largely unknown. This review provides a systematic overview of the current knowledge on the molecular and cellular mechanisms that guide initial stages of tooth development and outlines the challenges which temper the progress. Stem Cells 2019;37:26-32.


Subject(s)
Cell Biology , Odontogenesis/immunology , Tooth/embryology , Humans
16.
Bioessays ; 40(12): e1800140, 2018 12.
Article in English | MEDLINE | ID: mdl-30387177

ABSTRACT

Efforts from diverse disciplines, including evolutionary studies and biomechanical experiments, have yielded new insights into the genetic, signaling, and mechanical control of tooth formation and functions. Evidence from fossils and non-model organisms has revealed that a common set of genes underlie tooth-forming potential of epithelia, and changes in signaling environments subsequently result in specialized dentitions, maintenance of dental stem cells, and other phenotypic adaptations. In addition to chemical signaling, tissue forces generated through epithelial contraction, differential growth, and skeletal constraints act in parallel to shape the tooth throughout development. Here recent advances in understanding dental development from these studies are reviewed and important gaps that can be filled through continued application of evolutionary and biomechanical approaches are discussed.


Subject(s)
Biological Evolution , Fossils , Tooth/embryology , Tooth/growth & development , Animals , Biomechanical Phenomena , Cell Differentiation , Cell Proliferation , Dentition , Fishes/growth & development , Gene Expression Regulation, Developmental , Stem Cells/cytology , Stem Cells/physiology , Tooth/cytology , Tooth/metabolism
17.
Proc Natl Acad Sci U S A ; 114(3): 540-545, 2017 01 17.
Article in English | MEDLINE | ID: mdl-28049837

ABSTRACT

Birds stand out from other egg-laying amniotes by producing relatively small numbers of large eggs with very short incubation periods (average 11-85 d). This aspect promotes high survivorship by limiting exposure to predation and environmental perturbation, allows for larger more fit young, and facilitates rapid attainment of adult size. Birds are living dinosaurs; their rapid development has been considered to reflect the primitive dinosaurian condition. Here, nonavian dinosaurian incubation periods in both small and large ornithischian taxa are empirically determined through growth-line counts in embryonic teeth. Our results show unexpectedly slow incubation (2.8 and 5.8 mo) like those of outgroup reptiles. Developmental and physiological constraints would have rendered tooth formation and incubation inherently slow in other dinosaur lineages and basal birds. The capacity to determine incubation periods in extinct egg-laying amniotes has implications for dinosaurian embryology, life history strategies, and survivorship across the Cretaceous-Paleogene mass extinction event.


Subject(s)
Dinosaurs/embryology , Tooth/embryology , Animals , Biological Evolution , Birds/embryology , Extinction, Biological , Female , Fossils/anatomy & histology , Odontogenesis , Reptiles/embryology , Species Specificity
18.
Int J Mol Sci ; 21(15)2020 07 24.
Article in English | MEDLINE | ID: mdl-32722078

ABSTRACT

MicroRNAs (miRNAs) are a class of naturally occurring small non-coding RNAs that post-transcriptionally regulate gene expression in organisms. Most mammalian miRNAs influence biological processes, including developmental changes, tissue morphogenesis and the maintenance of tissue identity, cell growth, differentiation, apoptosis, and metabolism. The miR-206-3p has been correlated with cancer; however, developmental roles of this miRNA are unclear. In this study, we examined the expression pattern and evaluated the developmental regulation of miR-206-3p during tooth morphogenesis using ex-vivo culture method. The expression pattern of miR-206-3p was examined in the epithelium and mesenchyme of developing tooth germ with stage-specific manners. Perturbation of the expression of miR-206-3p clearly altered expression patterns of dental-development-related signaling molecules, including Axin2, Bmp2, Fgf4, Lef1 and Shh. The gene expression complemented with change in cellular events including, apoptosis and proliferation which caused altered crown and pulp morphogenesis in renal-capsule-calcified teeth. Especially, mislocalization of ß-Catenin and SMAD1/5/8 were observed alongside dramatic alterations in the expression patterns of Fgf4 and Shh. Overall, our data suggest that the miR-206-3p regulate the cellular physiology during tooth morphogenesis through modulation of the Wnt, Bmp, Fgf, and Shh signaling pathways to form proper tooth pulp and crown.


Subject(s)
Gene Expression Regulation, Developmental , MicroRNAs/metabolism , Organogenesis , Tooth/embryology , Wnt Signaling Pathway , Animals , Mice , Mice, Inbred ICR , MicroRNAs/genetics
19.
Int J Mol Sci ; 21(21)2020 Oct 29.
Article in English | MEDLINE | ID: mdl-33138041

ABSTRACT

FUSE binding protein 1 (Fubp1), a regulator of the c-Myc transcription factor and a DNA/RNA-binding protein, plays important roles in the regulation of gene transcription and cellular physiology. In this study, to reveal the precise developmental function of Fubp1, we examined the detailed expression pattern and developmental function of Fubp1 during tooth morphogenesis by RT-qPCR, in situ hybridization, and knock-down study using in vitro organ cultivation methods. In embryogenesis, Fubp1 is obviously expressed in the enamel organ and condensed mesenchyme, known to be important for proper tooth formation. Knocking down Fubp1 at E14 for two days, showed the altered expression patterns of tooth development related signalling molecules, including Bmps and Fgf4. In addition, transient knock-down of Fubp1 at E14 revealed changes in the localization patterns of c-Myc and cell proliferation in epithelium and mesenchyme, related with altered tooth morphogenesis. These results also showed the decreased amelogenin and dentin sialophosphoprotein expressions and disrupted enamel rod and interrod formation in one- and three-week renal transplanted teeth respectively. Thus, our results suggested that Fubp1 plays a modulating role during dentinogenesis and amelogenesis by regulating the expression pattern of signalling molecules to achieve the proper structural formation of hard tissue matrices and crown morphogenesis in mice molar development.


Subject(s)
DNA-Binding Proteins/metabolism , Embryo, Mammalian/cytology , Gene Expression Regulation, Developmental , Morphogenesis , Odontogenesis , RNA-Binding Proteins/metabolism , Tooth/embryology , Animals , Cell Proliferation , DNA-Binding Proteins/genetics , Embryo, Mammalian/metabolism , Mice , Mice, Inbred ICR , RNA-Binding Proteins/genetics , Signal Transduction , Tooth/metabolism
20.
Dev Biol ; 441(2): 235-241, 2018 09 15.
Article in English | MEDLINE | ID: mdl-30017604

ABSTRACT

The phenotype of lens-ablated Mexican tetra (Astyanax mexicanus) compared to wild-type surface fish has been described and includes, among other effects, eye degeneration, changes in tooth number and cranial bone changes. Here, we investigate the spatiotemporal expression patterns of several key genes involved in the development of these structures. Specifically, we show that the expression of pitx2, bmp4 and shh is altered in the eye, oral jaw, nasal pit and forebrain in these lens-ablated fish. Furthermore, for the first time, we show altered pitx2 expression in the cavefish, which also has altered eye and tooth phenotypes. We thus provide evidence for a genetic linkage between the eye and tooth modules in this fish species. Furthermore, the altered pitx2 expression pattern, together with the described morphological features of the lens-ablated fish suggests that Astyanax mexicanus could be considered as an alternative teleost model organism in which to study Axenfeld-Rieger syndrome (ARS), a rare autosomal dominant developmental disorder that is associated with PITX2 and which has both ocular and non-ocular abnormalities.


Subject(s)
Characiformes , Fish Proteins , Gene Expression Regulation, Developmental , Genetic Linkage , Lens, Crystalline/embryology , Tooth/embryology , Animals , Anterior Eye Segment/abnormalities , Anterior Eye Segment/embryology , Characiformes/embryology , Characiformes/genetics , Disease Models, Animal , Eye Abnormalities/embryology , Eye Abnormalities/genetics , Eye Diseases, Hereditary , Fish Proteins/biosynthesis , Fish Proteins/genetics , Lens, Crystalline/pathology
SELECTION OF CITATIONS
SEARCH DETAIL