Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 648
Filter
Add more filters

Publication year range
1.
Development ; 151(9)2024 May 01.
Article in English | MEDLINE | ID: mdl-38619396

ABSTRACT

Piezo1 and Piezo2 are recently reported mechanosensory ion channels that transduce mechanical stimuli from the environment into intracellular biochemical signals in various tissues and organ systems. Here, we show that Piezo1 and Piezo2 display a robust expression during jawbone development. Deletion of Piezo1 in neural crest cells causes jawbone malformations in a small but significant number of mice. We further demonstrate that disruption of Piezo1 and Piezo2 in neural crest cells causes more striking defects in jawbone development than any single knockout, suggesting essential but partially redundant roles of Piezo1 and Piezo2. In addition, we observe defects in other neural crest derivatives such as malformation of the vascular smooth muscle in double knockout mice. Moreover, TUNEL examinations reveal excessive cell death in osteogenic cells of the maxillary and mandibular arches of the double knockout mice, suggesting that Piezo1 and Piezo2 together regulate cell survival during jawbone development. We further demonstrate that Yoda1, a Piezo1 agonist, promotes mineralization in the mandibular arches. Altogether, these data firmly establish that Piezo channels play important roles in regulating jawbone formation and maintenance.


Subject(s)
Ion Channels , Jaw , Neural Crest , Animals , Mice , Gene Expression Regulation, Developmental , Ion Channels/metabolism , Ion Channels/genetics , Jaw/embryology , Jaw/metabolism , Mandible/embryology , Mandible/metabolism , Mice, Knockout , Neural Crest/metabolism , Osteogenesis/genetics , Pyrazines , Thiadiazoles
2.
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 , Animals , Mice , Cell Lineage/genetics , Epithelium/metabolism , Gene Regulatory Networks , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mandible/metabolism , Odontogenesis/genetics , Signal Transduction , SOXB1 Transcription Factors/metabolism , SOXB1 Transcription Factors/genetics , Tooth/metabolism , Tooth/growth & development , Tooth/embryology , Transcription Factor AP-2/metabolism , Transcription Factor AP-2/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
3.
Differentiation ; 138: 100782, 2024.
Article in English | MEDLINE | ID: mdl-38810379

ABSTRACT

The mandible is composed of several musculoskeletal tissues including bone, cartilage, and tendon that require precise patterning to ensure structural and functional integrity. Interestingly, most of these tissues are derived from one multipotent cell population called cranial neural crest cells (CNCCs). How CNCCs are properly instructed to differentiate into various tissue types remains nebulous. To better understand the mechanisms necessary for the patterning of mandibular musculoskeletal tissues we utilized the avian mutant talpid2 (ta2) which presents with several malformations of the facial skeleton including dysplastic tendons, mispatterned musculature, and bilateral ectopic cartilaginous processes extending off Meckel's cartilage. We found an ectopic epithelial BMP signaling domain in the ta2 mandibular prominence (MNP) that correlated with the subsequent expansion of SOX9+ cartilage precursors. These findings were validated with conditional murine models suggesting an evolutionarily conserved mechanism for CNCC-derived musculoskeletal patterning. Collectively, these data support a model in which cilia are required to define epithelial signal centers essential for proper musculoskeletal patterning of CNCC-derived mesenchyme.


Subject(s)
Mandible , Neural Crest , Animals , Chick Embryo , Mice , Avian Proteins/genetics , Avian Proteins/metabolism , Body Patterning/genetics , Cartilage/metabolism , Cartilage/growth & development , Cartilage/cytology , Cell Differentiation , Chickens/genetics , Cilia/metabolism , Cilia/genetics , Gene Expression Regulation, Developmental , Mandible/growth & development , Mandible/metabolism , Mesoderm/cytology , Mesoderm/metabolism , Mesoderm/growth & development , Neural Crest/cytology , Neural Crest/metabolism , Signal Transduction , SOX9 Transcription Factor/metabolism , SOX9 Transcription Factor/genetics
4.
Development ; 147(11)2020 06 11.
Article in English | MEDLINE | ID: mdl-32439763

ABSTRACT

Craniofacial development is a complex morphogenic process that requires highly orchestrated interactions between multiple cell types. Blood vessel-derived angiocrine factors are known to promote proliferation of chondrocytes in Meckel's cartilage to drive jaw outgrowth, however the specific factors controlling this process remain unknown. Here, we use in vitro and ex vivo cell and tissue culture, as well as genetic mouse models, to identify IGF1 as a novel angiocrine factor directing Meckel's cartilage growth during embryonic development. We show that IGF1 is secreted by blood vessels and that deficient IGF1 signalling underlies mandibular hypoplasia in Wnt1-Cre; Vegfafl/fl mice that exhibit vascular and associated jaw defects. Furthermore, conditional removal of IGF1 from blood vessels causes craniofacial defects including a shortened mandible, and reduced proliferation of Meckel's cartilage chondrocytes. This demonstrates a crucial angiocrine role for IGF1 during craniofacial cartilage growth, and identifies IGF1 as a putative therapeutic for jaw and/or cartilage growth disorders.


Subject(s)
Blood Vessels/metabolism , Insulin-Like Growth Factor I/metabolism , Maxillofacial Development/physiology , Animals , Antigens, CD/genetics , Cadherins/deficiency , Cadherins/genetics , Cartilage/cytology , Cartilage/metabolism , Cartilage/pathology , Cell Line , Cell Proliferation , Chondrocytes/cytology , Chondrocytes/metabolism , Embryo, Mammalian/metabolism , Embryonic Development , Insulin-Like Growth Factor I/genetics , Mandible/cytology , Mandible/metabolism , Mice , Mice, Knockout , Signal Transduction , Tissue Culture Techniques , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Wnt1 Protein/deficiency , Wnt1 Protein/genetics
5.
J Anat ; 243(1): 90-99, 2023 07.
Article in English | MEDLINE | ID: mdl-36899483

ABSTRACT

The Hedgehog pathway gene Gli1 has been proposed to mark a subpopulation of skeletal stem cells (SSCs) in craniofacial bone. Skeletal stem cells (SSCs) are multi-potent cells crucial for the development and homeostasis of bone. Recent studies on long bones have suggested that skeletal stem cells in endochondral or intramembranous ossification sites have different differentiation capacities. However, this has not been well-defined in neural crest derived bones. Generally, the long bones are derived from mesoderm and follow an endochondral ossification model, while most of the cranial bones are neural crest (NC) in origin and follow an intramembranous ossification model. The mandible is unique: It is derived from the neural crest lineage but makes use of both modes of ossification. Early in fetal development, the mandibular body is generated by intramembranous ossification with subsequent endochondral ossification forming the condyle. The identities and properties for SSCs in these two sites remain unknown. Here, we use genetic lineage tracing in mouse to identify cells expressing the Hedgehog responsive gene Gli1, which is thought to mark the tissue resident SSCs. We track the Gli1+ cells, comparing cells within the perichondrium to those in the periosteum covering the mandibular body. In juvenile mice, these have distinct differentiation and proliferative potential. We also assess the presence of Sox10+ cells, thought to mark neural crest stem cells, but find no substantial population associated with the mandibular skeleton, suggesting that Sox10+ cells have limited contribution to maintaining postnatal mandibular bone. All together, our study indicates that the Gli1+ cells display distinct and limited differentiation capacity dependent on their regional associations.


Subject(s)
Hedgehog Proteins , Osteogenesis , Mice , Animals , Zinc Finger Protein GLI1/metabolism , Hedgehog Proteins/metabolism , Mandible/metabolism , Skull , Neural Crest
6.
Orthod Craniofac Res ; 26(3): 415-424, 2023 Aug.
Article in English | MEDLINE | ID: mdl-36458927

ABSTRACT

OBJECTIVES: Antidepressants, specifically Selective Serotonin Re-uptake Inhibitors (SSRIs), that alter serotonin metabolism are currently the most commonly prescribed drugs for the treatment of depression. There is some evidence to suggest these drugs contribute to birth defects. As jaw development is often altered in craniofacial birth defects, the purpose of this study was to interrogate the effects of in utero SSRI exposure in a preclinical model of mandible development. MATERIALS AND METHODS: Wild-type C57BL6 mice were used to produce litters that were exposed in utero to an SSRI, Citalopram (500 µg/day). Murine mandibles from P15 pups were analysed for a change in shape and composition. RESULTS: Analysis indicated an overall shape change with total mandibular length and ramus height being shorter in exposed pups as compared to controls. Histomorphometric analysis revealed that first molar length was longer in exposed pups while third molar length was shorter in exposed as compared to control. Histological investigation of molars and surrounding periodontium revealed no change in collagen content of the molar in exposed pups, some alteration in collagen composition in the periodontium, increased alkaline phosphatase in molars and periodontium and decreased mesenchymal cell marker presence in exposed mandibles. CONCLUSION: The results of this study reveal SSRI exposure may interrupt mandible growth as well as overall dental maturation in a model of development giving insight into the expectation that children exposed to SSRIs may require orthodontic intervention.


Subject(s)
Selective Serotonin Reuptake Inhibitors , Serotonin , Animals , Mice , Selective Serotonin Reuptake Inhibitors/adverse effects , Serotonin/metabolism , Mice, Inbred C57BL , Citalopram/adverse effects , Mandible/metabolism
7.
Orthod Craniofac Res ; 26 Suppl 1: 20-28, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37231583

ABSTRACT

OBJECTIVES: Colony-stimulating factor-1 receptor (CSF1R) is vital for the recruitment of monocytes, and their proliferation and differentiation into functional osteoclasts. Mouse studies, where CSF1R and its cognate ligand are absent, have significant craniofacial phenotypes, but these have not been studied in detail. MATERIALS AND METHODS: Pregnant CD1 mice were fed diets laced with CSF1R inhibitor-PLX5622 starting at embryonic day 3.5 (E3.5) up to birth. Pups were collected at E18.5 to study CSF1R expression using immunofluorescence. Additional pups were studied at postnatal day 21 (P21) and P28 using microcomputed tomography (µCT) and Geometric Morphometrics, to evaluate craniofacial form. RESULTS: CSF1R-positive cells were present throughout the developing craniofacial region, including the jaw bones, surrounding teeth, tongue, nasal cavities, brain, cranial vault and base regions. Animals exposed to the CSF1R inhibitor in utero had severe depletion of CSF1R-positive cells at E18.5 and had significant differences in craniofacial form (size and shape) at postnatal timepoints. Centroid sizes for the mandibular and cranio-maxillary regions were significantly smaller in CSF1R-inhibited animals. Proportionally, these animals had a domed skull, with taller and wider cranial vaults and shortening of their midfacial regions. Mandibles were smaller vertically and anterio-posteriorly, with proportionally wider inter-condylar distances. CONCLUSIONS: Embryonic inhibition of CSF1R impacts postnatal craniofacial morphogenesis, with significant influences on the mandibular and cranioskeletal size and shape. These data indicate that CSF1R plays a role in early cranio-skeletal patterning, likely through osteoclast depletion.


Subject(s)
Macrophage Colony-Stimulating Factor , Skull , Animals , Female , Mice , Pregnancy , Macrophage Colony-Stimulating Factor/pharmacology , Mandible/metabolism , Morphogenesis , Skull/diagnostic imaging , Skull/metabolism , X-Ray Microtomography
8.
Dev Biol ; 479: 23-36, 2021 11.
Article in English | MEDLINE | ID: mdl-34332994

ABSTRACT

The mandibular gland is an important exocrine gland of worker bees, which mainly secretes fatty acids and pheromones. Lipids have important roles in energy storage, membrane structure stabilization, and signaling. However, molecular underpinnings of mandibular gland development and lipid remodeling at the different physiological stages of worker bees is still lacking. In this study, we used scanning and transmission electron microscopy to reveal the morphological changes in secretory cells, and liquid chromatography-mass spectrometry and RNA-seq to investigate the lipidome and gene transcripts during development. The morphology of secretory cells was flat in newly emerged workers, becoming vacuolated and turgid when they were activated in nurse bees and foragers. Transport vesicles became denser from newly emerged bees to 21-day worker bees. Concentrations of 10-HDA reached a maximum within 15d workers and changes in genes expression were consistent with 10-HDA content. Non-targeted lipidomics analysis of newly emerged, 6d, and 15d worker bees revealed that PC and TAG were the main lipids in mandibular gland, and lipids dramatically altered across developmental stages. TAG 54:4 was increased most strongly at 6d and 15d worker bees, meanwhile, the abundances of TAG 54:1 and TAG 54:2 were decreased sharply. Further, transcriptomics analysis showed that differentially expressed genes were significantly enriched in key nutrient metabolic pathways, particularly lipid metabolism, in 6d and 15d bees. This multi-omic perspective provides a unique resource and deeper insight into bee mandibular gland development and baseline data for further study of the mandibular gland in worker bees.


Subject(s)
Bees/embryology , Exocrine Glands/embryology , Mandible/embryology , Animals , Bees/metabolism , Behavior, Animal/physiology , Exocrine Glands/metabolism , Gene Expression Profiling/methods , Insect Proteins/genetics , Lipid Metabolism/genetics , Lipidomics/methods , Mandible/metabolism , Metabolic Networks and Pathways , Organogenesis , Proteome/metabolism , Proteomics/methods , Transcriptome/genetics
9.
Oral Dis ; 28(4): 1215-1227, 2022 May.
Article in English | MEDLINE | ID: mdl-33759298

ABSTRACT

OBJECTIVE: In chondrogenesis, BMP signaling was inferred to exhibit regional specificity during Meckel's cartilage morphogenesis. This study aimed to explore the differences in BMP signaling activity between different parts of Meckel's cartilage and the impacts of BMP4 or ALK3 deficiency on the development of Meckel's cartilage during embryogenesis. MATERIALS AND METHODS: The BRE-gal reporter mouse line was utilized to gain an overall picture of canonical BMP signaling activity, as assessed by X-gal staining. Mouse models lacking either Bmp4 or Alk3 in neural crest cells (Wnt1-Cre;Bmp4fl/fl and Wnt1-Cre;Alk3fl/fl ) were generated to explore the morphogenesis of Meckel's cartilage and the mandibular symphysis, as assessed by skeletal staining, histology, and immunostaining. RESULTS: Different parts of Meckel's cartilage exhibited activation of different combinations of BMP signaling pathways. In Wnt1-Cre;Bmp4fl/fl mutants, Sox9+ condensation of the chondrogenic rostral process failed to form, and the V-shaped Runx2+ tissue was split in the median mandibular symphysis. The Wnt1-Cre;Bmp4fl/fl and Wnt1-Cre;Alk3fl/fl mouse models both exhibited truncated Meckel's cartilage, aberrant mandibular intramembranous bone, and tongue muscle abnormalities. CONCLUSIONS: The central hard-tissue loss of both mutant mouse models led to a mandibular symphysis cleft, mimicking the typical sign of the median mandible Tessier 30 cleft in humans.


Subject(s)
Bone Morphogenetic Protein 4 , Bone Morphogenetic Protein Receptors, Type I , Chondrogenesis , Mandible , Animals , Bone Morphogenetic Protein 4/deficiency , Bone Morphogenetic Protein 4/metabolism , Bone Morphogenetic Protein Receptors, Type I/deficiency , Bone Morphogenetic Protein Receptors, Type I/metabolism , Cartilage/metabolism , Mandible/metabolism , Mice , Neural Crest/metabolism , Signal Transduction
10.
Proc Natl Acad Sci U S A ; 116(14): 6954-6963, 2019 04 02.
Article in English | MEDLINE | ID: mdl-30886100

ABSTRACT

Large mandibular defects are clinically challenging to reconstruct due to the complex anatomy of the jaw and the limited availability of appropriate tissue for repair. We envision leveraging current advances in fabrication and biomaterials to create implantable devices that generate bone within the patients themselves suitable for their own specific anatomical pathology. The in vivo bioreactor strategy facilitates the generation of large autologous vascularized bony tissue of customized geometry without the addition of exogenous growth factors or cells. To translate this technology, we investigated its success in reconstructing a mandibular defect of physiologically relevant size in sheep. We fabricated and implanted 3D-printed in vivo bioreactors against rib periosteum and utilized biomaterial-based space maintenance to preserve the native anatomical mandibular structure in the defect site before reconstruction. Nine weeks after bioreactor implantation, the ovine mandibles were repaired with the autologous bony tissue generated from the in vivo bioreactors. We evaluated tissues generated in bioreactors by radiographic, histological, mechanical, and biomolecular assays and repaired mandibles by radiographic and histological assays. Biomaterial-aided mandibular reconstruction was successful in a large superior marginal defect in five of six (83%) sheep. Given that these studies utilized clinically available biomaterials, such as bone cement and ceramic particles, this strategy is designed for rapid human translation to improve outcomes in patients with large mandibular defects.


Subject(s)
Bone Substitutes , Mandible , Mandibular Injuries , Periosteum , Printing, Three-Dimensional , Tissue Engineering , Animals , Bioreactors , Female , Mandible/metabolism , Mandible/pathology , Mandibular Injuries/metabolism , Mandibular Injuries/pathology , Mandibular Injuries/therapy , Periosteum/metabolism , Periosteum/pathology , Sheep
11.
Dev Dyn ; 250(4): 562-573, 2021 04.
Article in English | MEDLINE | ID: mdl-33034111

ABSTRACT

BACKGROUND: Previous studies showed that mice lacking Fgf18 function had cleft palate defects and that the FGF18 locus was associated with cleft lip and palate in humans, but what specific roles Fgf18 plays during palatogenesis are unclear. RESULTS: We show that Fgf18 exhibits regionally restricted expression in developing palatal shelves, mandible, and tongue, during palatal outgrowth and fusion in mouse embryos. Tissue-specific inactivation of Fgf18 throughout neural crest-derived craniofacial mesenchyme caused shortened mandible and reduction in ossification of the frontal, nasal, and anterior cranial base skeletal elements in Fgf18c/c ;Wnt1-Cre mutant mice. About 64% of Fgf18c/c ;Wnt1-Cre mice exhibited cleft palate. Whereas palatal shelf elevation was impaired in many Fgf18c/c ;Wnt1-Cre embryos, no significant difference in palatal cell proliferation was detected between Fgf18c/c ;Wnt1-Cre embryos and their control littermates. Embryonic maxillary explants from Fgf18c/c ;Wnt1-Cre embryos showed successful palatal shelf elevation and fusion in organ culture similar to the maxillary explants from control embryos. Furthermore, tissue-specific inactivation of Fgf18 in the early palatal mesenchyme did not cause cleft palate. CONCLUSION: These results demonstrate a critical role for Fgf18 expression in the neural crest-derived mesenchyme for the development of the mandible and multiple craniofacial bones but Fgf18 expression in the palatal mesenchyme is dispensable for palatogenesis.


Subject(s)
Cleft Palate/etiology , Fibroblast Growth Factors/physiology , Palate/embryology , Animals , Female , Male , Mandible/embryology , Mandible/metabolism , Mesoderm/metabolism , Mice, Knockout , Micrognathism/etiology , Neural Crest/physiology , Palate/metabolism
12.
J Cell Physiol ; 236(6): 4580-4591, 2021 06.
Article in English | MEDLINE | ID: mdl-33251612

ABSTRACT

Bone morphogenetic protein (BMP) signaling is well known in bone homeostasis. However, the physiological effects of BMP signaling on mandibles are largely unknown, as the mandible has distinct functions and characteristics from other bones. In this study, we investigated the roles of BMP signaling in bone homeostasis of the mandibles by deleting BMP type I receptor Acvr1 in osteoblast lineage cells with Osterix-Cre. We found mandibular bone loss in conditional knockout mice at the ages of postnatal day 21 and 42 in an age-dependent manner. The decreased bone mass was related to compromised osteoblast differentiation together with enhanced osteoclastogenesis, which was secondary to the changes in osteoblasts in vivo. In vitro study revealed that deletion of Acvr1 in the mandibular bone marrow stromal cells (BMSCs) significantly compromised osteoblast differentiation. When wild type bone marrow macrophages were cocultured with BMSCs lacking Acvr1 both directly and indirectly, both proliferation and differentiation of osteoclasts were induced as evidenced by an increase of multinucleated cells, compared with cocultured with control BMSCs. Furthermore, we demonstrated that the increased osteoclastogenesis in vitro was at least partially due to the secretion of soluble receptor activator of nuclear factor-κB ligand (sRANKL), which is probably the reason for the mandibular bone loss in vivo. Overall, our results proposed that ACVR1 played essential roles in maintaining mandibular bone homeostasis through osteoblast differentiation and osteoblast-osteoclast communication via sRANKL.


Subject(s)
Activin Receptors, Type I/deficiency , Cell Differentiation , Gene Deletion , Mandible/metabolism , Mesenchymal Stem Cells/metabolism , Osteoblasts/metabolism , Osteoclasts/metabolism , Osteogenesis , RANK Ligand/metabolism , Activin Receptors, Type I/genetics , Animals , Bone Morphogenetic Proteins/metabolism , Bone Resorption , Cell Lineage , Cell Proliferation , Cells, Cultured , Coculture Techniques , Female , Macrophages/metabolism , Male , Mandible/pathology , Mesenchymal Stem Cells/pathology , Mice, Knockout , Osteoblasts/pathology , Osteoclasts/pathology , Signal Transduction
13.
J Anat ; 238(3): 711-719, 2021 03.
Article in English | MEDLINE | ID: mdl-33011977

ABSTRACT

Mandibular anomalies are often seen in various congenital diseases, indicating that mandibular development is under strict molecular control. Therefore, it is crucial to understand the molecular mechanisms involved in mandibular development. MicroRNAs (miRNAs) are noncoding small single-stranded RNAs that play a critical role in regulating the level of gene expression. We found that the mesenchymal conditional deletion of miRNAs arising from a lack of Dicer (an essential molecule for miRNA processing, Dicerfl/fl ;Wnt1Cre), led to an abnormal groove formation at the distal end of developing mandibles. At E10.5, when the region forms, inhibitors of Hh signaling, Ptch1 and Hhip1 showed increased expression at the region in Dicer mutant mandibles, while Gli1 (a major mediator of Hh signaling) was significantly downregulated in mutant mandibles. These suggest that Hh signaling was downregulated at the distal end of Dicer mutant mandibles by increased inhibitors. To understand whether the abnormal groove formation inDicer mutant mandibles was caused by the downregulation of Hh signaling, mice with a mesenchymal deletion of Hh signaling activity arising from a lack of Smo (an essential molecule for Hh signaling activation, Smofl/fl ;Wnt1Cre) were examined. Smofl/fl ;Wnt1Cre mice showed a similar phenotype in the distal region of their mandibles to those in Dicerfl/fl ;Wnt1Cre mice. We also found that approximately 400 miRNAs were expressed in wild-type mandibular mesenchymes at E10.5, and six microRNAs were identified as miRNAs with binding potential against both Ptch1 and Hhip1. Their expressions at the distal end of the mandible were confirmed by in situ hybridization. This indicates that microRNAs regulate the distal part of mandibular formation at an early stage of development by involving Hh signaling activity through controlling its inhibitor expression level.


Subject(s)
Hedgehog Proteins/metabolism , Mandible/growth & development , MicroRNAs/metabolism , Animals , Mandible/metabolism , Mice , Mice, Transgenic
14.
Electrophoresis ; 42(23): 2552-2562, 2021 12.
Article in English | MEDLINE | ID: mdl-34453862

ABSTRACT

Proteomic characterization of alveolar bones in oral surgery represents an analytical challenge due to their insoluble character. The implementation of a straightforward technique could lead to the routine use of proteomics in this field. This work thus developed a simple technique for the characterization of bone tissue for human maxillary and mandibular bones. It is based on the direct in-bone tryptic digestion of proteins in both healthy and pathological human maxillary and mandibular bone samples. The released peptides were then identified by the LC-MS/MS. Using this approach, a total of 1120 proteins were identified in the maxillary bone and 1151 proteins in the mandibular bone. The subsequent partial least squares-discrimination analysis (PLS-DA) of protein data made it possible to reach 100% discrimination between the samples of healthy alveolar bones and those of the bone tissue surrounding the inflammatory focus. These results indicate that the in-bone protein digestion followed by the LC-MS/MS and subsequent statistical analysis can provide a deeper insight into the field of oral surgery at the molecular level. Furthermore, it could also have a diagnostic potential in the differentiation between the proteomic patterns of healthy and pathological alveolar bone tissue. Data are available via ProteomeXchange with the identifier PXD026775.


Subject(s)
Mandible , Maxilla , Oral Surgical Procedures , Proteins , Proteomics , Chromatography, Liquid , Humans , Mandible/metabolism , Maxilla/metabolism , Peptides , Proteins/metabolism , Proteolysis , Tandem Mass Spectrometry , Trypsin/metabolism
15.
Chem Senses ; 462021 01 01.
Article in English | MEDLINE | ID: mdl-33855345

ABSTRACT

We have characterized a recently rediscovered chemosensory structure at the rear of the mandibular mucosa in the mouse oral cavity originally reported in the 1980s. This consists of unorganized taste buds, not contained within troughs, associated with the ducts of an underlying minor salivary gland. Using whole-mount preparations of transgenic mice expressing green fluorescent protein under the promoter of taste-signaling-specific genes, we determined that the structure contains taste bud clusters and salivary gland orifices at the rear of each mandible, distal to the last molar and anterior to the ascending ramus. Immunohistochemical analysis shows in the retromolar taste buds expression of the taste receptors Tas2R131 and T1R3 and taste cascade molecules TrpM5, PLCß2, and GNAT3, consistent with type II taste cells, and expression of GAD1, consistent with type III taste cells. Furthermore, the neuronal marker, calcitonin gene-related peptide, in retromolar mucosa tissue wrapping around TrpM5+ taste buds was observed. RT-PCR showed that retromolar taste buds express all 3 mouse tas1r genes, 28 of the 35 tas2r genes, and taste transduction signaling genes gnat3, plcb2, and trpm5, making the retromolar taste buds similar to other lingual and palate taste buds. Finally, histochemistry demonstrated that the mandibular retromolar secretory gland is a minor salivary gland of mucous type. The mandibular retromolar taste structure may thus play a role in taste sensation and represent a potential novel pharmacological target for taste disorders.


Subject(s)
Mandible/metabolism , Mucus/metabolism , Salivary Glands/metabolism , Taste Buds/metabolism , Animals , Mice , Mice, Inbred C57BL
16.
J Muscle Res Cell Motil ; 42(2): 399-417, 2021 06.
Article in English | MEDLINE | ID: mdl-34255253

ABSTRACT

Ants use their mandibles for a variety of functions and behaviors. We investigated mandibular muscle structure and function from major workers of the Florida carpenter ant Camponotus floridanus: force-pCa relation and velocity of unloaded shortening of single, permeabilized fibres, primary sequences of troponin subunits (TnC, TnI and TnT) from a mandibular muscle cDNA library, and muscle fibre ultrastructure. From the mechanical measurements, we found Ca2+-sensitivity of isometric force was markedly shifted rightward compared with vertebrate striated muscle. From the troponin sequence results, we identified features that could explain the rightward shift of Ca2+-activation: the N-helix of TnC is effectively absent and three of the four EF-hands of TnC (sites I, II and III) do not adhere to canonical sequence rules for divalent cation binding; two alternatively spliced isoforms of TnI were identified with the alternatively spliced exon occurring in the region of the IT-arm α-helical coiled-coil, and the N-terminal extension of TnI may be involved in modulation of regulation, as in mammalian cardiac muscle; and TnT has a Glu-rich C-terminus. In addition, a structural homology model was built of C. floridanus troponin on the thin filament. From analysis of electron micrographs, we found thick filaments are almost as long as the 6.8 µm sarcomeres, have diameter of ~ 16 nm, and typical center-to-center spacing of ~ 46 nm. These results have implications for the mechanisms by which mandibular muscle fibres perform such a variety of functions, and how the structure of the troponin complex aids in these tasks.


Subject(s)
Ants , Troponin C , Animals , Ants/metabolism , Calcium/metabolism , Humans , Invertebrates/metabolism , Mandible/metabolism , Muscle, Skeletal/metabolism , Troponin C/genetics , Troponin C/metabolism , Troponin T/genetics , Troponin T/metabolism
17.
Int J Mol Sci ; 22(23)2021 Dec 02.
Article in English | MEDLINE | ID: mdl-34884839

ABSTRACT

Skeletal class II and III malocclusions are craniofacial disorders that negatively impact people's quality of life worldwide. Unfortunately, the growth patterns of skeletal malocclusions and their clinical correction prognoses are difficult to predict largely due to lack of knowledge of their precise etiology. Inspired by the strong inheritance pattern of a specific type of skeletal malocclusion, previous genome-wide association studies (GWAS) were reanalyzed, resulting in the identification of 19 skeletal class II malocclusion-associated and 53 skeletal class III malocclusion-associated genes. Functional enrichment of these genes created a signal pathway atlas in which most of the genes were associated with bone and cartilage growth and development, as expected, while some were characterized by functions related to skeletal muscle maturation and construction. Interestingly, several genes and enriched pathways are involved in both skeletal class II and III malocclusions, indicating the key regulatory effects of these genes and pathways in craniofacial development. There is no doubt that further investigation is necessary to validate these recognized genes' and pathways' specific function(s) related to maxillary and mandibular development. In summary, this systematic review provides initial insight on developing novel gene-based treatment strategies for skeletal malocclusions and paves the path for precision medicine where dental care providers can make an accurate prediction of the craniofacial growth of an individual patient based on his/her genetic profile.


Subject(s)
Malocclusion/metabolism , Muscle, Skeletal/metabolism , Genome-Wide Association Study , Humans , Malocclusion/genetics , Malocclusion/pathology , Mandible/growth & development , Mandible/metabolism , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , Phospholipase C gamma/genetics , Phospholipase C gamma/metabolism , Polymorphism, Single Nucleotide , Receptor, Fibroblast Growth Factor, Type 2/genetics , Signal Transduction/genetics
18.
Dev Dyn ; 248(12): 1264-1272, 2019 12.
Article in English | MEDLINE | ID: mdl-31464047

ABSTRACT

BACKGROUND: Six1 is a transcriptional factor that plays an important role in embryonic development. Mouse and chick embryos deficient for Six1 have multiple craniofacial anomalies in the facial bones and cartilages. Multiple Six1 enhancers have been identified, but none of them has been reported to be active in the maxillary and mandibular process. RESULTS: We studied two Six1 enhancers in the chick neural crest tissues during craniofacial development. We showed that two evolutionarily conserved enhancers, Six1E1 and Six1E2, act synergistically. Neither Six1E1 nor Six1E2 alone can drive enhancer reporter signal in the maxillary or mandibular processes. However, their combination, Six1E, showed robust enhancer activity in these tissues. Similar reporter signal can also be driven by the mouse homolog of Six1E. Mutations of multiple conserved transcriptional factor binding sites altered the enhancer activity of Six1E, especially mutation of the LIM homeobox binding site, dramatically reduced the enhancer activity, implying that the Lhx protein family be an important regulator of Six1 expression. CONCLUSION: This study, for the first time, described the synergistic activation of two Six1 enhancers in the maxillary and mandibular processes and will facilitate more detailed studies of the regulation of Six1 in craniofacial development.


Subject(s)
Enhancer Elements, Genetic/physiology , Facial Bones/embryology , Homeodomain Proteins/genetics , Neural Crest/embryology , Skull/embryology , Animals , Animals, Genetically Modified , Chick Embryo , Craniofacial Abnormalities/genetics , Embryonic Development/genetics , Facial Bones/metabolism , Gene Expression Regulation, Developmental , Mandible/embryology , Mandible/metabolism , Maxilla/embryology , Maxilla/metabolism , Neural Crest/metabolism , Skull/metabolism
19.
Genesis ; 57(1): e23275, 2019 01.
Article in English | MEDLINE | ID: mdl-30561090

ABSTRACT

The mandibular or first pharyngeal arch forms the upper and lower jaws in all gnathostomes. A gene regulatory network that defines ventral, intermediate, and dorsal domains along the dorsal-ventral (D-V) axis of the arch has emerged from studies in zebrafish and mice, but the temporal dynamics of this process remain unclear. To define cell fate trajectories in the arches we have performed quantitative gene expression analyses of D-V patterning genes in pharyngeal arch primordia in zebrafish and mice. Using NanoString technology to measure transcript numbers per cell directly we show that, in many cases, genes expressed in similar D-V domains and induced by similar signals vary dramatically in their temporal profiles. This suggests that cellular responses to D-V patterning signals are likely shaped by the baseline kinetics of target gene expression. Furthermore, similarities in the temporal dynamics of genes that occupy distinct pathways suggest novel shared modes of regulation. Incorporating these gene expression kinetics into our computational models for the mandibular arch improves the accuracy of patterning, and facilitates temporal comparisons between species. These data suggest that the magnitude and timing of target gene expression help diversify responses to patterning signals during craniofacial development.


Subject(s)
Gene Expression Regulation, Developmental , Mandible/embryology , Transcriptome , Animals , Body Patterning , Mandible/metabolism , Mice , Organogenesis , Zebrafish
20.
J Struct Biol ; 206(1): 128-137, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30849471

ABSTRACT

Teeth are subjected to compressive loads during mastication. Under small loads the soft tissue periodontal ligament (PDL) deforms most. However when the loads increase and the PDL is highly compressed, the tooth and the alveolar bone supporting the tooth, begin to deform. Here we report on the structure of this alveolar bone in the upper furcation region of the first molars of mature minipigs. Using light microscopy and scanning electron microscopy (SEM) of bone cross-sections, we show that this bone is hypermineralized, containing abundant small pores around 1-5 µm in diameter, lacunae around 10-20 µm as well as larger spaces. This bone does not possess the typical lamellar motif or other repeating structures normally found in cortical or trabecular mammalian bone. We also use high resolution focused ion beam scanning electron microscopy (FIB-SEM) in the serial surface mode to image the 3D organization of the demineralized bone matrix. We show that the upper furcation bone matrix has a disordered isotropic structure composed mainly of individual collagen fibrils with no preferred orientation, as well as highly staining material that is probably proteoglycans. Much larger aligned arrays of collagen fibers - presumably Sharpey's fibers - are embedded in this material. This unusual furcation bone material is similar to the disordered material found in human lamellar bone. In the upper furcation region this disordered bone comprises almost all the volume excluding Sharpey's fibers. We surmise that this most unusual bone type functions to resist the repeating compressive loads incurred by molars during mastication.


Subject(s)
Alveolar Process/metabolism , Dental Cementum/chemistry , Mandible/chemistry , Molar/chemistry , Molecular Conformation , Periodontal Ligament/chemistry , Alveolar Process/chemistry , Alveolar Process/pathology , Animals , Collagen/metabolism , Dental Cementum/metabolism , Dental Cementum/ultrastructure , Mandible/metabolism , Mandible/ultrastructure , Microscopy, Electron, Scanning , Molar/metabolism , Molar/ultrastructure , Periodontal Ligament/metabolism , Periodontal Ligament/ultrastructure , Swine , Swine, Miniature , Tooth Demineralization/diagnosis , Tooth Demineralization/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL