Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
J Periodontal Res ; 58(1): 184-194, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36517910

ABSTRACT

BACKGROUND AND OBJECTIVES: Hertwig's epithelial root sheath (HERS) plays a role in root dentin formation. It produces the epithelial rests of Malassez (ERM) for the induction of periodontal tissue development during root formation. Although ERM is thought to be caused by epithelial-mesenchymal transition (EMT), the mechanism by which HERS is maintained as epithelium is unknown. Here, we aimed to elucidate the molecular mechanisms regulating the relationship between HERS maintenance and ERM development. METHODS: To understand the relationship between HERS and ERM development during root formation, we observed the developing molar root using cytokeratin14 (CK14) Cre/tdTomato mice via stereomicroscopy. The relationship between semaphorin and transforming growth factor (TGF) signaling in the maintenance of HERS and ERM development was examined using CK14cre/R26-tdTomato mice and a HERS cell line. RESULTS: tdTomato-positive cells were observed on HERS and the migrating cells from HERS. The migrating cells showed reduced E-cadherin expression. In contrast, HERS cells expressed semaphorin receptors and active RhoA. Semaphorin signaling was associated with RhoA activation and cell-cell adhesion, while TGF-ß induced decreased E-cadherin and active RhoA expression, and consequently enhanced cell migration. CONCLUSION: HERS induces root formation by controlling epithelial maintenance and EMT through the opposing effects of semaphorin and TGF-ß signaling.


Subject(s)
Epithelial-Mesenchymal Transition , Transforming Growth Factor beta , Female , Mice , Animals , Transforming Growth Factor beta/pharmacology , Epithelial Cells , Tooth Root/physiology , Cadherins/metabolism
2.
J Anat ; 234(4): 465-479, 2019 04.
Article in English | MEDLINE | ID: mdl-30793310

ABSTRACT

Teeth are richly supported by blood vessels and peripheral nerves. The aim of this study was to describe in detail the developmental time-course and localization of blood vessels during early tooth formation and to compare that to innervation, as well as to address the putative role of vascular endothelial growth factor (VEGF), which is an essential regulator of vasculature development, in this process. The localization of blood vessels and neurites was compared using double immunofluorescence staining on sections at consecutive stages of the embryonic (E) and postnatal (PN) mandibular first molar tooth germ (E11-PN7). Cellular mRNA expression domains of VEGF and its signaling receptor VEGFR2 were studied using sectional radioactive in situ hybridization. Expression of VEGF mRNA and the encoded protein were studied by RT-PCR and western blot analysis, respectively, in the cap and early bell stage tooth germs, respectively. VEGFR2 was immunolocalized on tooth tissue sections. Smooth muscle cells were investigated by anti-alpha smooth muscle actin (αSMA) antibodies. VEGF showed developmentally regulated epithelial and mesenchymal mRNA expression domains including the enamel knot signaling centers that correlated with the growth and navigation of the blood vessels expressing Vegfr2 and VEGFR2 to the dental papilla and enamel organ. Developing blood vessels were present in the jaw mesenchyme including the presumptive dental mesenchyme before the appearance of the epithelial dental placode and dental neurites. Similarly, formation of a blood vessel plexus around the bud stage tooth germ and ingrowth of vessels into dental papilla at E14 preceded ingrowth of neurites. Subsequently, pioneer blood vessels in the dental papilla started to receive smooth muscle coverage at the early embryonic bell stage. Establishment and patterning of the blood vessels and nerves during tooth formation are developmentally regulated, stepwise processes that likely involve differential patterning mechanisms. Development of tooth vascular supply is proposed to be regulated by local, tooth-specific regulation by epithelial-mesenchymal tissue interactions and involving tooth target expressed VEGF signaling. Further investigations on tooth vascular development by local VEGF signaling, as well as how tooth innervation and development of blood vessels are integrated with advancing tooth organ formation by local signaling mechanisms, are warranted.


Subject(s)
Blood Vessels/growth & development , Tooth , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Animals , Body Patterning , Gene Expression Regulation, Developmental , Mice , Muscle, Smooth, Vascular/growth & development , Odontogenesis , Signal Transduction , Tooth/embryology , Tooth/innervation , Tooth/metabolism , Tooth Germ/embryology , Tooth Germ/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
3.
Cell Adh Migr ; 10(6): 618-626, 2016 11.
Article in English | MEDLINE | ID: mdl-27715429

ABSTRACT

The tooth, like many other organs, develops from both epithelial and mesenchymal tissues, and has proven to be a valuable tool with which to investigate organ formation and peripheral innervation. Tooth formation is regulated by local epithelial-mesenchymal tissue interactions, and is closely integrated with stereotypic dental nerve navigation and patterning. Recent analyses of the function and regulation of semaphorin 3A (SEMA3A) have shed light on the regulatory mechanisms that coordinate organogenesis and innervation at the tissue and molecular levels. In the tooth, SEM3A acts as a developmentally regulated secretory chemo-repellent, that controls tooth innervation during embryonic and postnatal development. The tooth germ governs its own innervation by a combination of local tissue interactions and SEMA3A expression. SEMA3A signaling, in turn, is controlled by a number of conserved signaling effectors, including TGF-ß superfamily members, FGF, and WNT; all function in embryo and organ development, and are essential for tooth histo-morphogenesis. Thus, SEMA3A driven axon guidance is integrated into key odontogenic signaling networks, establishing this protein as a critical molecular tether between 2 distinct developmental processes (morphogenesis and sensory innervation), both of which are required to obtain a functional tooth.


Subject(s)
Morphogenesis , Semaphorin-3A/metabolism , Signal Transduction , Tooth/growth & development , Tooth/innervation , Animals , Humans , Models, Biological , Organogenesis
4.
Cell Tissue Res ; 357(1): 15-29, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24752460

ABSTRACT

Semaphorin 3A (Sema3A) axon repellant serves multiple developmental functions. Sema3A mRNAs are expressed in epithelial and mesenchymal components of the developing incisor in a dynamic manner. Here, we investigate the functions of Sema3A during development of incisors using Sema3A-deficient mice. We analyze histomorphogenesis and innervation of mandibular incisors using immunohistochemistry as well as computed tomography and thick tissue confocal imaging. Whereas no apparent disturbances in histomorphogenesis or hard tissue formation of Sema3A (-/-) incisors were observed, nerve fibers were prematurely seen in the presumptive dental mesenchyme of the bud stage Sema3A (-/-) tooth germ. Later, nerves were ectopically present in the Sema3A (-/-) dental papilla mesenchyme during the cap and bell stages, whereas in the Sema3A (+/+) mice the first nerve fibers were seen in the pulp after the onset of dental hard tissue formation. However, no apparent topographic differences in innervation pattern or nerve fasciculation were seen inside the pulp between postnatal and adult Sema3A (+/+) or Sema3A (-/-) incisors. In contrast, an abnormally large number of nerves and arborizations were observed in the Sema3A (-/-) developing dental follicle target field and periodontium and, unlike in the wild-type mice, nerve fibers were abundant in the labial periodontium. Of note, the observed defects appeared to be mostly corrected in the adult incisors. The expressions of Ngf and Gdnf neurotrophins and their receptors were not altered in the Sema3A (-/-) postnatal incisor or trigeminal ganglion, respectively. Thus, Sema3A is an essential, locally produced chemorepellant, which by creating mesenchymal exclusion areas, regulates the timing and patterning of the dental nerves during the development of incisor tooth germ.


Subject(s)
Incisor/growth & development , Incisor/innervation , Semaphorin-3A/physiology , Tooth Germ/growth & development , Tooth Germ/innervation , Animals , Mice , Mice, Transgenic , Odontogenesis/physiology , Semaphorin-3A/deficiency
5.
Exp Cell Res ; 325(2): 72-7, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24631295

ABSTRACT

In addition to being an advantageous model to investigate general molecular mechanisms of organ formation, the tooth is a distinct target organ for peripheral nerve innervation. These nerves are required for the function and protection of the teeth and, as shown in fish, also for their regeneration. This review focuses on recent findings of the local tissue interactions and molecular signaling mechanisms that regulate the early nerve arrival and patterning of mouse mandibular molar tooth sensory innervation. Dental sensory nerve growth and patterning is a stepwise process that is intimately linked to advancing tooth morphogenesis. In particular, nerve growth factor and semaphorin 3A serve as essential functions during and are iteratively used at different stages of tooth innervation. The tooth germ controls development of its own nerve supply, and similar to the development of the tooth organ proper, tissue interactions between dental epithelial and mesenchymal tissues control the establishment of tooth innervation. Tgf-ß, Wnt, and Fgf signaling, which regulate tooth formation, are implicated to mediate these interactions. Therefore, tissue interactions mediated by conserved signal families may constitute key mechanism for the integration of tooth organogenesis and development of its peripheral nerve supply.


Subject(s)
Cell Communication , Models, Animal , Morphogenesis/physiology , Neurogenesis/physiology , Tooth/embryology , Tooth/innervation , Animals , Humans , Mice
6.
Differentiation ; 84(5): 371-9, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23142733

ABSTRACT

Timing and patterning of dental pulp innervation are strictly spatio-temporally regulated but it is still not known how they are controlled at molecular level. We analyzed postnatal innervation of the dental pulp in the mandibular first molar of mice deficient for Semaphorin 3A (Sema3A) axon repellant molecule. Immunohistochemical localization of nerve fibers on serial sections covering the whole tooth germs using anti-peripherin antibody revealed that nerve fibers were prematurely present within the preodontoblast layer next to the inner enamel epithelium already at PN0 in Sema3A(-/-) mice. In contrast, in the wild-type (Sema3A(+/+)) mice nerve fibers were seen in the pulp only after enamel formation at PN3. The nerves in Sema3A(-/-) pulp were notably defasciculated and thinner compared to that of Sema3A(+/+) mice. A premature formation of an abnormal, enlarged nerve plexus with a high number of arborizations was apparent in the pulp-dentin border target area in Sema3A(-/-) already at PN2 whereas in the wild-type mice the first sign of plexus formation was seen at PN7. The expression of mRNAs for Ngf, Gdnf and Ncam neuroregulatory molecules in mandibular molar as well as receptors for neurotrophic factors and class 3 semaphorins including Sema3A (TrkA, p75, TrkB, TrkC, Ret, Npn1, Npn2, PlxA4) in trigeminal ganglia were not altered in the Sema3A(-/-) mice. Collectively, this data show that Sema3A serves an essential role in molar tooth pulp innervation controlling the timing of nerve fiber penetration into the pulp, their patterning and the formation of nerve plexus at pulp-dentin border area, and provide further support for the hypothesis that tooth innervation is regulated by the coordinated activity of locally expressed neuroregulatory molecules exerting positive and negative influences on growing dental nerve fibers.


Subject(s)
Dental Pulp/innervation , Semaphorin-3A/metabolism , Animals , Dental Enamel/metabolism , Dental Pulp/metabolism , Dentin/metabolism , Gene Expression Regulation , Glial Cell Line-Derived Neurotrophic Factor/genetics , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Mice , Mice, Inbred C57BL , Molar/anatomy & histology , Molar/metabolism , Nerve Fibers/metabolism , Nerve Growth Factor/genetics , Nerve Growth Factor/metabolism , Neural Cell Adhesion Molecules/genetics , Neural Cell Adhesion Molecules/metabolism , RNA, Messenger/biosynthesis , Semaphorin-3A/genetics , Trigeminal Ganglion/metabolism
7.
Acta Odontol Scand ; 70(2): 140-8, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21815834

ABSTRACT

OBJECTIVE: Semaphorins form a family of axon wiring molecules but still little is known about their role in tooth formation. A class 3 semaphorin, Semaphorin3F (Sema3F), besides acting as a chemorepellant for different types of axons, controls a variety of non-neuronal developmental processes. MATERIALS AND METHODS: Cellular mRNA expression patterns of Sema3F as well as neuropilin 1 (Npn1), neuropilin 2 (Npn2), plexinA3 and plexinA4 receptors were analyzed by sectional in situ hybridization in the mouse molar tooth during postnatal days 0-7. The expression of the receptors was studied in PN5 trigeminal ganglia. RESULTS: Sema3F, Npn1, -2 and plexinA4 exhibited distinct, spatiotemporally changing expression patterns, whereas plexinA3 was not observed in the tooth germs. Besides being expressed in the base of the dental mesenchyme Sema3F, like plexinA4, Npn1 and -2, was present in the ameloblast cell lineage. Npn1 and Npn2 were additionally seen in the pulp horns and endothelial cells and like PlexinA4 in the developing alveolar bone. Npn1, plexinA3 and -A4 were observed in trigeminal ganglion neurons. CONCLUSIONS: Sema3F may act as a tooth target-derived axonal chemorepellant controlling establishment of the tooth nerve supply. Furthermore, Sema3F, like Npn1, -2 and plexinA4 may serve non-neuronal functions by controlling the development of the ameloblast cell lineage. Moreover, Npn1 and Npn2 may regulate dental vasculogenesis and, together with PlexinA4, alveolar bone formation. Further analyses such as investigation of transgenic mouse models will be required to elucidate in vivo signaling functions of Sema3F and the receptors in odontogenesis.


Subject(s)
Dental Pulp/innervation , Nerve Tissue Proteins/biosynthesis , Neuropilins/biosynthesis , Semaphorins/biosynthesis , Tooth/innervation , Ameloblasts/cytology , Animals , Cell Differentiation , Dental Pulp/blood supply , Dental Pulp/metabolism , Gene Expression , In Situ Hybridization , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Membrane Proteins/physiology , Mice , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Neuropilin-1/biosynthesis , Neuropilin-1/genetics , Neuropilin-2/biosynthesis , Neuropilin-2/genetics , Neuropilins/genetics , RNA, Messenger/biosynthesis , Receptors, Cell Surface/biosynthesis , Receptors, Cell Surface/genetics , Semaphorins/genetics , Semaphorins/physiology , Tooth/blood supply , Tooth/metabolism , Tooth Crown/metabolism , Tooth Germ/metabolism , Trigeminal Ganglion/metabolism
8.
Acta Odontol Scand ; 70(3): 184-9, 2012 May.
Article in English | MEDLINE | ID: mdl-21793640

ABSTRACT

OBJECTIVE: Semaphorin 3A (Sema3A) is an essential chemorepellant controlling peripheral axon pathfinding and patterning, but also serves non-neuronal cellular functions. Incisors of rodent are distinctive from molars as they erupt continuously, have only one root and enamel is present only on the labial side. The aim of this study is to address putative regulatory roles of Sema3A chemorepellant in the development of incisor innervation and formation. MATERIALS AND METHODS: This study analyzed expression of Sema3A mRNAs during embryonic and early post-natal stages of mouse mandibular incisor using sectional radioactive in situ hybridization. RESULTS: Although Sema3A mRNAs were observed in condensed dental mesenchyme during the early bud stage, they were absent in dental papilla or pulp at later stages. Sema3A mRNAs were observed in the dental epithelium including the cervical loops and a prominent expression was also seen in alveolar bone. Interestingly, transcripts were absent from the mesenchymal dental follicle target area (future periodontal ligament) throughout the studied stages. CONCLUSION: The expression patterns of Sema3A indicate that it may control the timing and patterning of the incisor innervation. In particular, Sema3A appears to regulate innervation of the periodontal ligament, while nerve penetration into the incisor dental pulp appears not to be dependent on Sema3A. Moreover, Sema3A may regulate the functions of cervical loops and the development of alveolar bone. Future study with Sema3A deficient mice will help to elucidate the putative neuronal and non-neuronal functions of Sema3A in incisor tooth development.


Subject(s)
Dental Pulp/embryology , Incisor/metabolism , Odontogenesis/physiology , Periodontal Ligament/innervation , Semaphorin-3A/metabolism , Animals , Axons/physiology , Dental Pulp/innervation , Gene Expression Regulation, Developmental , Incisor/embryology , Mandible , Mice , Periodontal Ligament/embryology , RNA, Messenger/analysis , Semaphorin-3A/genetics , Tooth Germ/embryology , Tooth Germ/innervation , Trigeminal Nerve/embryology , Trigeminal Nerve/physiology
9.
Acta Odontol Scand ; 69(6): 360-6, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21449687

ABSTRACT

OBJECTIVE: To investigate and compare the cellular expression of non-secreted Fgf11-14 and secreted Fgf15-18 and -20 mRNAs during tooth formation. MATERIALS AND METHODS: mRNA expression was analyzed from the morphological initiation of the mouse mandibular first molar development to the onset of crown calcification using sectional in situ hybridization. RESULTS: This study found distinct, differentially regulated expression patterns for the Fgf11-13, -15-17 and -20, in particular in the epithelial-mesenchymal interface, whereas Fgf14 and 18 mRNAs were not detected. Fgf11, -15, -16, -17 and -20 were seen in the epithelium, whereas Fgf12 and -13 signals were restricted to the mesenchymal tissue component of the tooth. Fgf11 was observed in the putative epithelial signaling areas, the tertiary enamel knots and enamel free areas of the calcifying crown. Fgf15, Fgf17 and -20 were transiently colocalized in the thickened dental epithelium at E11.5. Later Fgf15 and -20 were exclusively expressed in the epithelial enamel knot signaling centers. In contrast, Fgf13 was present in the dental mesenchyme including odontoblasts cell lineage, whereas Fgf12 appeared transiently in the preodontoblasts. CONCLUSIONS: The expression of the Fgf11-13, -15, -17 and -20 in the epithelial signaling centers and/or epithelial-mesenchymal interfaces at key stages of the tooth formation suggest important functions in odontogenesis. Future analyses of the transgenic mice will help elucidate in vivo functions of the studied Fgfs during odontogenesis and whether any of the functions of the tooth expressed epithelial and mesenchymal Fgfs of different sub-families are redundant.


Subject(s)
Fibroblast Growth Factors/genetics , Gene Expression Regulation, Developmental/genetics , Molar/embryology , Odontogenesis/genetics , Ameloblasts/cytology , Animals , Cell Lineage , Dental Papilla/embryology , Enamel Organ/embryology , Epithelium/embryology , Fibroblast Growth Factors/analysis , In Situ Hybridization , Mesoderm/embryology , Mice , Odontoblasts/cytology , Tooth Calcification/genetics , Tooth Crown/embryology , Tooth Germ/embryology
10.
Article in English | MEDLINE | ID: mdl-20493789

ABSTRACT

The Atlantic cod (Gadus morhua) is an emerging aquaculture species. Efforts to develop and characterize its genomic recourses, including draft-grade genome sequencing, have been initiated by the research community. The transcriptome represents the whole complement of RNA transcripts in cells and tissues and reflects the expressed genes at various life stages, tissue types, physiological states, and environmental conditions. We are investigating the Atlantic cod transcriptome by Roche 454, Illumina GA, and ABI SOLiD deep sequencing platforms and corresponding bioinformatics. Both embryonic developmental stages and adult tissues are studied. Here we summarize our recent progress in the analyses of nuclear and mitochondrial polyA mRNAs, non-protein-coding intermediate RNAs, and regulatory microRNAs.


Subject(s)
Gadus morhua/genetics , Genome/genetics , Sequence Analysis, DNA/methods , Animals , Computational Biology , Gene Expression Profiling , MicroRNAs/genetics
11.
Gene Expr Patterns ; 10(2-3): 140-3, 2010.
Article in English | MEDLINE | ID: mdl-20170752

ABSTRACT

Wnt signaling is essential for tooth formation and Dact proteins modulate Wnt signaling by binding to the intracellular protein Dishevelled (Dvl). Comparison of the three known mouse Dact genes, Dact1-3, from the morphological initiation of mandibular first molar development through the onset of root formation using section in situ hybridization showed distinct, complementary and overlapping expression patterns for these genes. Whereas Dact2 expression was restricted to the dental epithelium, including the enamel knot signaling centers and pre-ameloblasts, Dact1 and Dact3 showed developmentally regulated expression in the dental mesenchyme. Both Dact1 and Dact3 mRNAs were first detected in the presumptive dental mesenchyme. After being downregulated from the condensing dental mesenchyme of the bud stage tooth germ, Dact1 was upregulated in the dental follicle mesenchyme at the cap stage and subsequently also in the dental papilla at the bell stage, where the expression persisted to the postnatal stages. In contrast, Dact3 transcripts persisted throughout the dental mesenchyme, including the preodontoblasts, during embryogenesis before transcripts were largely downregulated from the tooth germ postnatally. Collectively, these results suggest that Dact1 and -3 may contribute to early tooth formation by modulation of Wnt signaling pathways in the mesenchyme, including preodontoblasts, whereas Dact2 may play important signal-modulating roles in the adjacent epithelial cells including the enamel knot signaling centers and pre-ameloblasts. Future loss-of-function studies will help elucidate whether any of these functions are redundant, particularly for Dact1 and Dact3.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Intracellular Signaling Peptides and Proteins/genetics , Mesoderm/metabolism , Odontogenesis/genetics , Adaptor Proteins, Signal Transducing/biosynthesis , Animals , Epithelium/metabolism , Gene Expression Regulation, Developmental , Mice , Molar/metabolism , RNA, Messenger/metabolism , RNA-Binding Proteins , Tooth Germ/metabolism , Wnt Proteins/physiology
12.
Ann Anat ; 190(2): 178-87, 2008.
Article in English | MEDLINE | ID: mdl-18413271

ABSTRACT

During embryogenesis, dental trigeminal axon navigation and patterning in the developing tooth take place in a highly spatio-temporally directed manner that is tightly linked to tooth morphogenesis and cell differentiation. Tooth formation is regulated by sequential and reciprocal tissue interactions between dental epithelium and neural crest-derived ectomesenchymal cells. This odontogenic secondary induction is mediated by signal molecules of different conserved families. Recent molecular and experimental data have provided evidence that local instructive signaling from the early odontogenic epithelium also controls dental axon navigation in the dental mesenchyme. In this review, we discuss recent molecular data regarding tooth formation and innervation and the putative role of the secondary induction in coordinating these two developmental processes. Importantly, because it has not yet been shown that the interactions that regulate tooth innervation include signaling to the dental epithelium and that they are reciprocal, it remains to be demonstrated that secondary induction controls the establishment of tooth nerve supply. Moreover, the key question of which molecule(s), if any, integrate tooth morphogenesis and the development of dental sensory trigeminal innervation remains to be answered.


Subject(s)
Axons/physiology , Odontogenesis/physiology , Tooth/embryology , Tooth/innervation , Body Patterning , Dentition , Embryonic Development , Humans , Nerve Fibers , Trigeminal Nerve/embryology
13.
Arch Oral Biol ; 53(9): 865-73, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18436190

ABSTRACT

OBJECTIVE: Our goal was to study the development of pioneer sympathetic innervation of dental pulp of mouse mandibular first molar. DESIGN: We used double fluorescent immunohistochemistry with tyrosine hydroxylase (TH) and anti-medium-chain neurofilament (2H3) antibodies to detect sympathetic and sensory nerve fibres. Serial sections of whole teeth from postnatal days (PN) 0-14, trigeminal and sympathetic superior cervical ganglia of PN 15 mice were examined with confocal microscope. RESULTS: There were two main findings. The unexpected finding was that 2H3 antibody was specific only for sensory nerve fibres and neurons and failed to stain either sympathetic nerve fibres or neurons. The main finding was that although both sympathetic and sensory nerve fibres were already seen near the tooth germ at the newborn stage, the pioneer sympathetic nerve fibres were first observed in the dental pulp only after the onset of root formation on day 9, in contrast to sensory nerve fibres which entered the tooth already on day 4. CONCLUSION: Pioneer sympathetic innervation of dental pulp starts on postnatal day 9 and follows sensory innervation. This indicates differential developmental regulation of the initial sensory and sympathetic innervation of teeth and provides essential background data for further studies on the molecular regulation of pulp innervation.


Subject(s)
Dental Pulp/innervation , Mandible/innervation , Molar/innervation , Nerve Fibers/physiology , Tooth Germ/innervation , Trigeminal Ganglion/physiology , Animals , Animals, Newborn , Immunohistochemistry , Mice , Nerve Growth Factors/physiology
14.
Mech Dev ; 124(11-12): 868-83, 2007.
Article in English | MEDLINE | ID: mdl-17951031

ABSTRACT

Dental trigeminal nerve fiber growth and patterning are strictly integrated with tooth morphogenesis, but it is still unknown, how these two developmental processes are coordinated. Here we show that targeted inactivation of the dental epithelium expressed Fgfr2b results in cessation of the mouse mandibular first molar development at the degenerated cap stage and the failure of the trigeminal molar nerve to establish the lingual branch at E13.5 stage while the buccal branch develops properly. This axon patterning defect correlates to the histological absence of the mesenchymal dental follicle and adjacent Semaphorin3A-free dental follicle target field as well as appearance of ectopic Sema3A expression domain in the lingual side of the epithelial bud. Although the mesenchymal ligands for Fgfr2b, Fgf3 and -10 were present in the Fgfr2b(-/)(-) dental mesenchyme, mutant dental epithelium showed dramatically reduced proliferation and the lack of Fgf3. Tgfbeta1, which controls Sema3A was absent from the Fgfr2b(-/-) tooth germ, and Sema3A was specifically downregulated in the dental mesenchyme at the bud and cap stage. In addition, the epithelial primary enamel knot signaling center although being molecularly present neither was histologically detectable nor expressed Bmp4 and Fgf3 as well as Fgf4, which is essential for tooth morphogenesis and stimulates mesenchymal Fgf3 and Tgfbeta1. Fgf4 beads rescued Tgfbeta1 in the Fgfr2b(-/-) dental mesenchyme explants and Tgfbeta1 induced de novo Sema3A expression in the dental mesenchyme. Collectively these results demonstrate that epithelial Fgfr2b controls tooth morphogenesis and dental axon patterning, and suggests that Fgfr2b, by mediating local epithelial-mesenchymal interactions, integrates these two distinct developmental processes during odontogenesis.


Subject(s)
Axons/metabolism , Body Patterning , Epithelial Cells/metabolism , Mesoderm/metabolism , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Tooth/embryology , Trigeminal Ganglion/embryology , Animals , Apoptosis , Cell Proliferation , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Epithelial Cells/cytology , Mice , Models, Biological , Molar/cytology , Molar/embryology , Nerve Growth Factors/metabolism , Neurites/metabolism , Receptor, Fibroblast Growth Factor, Type 2/deficiency , Semaphorin-3A/metabolism , Tooth/cytology , Tooth Germ/cytology , Tooth Germ/embryology , Transforming Growth Factor beta1/metabolism
15.
Anat Rec A Discov Mol Cell Evol Biol ; 288(12): 1250-8, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17066377

ABSTRACT

The cranial base is formed by endochondral ossification and is characterized by the presence of the synchondrosis growth centers. The aim of this study was to describe the histological development of the mouse midsagittal cranial base area from embryonic day 10 (E10) to the postnatal age of 2 months. The Bmp family of signaling molecules serves important functions in embryo and bone development and may therefore play a significant role in the early formation of the cranial base. To investigate this, we analyzed the mRNA pattern of expression of Bmp2-6 in the mouse cranial base from E10 to 5 days postnatally using radioactive in situ hybridization. We found that the formation of the mouse cranial base corresponds to that of rat and proceeds in a caudorostral sequence. Moreover, all Bmps studied showed distinct and overlapping developmentally regulated expression domains. Bmp2, Bmp5, and Bmp6 were expressed in the early mesenchymal condensations. Later, Bmp2, Bmp3, Bmp4, and Bmp5 were detected in the perichondrium and in the adjacent mesenchyme. Subsequently, Bmp2 and Bmp6 expressions were confined to hypertrophic chondrocytes, while Bmp3, Bmp4, and Bmp5 were expressed in the osteoblasts of the trabecular bone and bone collar. Interestingly, Bmp3 was uniquely expressed postnatally in the resting zone of the synchondrosis growth center, suggesting a role in the regulation of cranial base growth. These results suggest that Bmp signaling may serve specific and synergistic functions at different key stages of cranial base development and growth.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Gene Expression Regulation, Developmental , RNA, Messenger/metabolism , Skull Base/metabolism , Transforming Growth Factor beta/metabolism , Animals , Bone Morphogenetic Protein 2 , Bone Morphogenetic Protein 3 , Bone Morphogenetic Protein 4 , Bone Morphogenetic Protein 5 , Bone Morphogenetic Protein 6 , In Situ Hybridization , Mice , Skull Base/embryology , Time Factors
16.
Int J Dev Biol ; 50(6): 511-21, 2006.
Article in English | MEDLINE | ID: mdl-16741866

ABSTRACT

The BMP signalling pathway is conserved throughout evolution and essential for mammalian embryonic and postnatal development and growth. In the vertebrate head, this signal is involved in the development of a variety of structures and shows divergent roles. During early head development, BMP signalling participates in the induction, formation, determination and migration of the cranial neural crest cells, which give rise to most of the craniofacial structures. Subsequently, it is also important for patterning and formation of facial primordia. During craniofacial skeletogenesis, BMP signalling is an early inductive signal required for committed cell migration, condensation, proliferation and differentiation. Thereafter, BMP signalling maintains regulatory roles in skeletons and skeletal growth centres. For myogenesis, BMP signalling is a negative regulator. Importantly, myostatin has been identified as a key mediator in this process. During palatogenesis, the crucial role of BMP signalling is demonstrated by mouse models with Alk2 or Alk3 (BMP type I receptors) deletion from the neural crest or craniofacial region, in which cleft palate is one of the major anomalies. BMP signalling is also an important participant for tooth development, regulating early tooth morphogenesis and subsequent odontoblast differentiation. In this review these aspects are discussed in detail with a focus on recent advances.


Subject(s)
Bone Morphogenetic Proteins/physiology , Facial Bones/embryology , Signal Transduction/physiology , Skull/embryology , Animals , Mice
17.
Dev Dyn ; 234(3): 482-8, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16217735

ABSTRACT

Like many other organs, the tooth develops as a result of the epithelial-mesenchymal interactions. In addition, the tooth is a well-defined peripheral target organ for sensory trigeminal nerves, which are required for the function and protection of the teeth. Dental trigeminal axon growth and patterning are tightly linked with advancing tooth morphogenesis and cell differentiation. This review summarizes recent findings on the regulation of dental axon pathfinding, which have provided evidence that the development of tooth trigeminal innervation is controlled by epithelial-mesenchymal interactions. The early dental epithelium possesses the information to instruct tooth nerve supply, and signals mediating these interactions are part of the signaling networks regulating tooth morphogenesis. Tissue interactions, thus, appear to provide a central mechanism of spatiotemporally orchestrating tooth formation and dental axon navigation and patterning.


Subject(s)
Axons/physiology , Morphogenesis , Tooth/embryology , Tooth/innervation , Animals , Semaphorin-3A/metabolism , Tooth/anatomy & histology , Tooth/cytology
18.
Anat Rec A Discov Mol Cell Evol Biol ; 286(2): 891-8, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16145660

ABSTRACT

The cranial base, located between the cranial vault and the facial bones, plays an important role in integrated craniofacial development and growth. Transgenic Shh and Sox9-deficient mice show similar defects in cranial base patterning. Therefore, in order to examine potential interactions of Shh, Ihh, another member of the Hh family, and Sox9 during cranial base development and growth, we investigated their cellular mRNA expression domains in the embryonic (E) and early postnatal (PN) cranial base from E10 to PN5 using sectional radioactive 35-S in situ hybridization. Of the Hhs, Shh was observed in the foregut epithelium and the notochord, while Sox9 showed broad expression in the loose mesenchyme of the cranial base area during E10-E11. Subsequently, from E12 onward, all genes were observed in the developing cranial base, and after birth the genes were prominently colocalized in the prehypertrophic chondrocytes of the synchondroses. Collectively, these data suggest that Hh-Sox9 auto- and paracrine signaling interactions may provide a critical mechanism for regulating the patterning of the cranial base as well as for its development and growth.


Subject(s)
Gene Expression Regulation, Developmental , High Mobility Group Proteins/genetics , Osteogenesis/genetics , Skull Base/embryology , Skull Base/growth & development , Trans-Activators/genetics , Transcription Factors/genetics , Animals , Animals, Newborn , Bone Development/genetics , Bone and Bones/embryology , Bone and Bones/metabolism , Embryo, Mammalian , Embryonic Development , Gestational Age , Hedgehog Proteins , High Mobility Group Proteins/metabolism , In Situ Hybridization , Mice , Mice, Inbred Strains , RNA, Messenger/metabolism , SOX9 Transcription Factor , Skull Base/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism
19.
Dev Dyn ; 233(1): 161-6, 2005 May.
Article in English | MEDLINE | ID: mdl-15759274

ABSTRACT

Wnt signaling is essential for tooth formation. Members of the Dickkopf (Dkk) family modulate the Wnt signaling pathway by binding to the Wnt receptor complex. Comparison of Dkk1, -2, and -3 mRNA expression during mouse tooth formation revealed that all three genes showed distinct spatiotemporally regulated expression patterns. Dkk1 was prominently expressed in the distal, incisor-bearing mesenchyme area of the mandibular process during the initial stages of tooth formation. During molar morphogenesis Dkk1 was detected in the dental mesenchyme, including the preodontoblasts. Dkk2 was seen in the dental papilla, whereas Dkk3 was specifically expressed in the putative epithelial signaling centers, the primary and secondary enamel knots. Postnatally, Dkk1 was prominently expressed in the preodonto- and odontoblasts, while Dkk3 mRNAs were transiently seen in the preameloblasts before the onset of enamel matrix secretion. These results suggest that modulation of Wnt-signaling by Dkks may serve important functions in patterning of dentition as well as in crown morphogenesis and dental hard-tissue formation.


Subject(s)
Intercellular Signaling Peptides and Proteins/metabolism , Proteins/metabolism , Tooth/embryology , Adaptor Proteins, Signal Transducing , Animals , Gene Expression Regulation, Developmental/physiology , Intercellular Signaling Peptides and Proteins/genetics , Mice , Proteins/genetics , RNA, Messenger/metabolism , Tooth/metabolism , Wnt Proteins
20.
J Mol Histol ; 36(6-7): 419-26, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16521043

ABSTRACT

The Dickkopf (Dkk) family and Mmp9 are important for apoptosis and a number of other developmental processes. However, little is known about their roles in the development of cranial base, which is an important structure for coordinated development and growth of the craniofacial skeletons. In order to establish whether and in what way these genes are involved in cranial base development, we examined their expression patterns and cell apoptosis. Dkk1 was first seen in the perichondral mesenchyme in restricted domains from E14, and later in the migrating mesenchymal cells within the cartilage. Thereafter, it was widespread throughout the bones of the cranial base. The expression was downregulated in postnatal stages. Dkk2 was localized in the perichondral mesenchyme outlining the anterior cranial base in embryogenesis. Dkk3 was mainly detected in the occipital-vertebral joint at E13 and E14. Mmp9 transcripts were clustered in the inner layer of perichondral mesenchyme, juxtaposed with the terminally differentiated hypertrophic chondrocytes from E14. Later Mmp9-expressing cells were found at the sites of chondrocyte apoptosis. This was particularly clear at the distal ends of the synchondroses. These data indicate that Mmp9 regulates skeletogenesis in cranial base in a manner that is largely similar to that of the appendicular skeletons. Expression of Dkks suggests other roles that remain to be defined.


Subject(s)
Apoptosis , Gene Expression Regulation, Developmental , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Adaptor Proteins, Signal Transducing , Animals , In Situ Hybridization , Mice , RNA, Messenger/genetics , RNA, Messenger/metabolism , Skull Base/cytology , Skull Base/embryology , Skull Base/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL
...