1
|
Sohail A, Nicoll O, Bendall AJ. Assessing candidate DLX-regulated genes in the first pharyngeal arch of chick embryos. Dev Dyn 2025. [PMID: 39810614 DOI: 10.1002/dvdy.765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 11/20/2024] [Accepted: 11/29/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Insights into the development and evolution of asymmetrical jaws will require an understanding of the gene regulatory networks that underpin the differential morphogenesis of the maxillary and mandibular domains of the first pharyngeal arch in a variety of gnathostomes. While a robust relationship has been demonstrated between jaw patterning and the Endothelin-Dlx gene axis, much less is known of the next level of genes in the jaw patterning hierarchy. RESULTS Several genes, whose expression depends on Dlx5 and/or Dlx6, have been identified in mice. Here, we examined the expression patterns of the chick orthologues of some of those genes, namely GSC, PITX1, HAND2, and GBX2, and tested their dependence on endothelin signaling to assess whether there is a conserved regulatory relationship between those genes in the chick embryo. To further validate these genes as direct DLX targets, we identified conserved non-coding sequences containing candidate DLX binding motifs and demonstrated DLX-responsiveness in vitro. CONCLUSIONS The evidence presented in this study combines to support the hypothesis that these four genes are direct targets of DLX transcription factors in the lower jaw-forming tissue.
Collapse
Affiliation(s)
- Afshan Sohail
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - Olivia Nicoll
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - Andrew J Bendall
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| |
Collapse
|
2
|
Lovely CB. Bone morphogenetic protein signaling pathway- Ethanol interactions disrupt palate formation independent of gata3. Reprod Toxicol 2025; 131:108754. [PMID: 39586481 PMCID: PMC11634638 DOI: 10.1016/j.reprotox.2024.108754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 11/06/2024] [Accepted: 11/17/2024] [Indexed: 11/27/2024]
Abstract
Fetal Alcohol Spectrum Disorders (FASD) describes a wide array of neurological defects and craniofacial malformations, associated with ethanol teratogenicity. While there is growing evidence for a genetic component to FASD, little is known of the genes underlying these ethanol-induced defects. Along with timing and dosage, genetic predispositions may help explain the variability within FASD. From a screen for gene-ethanol interactions, we found that mutants for Bmp signaling components are ethanol-sensitive leading to defects in the zebrafish palate. Loss of Bmp signaling results in reductions in gata3 expression in the maxillary domain of the neural crest in the 1st pharyngeal arch, leading to palate defects while upregulation of human GATA3 rescues these defects. Here, we show that ethanol-treated Bmp mutants exhibit misshaped and/or broken trabeculae. Surprisingly, up regulation of GATA3 does not rescue ethanol-induced palate defects and gata3 expression was not altered in ethanol-treated Bmp mutants or dorsomorphin-treated larvae. Timing of ethanol sensitivity shows that Bmp mutants are ethanol sensitive from 10 to 18 hours post-fertilization (hpf), prior to Bmp's regulation of gata3 in palate formation. This is consistent with our previous work with dorsomorphin-dependent knock down of Bmp signaling from 10 to 18 hpf disrupting endoderm formation and subsequent jaw development. Overall, this suggests that ethanol disrupts Bmp-dependent palate development independent of and earlier than the role of gata3 in palate formation by disrupting epithelial development. Ultimately, these data demonstrate that zebrafish is a useful model to identify and characterize gene-ethanol interactions and this work will directly inform our understanding of FASD.
Collapse
Affiliation(s)
- C Ben Lovely
- University of Louisville, School of Medicine, Department of Biochemistry and Molecular Genetics, 319 Abraham Flexner Way, Louisville, KY 40202, USA.
| |
Collapse
|
3
|
Lovely CB. Bone Morphogenetic Protein signaling pathway - ethanol interactions disrupt palate formation independent of gata3. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.15.623833. [PMID: 39605565 PMCID: PMC11601317 DOI: 10.1101/2024.11.15.623833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Fetal Alcohol Spectrum Disorders (FASD) describes a wide array of neurological defects and craniofacial malformations, associated with ethanol teratogenicity. While there is growing evidence for a genetic component to FASD, little is known of the genes underlying these ethanol-induced defects. Along with timing and dosage, genetic predispositions may help explain the variability within FASD. From a screen for gene-ethanol interactions, we found that mutants for Bmp signaling components are ethanol-sensitive leading to defects in the zebrafish palate. Loss of Bmp signaling results in reductions in gata3 expression in the maxillary domain of the neural crest in the 1st pharyngeal arch, leading to palate defects while upregulation of human GATA3 rescues these defects. Here, we show that ethanol-treated Bmp mutants exhibit misshaped and/or broken trabeculae. Surprisingly, up regulation of GATA3 does not rescue ethanol-induced palate defects and gata3 expression was not altered in ethanol-treated Bmp mutants or dorsomorphin-treated larvae. Timing of ethanol sensitivity shows that Bmp mutants are ethanol sensitive from 10-18 hours post-fertilization (hpf), prior to Bmp's regulation of gata3 in palate formation. This is consistent with our previous work with dorsomorphin-dependent knock down of Bmp signaling from 10-18 hpf disrupting endoderm formation and subsequent jaw development. Overall, this suggests that ethanol disrupts Bmp-dependent palate development independent of and earlier than the role of gata3 in palate formation by disrupting epithelial development. Ultimately, these data demonstrate that zebrafish is a useful model to identify and characterize gene-ethanol interactions and this work will directly inform our understanding of FASD. Highlights Bmp pathway mutants are ethanol sensitive resulting in palate defects. Ethanol disrupts Bmp-dependent palate development independent of gata3 . Timing of ethanol sensitivity suggests ethanol disrupts Bmp-dependent epithelial morphogenesis.
Collapse
|
4
|
Liu S, Lin Z, Wen S, Teng Y, Xie K, Huang Y. Epidemiological and CBCT characterizations of odontomas: A retrospective study of 87,590 subjects. Oral Dis 2024; 30:4585-4597. [PMID: 38129744 DOI: 10.1111/odi.14845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 11/14/2023] [Accepted: 12/06/2023] [Indexed: 12/23/2023]
Abstract
OBJECTIVES This study aimed to assess the epidemiological and three-dimensional (3D) radiological characterizations of odontomas, as well as the spatial relationship between odontomas and gubernaculum tracts (GT). MATERIALS AND METHODS We retrieved the cone-beam computed tomography (CBCT) data of 87,590 patients. Dentition, location, type, diameter of the odontomas, width of the dental follicle (DF), the spatial relationship between the odontoma and GT, and the influence on adjacent teeth were evaluated. RESULTS Significant differences were found in age, dentition, location, Max/Min diameter, width of DF, impaction, retention, and root bending of adjacent teeth among different spatial relationships between the odontoma and GT (all p < 0.05), as well as in age, type and size, absence, impaction, malposition, and retention of adjacent teeth among different locations of odontomas (all p < 0.05). Compared to the odontomas without impaction, those with impaction had larger diameter (p < 0.05 in all directions). This statistically significant association was consistent for odontomas with malposition, while no similar result was observed in the maximum diameter. CONCLUSION Our findings provide the preliminary data for clinicians to comprehensively understand the incidence, radiographic characterizations and symptoms of odontoma in Chinese population.
Collapse
Affiliation(s)
- Shu Liu
- Department of Radiology, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Zitong Lin
- Department of Radiology, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Shanhui Wen
- Department of Radiology, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yuehui Teng
- Department of Radiology, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Kaipeng Xie
- Department of Public Health, Nanjing Maternity and Child Health Care Hospital, Women's Hospital of Nanjing Medical University, Nanjing, China
| | - Yanhua Huang
- Department of Stomatology, Nanjing Maternity and Child Health Care Hospital, Women's Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
5
|
Chen J, Ying Y, Li H, Sha Z, Lin J, Wu Y, Wu Y, Zhang Y, Chen X, Zhang W. Abnormal dental follicle cells: A crucial determinant in tooth eruption disorders (Review). Mol Med Rep 2024; 30:168. [PMID: 39027997 DOI: 10.3892/mmr.2024.13292] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 06/20/2024] [Indexed: 07/20/2024] Open
Abstract
The dental follicle (DF) plays an indispensable role in tooth eruption by regulating bone remodeling through their influence on osteoblast and osteoclast activity. The process of tooth eruption involves a series of intricate regulatory mechanisms and signaling pathways. Disruption of the parathyroid hormone‑related protein (PTHrP) in the PTHrP‑PTHrP receptor signaling pathway inhibits osteoclast differentiation by DF cells (DFCs), thus resulting in obstructed tooth eruption. Furthermore, parathyroid hormone receptor‑1 mutations are linked to primary tooth eruption failure. Additionally, the Wnt/β‑catenin, TGF‑β, bone morphogenetic protein and Hedgehog signaling pathways have crucial roles in DFC involvement in tooth eruption. DFC signal loss or alteration inhibits osteoclast differentiation, affects osteoblast and cementoblast differentiation, and suppresses DFC proliferation, thus resulting in failed tooth eruptions. Abnormal tooth eruption is also associated with a range of systemic syndromes and genetic diseases, predominantly resulting from pathogenic gene mutations. Among these conditions, the following disorders arise due to genetic mutations that disrupt DFCs and impede proper tooth eruption: Cleidocranial dysplasia associated with Runt‑related gene 2 gene mutations; osteosclerosis caused by CLCN7 gene mutations; mucopolysaccharidosis type VI resulting from arylsulfatase B gene mutations; enamel renal syndrome due to FAM20A gene mutations; and dentin dysplasia caused by mutations in the VPS4B gene. In addition, regional odontodysplasia and multiple calcific hyperplastic DFs are involved in tooth eruption failure; however, they are not related to gene mutations. The specific mechanism for this effect requires further investigation. To the best of our knowledge, previous reviews have not comprehensively summarized the syndromes associated with DF abnormalities manifesting as abnormal tooth eruption. Therefore, the present review aims to consolidate the current knowledge on DFC signaling pathways implicated in abnormal tooth eruption, and their association with disorders of tooth eruption in genetic diseases and syndromes, thereby providing a valuable reference for future related research.
Collapse
Affiliation(s)
- Jiahao Chen
- Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, P.R. China
| | - Ying Ying
- Department of Child Health, Yongkang Women and Children's Health Hospital, Yongkang, Zhejiang 321300, P.R. China
| | - Huimin Li
- Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, P.R. China
| | - Zhuomin Sha
- Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, P.R. China
| | - Jiaqi Lin
- Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, P.R. China
| | - Yongjia Wu
- Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, P.R. China
| | - Yange Wu
- Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, P.R. China
| | - Yun Zhang
- Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, P.R. China
| | - Xuepeng Chen
- Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, P.R. China
| | - Weifang Zhang
- Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, P.R. China
| |
Collapse
|
6
|
Xu C, Xie X, Wu Y, Wang J, Feng JQ. Bone or Tooth dentin: The TGF-β signaling is the key. Int J Biol Sci 2024; 20:3557-3569. [PMID: 38993575 PMCID: PMC11234226 DOI: 10.7150/ijbs.97206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 06/15/2024] [Indexed: 07/13/2024] Open
Abstract
To investigate the cell linkage between tooth dentin and bones, we studied TGF-β roles during postnatal dentin development using TGF-β receptor 2 (Tgfβr2) cKO models and cell lineage tracing approaches. Micro-CT showed that the early Tgfβr2 cKO exhibit short roots and thin root dentin (n = 4; p<0.01), a switch from multilayer pre-odontoblasts/odontoblasts to a single-layer of bone-like cells with a significant loss of ~85% of dentinal tubules (n = 4; p<0.01), and a matrix shift from dentin to bone. Mechanistic studies revealed a statistically significant decrease in odontogenic markers, and a sharp increase in bone markers. The late Tgfβr2 cKO teeth displayed losses of odontoblast polarity, a significant reduction in crown dentin volume, and the onset of massive bone-like structures in the crown pulp with high expression levels of bone markers and low levels of dentin markers. We thus concluded that bones and tooth dentin are in the same evolutionary linkage in which TGF-β signaling defines the odontogenic fate of dental mesenchymal cells and odontoblasts. This finding also raises the possibility of switching the pulp odontogenic to the osteogenic feature of pulp cells via a local manipulation of gene programs in future treatment of tooth fractures.
Collapse
Affiliation(s)
- Chunmei Xu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan China
| | - Xudong Xie
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan China
| | - Yafei Wu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan China
| | - Jun Wang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan China
| | - Jian Q Feng
- Shanxi Medical University School and Hospital of Stomatology, Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, China
| |
Collapse
|
7
|
Quilez S, Dumontier E, Baim C, Kam J, Cloutier JF. Loss of Neogenin alters branchial arch development and leads to craniofacial skeletal defects. Front Cell Dev Biol 2024; 12:1256465. [PMID: 38404688 PMCID: PMC10884240 DOI: 10.3389/fcell.2024.1256465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 01/22/2024] [Indexed: 02/27/2024] Open
Abstract
The formation of complex structures, such as the craniofacial skeleton, requires precise and intricate two-way signalling between populations of cells of different embryonic origins. For example, the lower jaw, or mandible, arises from cranial neural crest cells (CNCCs) in the mandibular portion of the first branchial arch (mdBA1) of the embryo, and its development is regulated by signals from the ectoderm and cranial mesoderm (CM) within this structure. The molecular mechanisms underlying CM cell influence on CNCC development in the mdBA1 remain poorly defined. Herein we identified the receptor Neogenin as a key regulator of craniofacial development. We found that ablation of Neogenin expression via gene-targeting resulted in several craniofacial skeletal defects, including reduced size of the CNCC-derived mandible. Loss of Neogenin did not affect the formation of the mdBA1 CM core but resulted in altered Bmp4 and Fgf8 expression, increased apoptosis, and reduced osteoblast differentiation in the mdBA1 mesenchyme. Reduced BMP signalling in the mdBA1 of Neogenin mutant embryos was associated with alterations in the gene regulatory network, including decreased expression of transcription factors of the Hand, Msx, and Alx families, which play key roles in the patterning and outgrowth of the mdBA1. Tissue-specific Neogenin loss-of-function studies revealed that Neogenin expression in mesodermal cells contributes to mandible formation. Thus, our results identify Neogenin as a novel regulator of craniofacial skeletal formation and demonstrates it impinges on CNCC development via a non-cell autonomous mechanism.
Collapse
Affiliation(s)
- Sabrina Quilez
- The Neuro—Montreal Neurological Institute and Hospital, 3801 University, Montréal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada
| | - Emilie Dumontier
- The Neuro—Montreal Neurological Institute and Hospital, 3801 University, Montréal, QC, Canada
| | - Christopher Baim
- The Neuro—Montreal Neurological Institute and Hospital, 3801 University, Montréal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada
| | - Joseph Kam
- The Neuro—Montreal Neurological Institute and Hospital, 3801 University, Montréal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada
| | - Jean-François Cloutier
- The Neuro—Montreal Neurological Institute and Hospital, 3801 University, Montréal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada
- Department of Anatomy and Cell Biology, McGill University, Montréal, QC, Canada
| |
Collapse
|
8
|
Alexander BE, Zhao H, Astrof S. SMAD4: A critical regulator of cardiac neural crest cell fate and vascular smooth muscle development. Dev Dyn 2024; 253:119-143. [PMID: 37650555 PMCID: PMC10842824 DOI: 10.1002/dvdy.652] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 06/07/2023] [Accepted: 08/09/2023] [Indexed: 09/01/2023] Open
Abstract
BACKGROUND During embryogenesis, cardiac neural crest-derived cells (NCs) migrate into the pharyngeal arches and give rise to the vascular smooth muscle cells (vSMCs) of the pharyngeal arch arteries (PAAs). vSMCs are critical for the remodeling of the PAAs into their final adult configuration, giving rise to the aortic arch and its arteries (AAAs). RESULTS We investigated the role of SMAD4 in NC-to-vSMC differentiation using lineage-specific inducible mouse strains. We found that the expression of SMAD4 in the NC is indelible for regulating the survival of cardiac NCs. Although the ablation of SMAD4 at E9.5 in the NC lineage led to a near-complete absence of NCs in the pharyngeal arches, PAAs became invested with vSMCs derived from a compensatory source. Analysis of AAA development at E16.5 showed that the alternative vSMC source compensated for the lack of NC-derived vSMCs and rescued AAA morphogenesis. CONCLUSIONS Our studies uncovered the requisite role of SMAD4 in the contribution of the NC to the pharyngeal arch mesenchyme. We found that in the absence of SMAD4+ NCs, vSMCs around the PAAs arose from a different progenitor source, rescuing AAA morphogenesis. These findings shed light on the remarkable plasticity of developmental mechanisms governing AAA development.
Collapse
Affiliation(s)
- Brianna E. Alexander
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, 07103
- Multidisciplinary Ph.D. Program in Biomedical Sciences: Cell Biology, Neuroscience and Physiology Track, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, 07103
| | - Huaning Zhao
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, 07103
| | - Sophie Astrof
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, 07103
- Multidisciplinary Ph.D. Program in Biomedical Sciences: Cell Biology, Neuroscience and Physiology Track, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, 07103
- Multidisciplinary Ph.D. Program in Biomedical Sciences: Molecular Biology, Genetics, and Cancer Track, New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, 07103
| |
Collapse
|
9
|
Kearney M, Cooper PR, Smith AJ, Duncan HF. Characterisation of miRNA Expression in Dental Pulp Cells during Epigenetically-Driven Reparative Processes. Int J Mol Sci 2023; 24:ijms24108631. [PMID: 37239975 DOI: 10.3390/ijms24108631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 05/08/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
Within regenerative endodontics, exciting opportunities exist for the development of next-generation targeted biomaterials that harness epigenetic machinery, including microRNAs (miRNAs), histone acetylation, and DNA methylation, which are used to control pulpitis and to stimulate repair. Although histone deacetylase inhibitors (HDACi) and DNA methyltransferase inhibitors (DNMTi) induce mineralisation in dental pulp cell (DPC) populations, their interaction with miRNAs during DPC mineralisation is not known. Here, small RNA sequencing and bioinformatic analysis were used to establish a miRNA expression profile for mineralising DPCs in culture. Additionally, the effects of a HDACi, suberoylanilide hydroxamic acid (SAHA), and a DNMTi, 5-aza-2'-deoxycytidine (5-AZA-CdR), on miRNA expression, as well as DPC mineralisation and proliferation, were analysed. Both inhibitors increased mineralisation. However, they reduced cell growth. Epigenetically-enhanced mineralisation was accompanied by widespread changes in miRNA expression. Bioinformatic analysis identified many differentially expressed mature miRNAs that were suggested to have roles in mineralisation and stem cell differentiation, including regulation of the Wnt and MAPK pathways. Selected candidate miRNAs were demonstrated by qRT-PCR to be differentially regulated at various time points in mineralising DPC cultures treated with SAHA or 5-AZA-CdR. These data validated the RNA sequencing analysis and highlighted an increased and dynamic interaction between miRNA and epigenetic modifiers during the DPC reparative processes.
Collapse
Affiliation(s)
- Michaela Kearney
- Division of Restorative Dentistry & Periodontology, Dublin Dental University Hospital, Trinity College Dublin, University of Dublin, D02 F859 Dublin, Ireland
| | - Paul R Cooper
- Department of Oral Sciences, Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin 9016, New Zealand
| | - Anthony J Smith
- Oral Biology, School of Dentistry, University of Birmingham, Birmingham B5 7EG, UK
| | - Henry F Duncan
- Division of Restorative Dentistry & Periodontology, Dublin Dental University Hospital, Trinity College Dublin, University of Dublin, D02 F859 Dublin, Ireland
| |
Collapse
|
10
|
Alexander BE, Zhao H, Astrof S. SMAD4: A Critical Regulator of Cardiac Neural Crest Cell Fate and Vascular Smooth Muscle Differentiation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.14.532676. [PMID: 36993156 PMCID: PMC10055180 DOI: 10.1101/2023.03.14.532676] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Background The pharyngeal arch arteries (PAAs) are precursor vessels which remodel into the aortic arch arteries (AAAs) during embryonic cardiovascular development. Cardiac neural crest cells (NCs) populate the PAAs and differentiate into vascular smooth muscle cells (vSMCs), which is critical for successful PAA-to-AAA remodeling. SMAD4, the central mediator of canonical TGFβ signaling, has been implicated in NC-to-vSMC differentiation; however, its distinct roles in vSMC differentiation and NC survival are unclear. Results Here, we investigated the role of SMAD4 in cardiac NC differentiation to vSMCs using lineage-specific inducible mouse strains in an attempt to avoid early embryonic lethality and NC cell death. We found that with global SMAD4 loss, its role in smooth muscle differentiation could be uncoupled from its role in the survival of the cardiac NC in vivo . Moreover, we found that SMAD4 may regulate the induction of fibronectin, a known mediator of NC-to-vSMC differentiation. Finally, we found that SMAD4 is required in NCs cell-autonomously for NC-to-vSMC differentiation and for NC contribution to and persistence in the pharyngeal arch mesenchyme. Conclusions Overall, this study demonstrates the critical role of SMAD4 in the survival of cardiac NCs, their differentiation to vSMCs, and their contribution to the developing pharyngeal arches.
Collapse
|
11
|
Zhang H, Gong X, Xu X, Wang X, Sun Y. Tooth number abnormality: from bench to bedside. Int J Oral Sci 2023; 15:5. [PMID: 36604408 PMCID: PMC9816303 DOI: 10.1038/s41368-022-00208-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 09/24/2022] [Accepted: 11/01/2022] [Indexed: 01/07/2023] Open
Abstract
Tooth number abnormality is one of the most common dental developmental diseases, which includes both tooth agenesis and supernumerary teeth. Tooth development is regulated by numerous developmental signals, such as the well-known Wnt, BMP, FGF, Shh and Eda pathways, which mediate the ongoing complex interactions between epithelium and mesenchyme. Abnormal expression of these crutial signalling during this process may eventually lead to the development of anomalies in tooth number; however, the underlying mechanisms remain elusive. In this review, we summarized the major process of tooth development, the latest progress of mechanism studies and newly reported clinical investigations of tooth number abnormality. In addition, potential treatment approaches for tooth number abnormality based on developmental biology are also discussed. This review not only provides a reference for the diagnosis and treatment of tooth number abnormality in clinical practice but also facilitates the translation of basic research to the clinical application.
Collapse
Affiliation(s)
- Han Zhang
- grid.24516.340000000123704535Department of Implantology, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - Xuyan Gong
- grid.24516.340000000123704535Department of Implantology, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - Xiaoqiao Xu
- grid.24516.340000000123704535Department of Implantology, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - Xiaogang Wang
- grid.64939.310000 0000 9999 1211Key Laboratory of Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, Beijing, China
| | - Yao Sun
- Department of Implantology, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China.
| |
Collapse
|
12
|
Liao J, Huang Y, Wang Q, Chen S, Zhang C, Wang D, Lv Z, Zhang X, Wu M, Chen G. Gene regulatory network from cranial neural crest cells to osteoblast differentiation and calvarial bone development. Cell Mol Life Sci 2022; 79:158. [PMID: 35220463 PMCID: PMC11072871 DOI: 10.1007/s00018-022-04208-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 02/02/2022] [Accepted: 02/14/2022] [Indexed: 11/03/2022]
Abstract
Calvarial bone is one of the most complex sequences of developmental events in embryology, featuring a uniquely transient, pluripotent stem cell-like population known as the cranial neural crest (CNC). The skull is formed through intramembranous ossification with distinct tissue lineages (e.g. neural crest derived frontal bone and mesoderm derived parietal bone). Due to CNC's vast cell fate potential, in response to a series of inductive secreted cues including BMP/TGF-β, Wnt, FGF, Notch, Hedgehog, Hippo and PDGF signaling, CNC enables generations of a diverse spectrum of differentiated cell types in vivo such as osteoblasts and chondrocytes at the craniofacial level. In recent years, since the studies from a genetic mouse model and single-cell sequencing, new discoveries are uncovered upon CNC patterning, differentiation, and the contribution to the development of cranial bones. In this review, we summarized the differences upon the potential gene regulatory network to regulate CNC derived osteogenic potential in mouse and human, and highlighted specific functions of genetic molecules from multiple signaling pathways and the crosstalk, transcription factors and epigenetic factors in orchestrating CNC commitment and differentiation into osteogenic mesenchyme and bone formation. Disorders in gene regulatory network in CNC patterning indicate highly close relevance to clinical birth defects and diseases, providing valuable transgenic mouse models for subsequent discoveries in delineating the underlying molecular mechanisms. We also emphasized the potential regenerative alternative through scientific discoveries from CNC patterning and genetic molecules in interfering with or alleviating clinical disorders or diseases, which will be beneficial for the molecular targets to be integrated for novel therapeutic strategies in the clinic.
Collapse
Affiliation(s)
- Junguang Liao
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Yuping Huang
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Qiang Wang
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Sisi Chen
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Chenyang Zhang
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Dan Wang
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Zhengbing Lv
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Xingen Zhang
- Department of Orthopedics, Jiaxing Key Laboratory for Minimally Invasive Surgery in Orthopaedics & Skeletal Regenerative Medicine, Zhejiang Rongjun Hospital, Jiaxing, 314001, China
| | - Mengrui Wu
- Institute of Genetics, College of Life Science, Zhejiang University, Hangzhou, 310058, China
| | - Guiqian Chen
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China.
- Institute of Genetics, College of Life Science, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
13
|
Stanwick M, Barkley C, Serra R, Kruggel A, Webb A, Zhao Y, Pietrzak M, Ashman C, Staats A, Shahid S, Peters SB. Tgfbr2 in Dental Pulp Cells Guides Neurite Outgrowth in Developing Teeth. Front Cell Dev Biol 2022; 10:834815. [PMID: 35265620 PMCID: PMC8901236 DOI: 10.3389/fcell.2022.834815] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 01/24/2022] [Indexed: 11/13/2022] Open
Abstract
Transforming growth factor β (TGFβ) plays an important role in tooth morphogenesis and mineralization. During postnatal development, the dental pulp (DP) mesenchyme secretes neurotrophic factors that guide trigeminal nerve fibers into and throughout the DP. This process is tightly linked with dentin formation and mineralization. Our laboratory established a mouse model in which Tgfbr2 was conditionally deleted in DP mesenchyme using an Osterix promoter-driven Cre recombinase (Tgfbr2 cko ). These mice survived postnatally with significant defects in bones and teeth, including reduced mineralization and short roots. Hematoxylin and eosin staining revealed reduced axon-like structures in the mutant mice. Reporter imaging demonstrated that Osterix-Cre activity within the tooth was active in the DP and derivatives, but not in neuronal afferents. Immunofluorescence staining for β3 tubulin (neuronal marker) was performed on serial cryosections from control and mutant molars on postnatal days 7 and 24 (P7, P24). Confocal imaging and pixel quantification demonstrated reduced innervation in Tgfbr2 cko first molars at both stages compared to controls, indicating that signals necessary to promote neurite outgrowth were disrupted by Tgfbr2 deletion. We performed mRNA-Sequence (RNA-Seq) and gene onotology analyses using RNA from the DP of P7 control and mutant mice to investigate the pathways involved in Tgfbr2-mediated tooth development. These analyses identified downregulation of several mineralization-related and neuronal genes in the Tgfbr2 cko DP compared to controls. Select gene expression patterns were confirmed by quantitative real-time PCR and immunofluorescence imaging. Lastly, trigeminal neurons were co-cultured atop Transwell filters overlying primary Tgfbr2 f/f DP cells. Tgfbr2 in the DP was deleted via Adenovirus-expressed Cre recombinase. Confocal imaging of axons through the filter pores showed increased axonal sprouting from neurons cultured with Tgfbr2-positive DP cells compared to neurons cultured alone. Axon sprouting was reduced when Tgfbr2 was knocked down in the DP cells. Immunofluorescence of dentin sialophosphoprotein in co-cultured DP cells confirmed reduced mineralization potential in cells with Tgfbr2 deletion. Both our proteomics and RNA-Seq analyses indicate that axonal guidance cues, particularly semaphorin signaling, were disrupted by Tgfbr2 deletion. Thus, Tgfbr2 in the DP mesenchyme appears to regulate differentiation and the cells' ability to guide neurite outgrowth during tooth mineralization and innervation.
Collapse
Affiliation(s)
- Monica Stanwick
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, United States
| | - Courtney Barkley
- Department of Cell Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Rosa Serra
- Department of Cell Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Andrew Kruggel
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, United States
| | - Amy Webb
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, United States
| | - Yue Zhao
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, United States
| | - Maciej Pietrzak
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, United States
| | - Chandler Ashman
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, United States
| | - Allie Staats
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, United States
| | - Shifa Shahid
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, United States
| | - Sarah B. Peters
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, United States,Department of Cell Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, United States,*Correspondence: Sarah B. Peters,
| |
Collapse
|
14
|
Cappuccio G, Brunetti-Pierri N, Clift P, Learn C, Dykes JC, Mercer CL, Callewaert B, Meerschaut I, Spinelli AM, Bruno I, Gillespie MJ, Dorfman AT, Grimberg A, Lindsay ME, Lin AE. Expanded cardiovascular phenotype of Myhre syndrome includes tetralogy of Fallot suggesting a role for SMAD4 in human neural crest defects. Am J Med Genet A 2022; 188:1384-1395. [PMID: 35025139 DOI: 10.1002/ajmg.a.62645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 11/16/2021] [Accepted: 12/11/2021] [Indexed: 11/08/2022]
Abstract
Tetralogy of Fallot (ToF) can be associated with a wide range of extracardiac anomalies, with an underlying etiology identified in approximately 10% of cases. Individuals affected with Myhre syndrome due to recurrent SMAD4 mutations frequently have cardiovascular anomalies, including congenital heart defects. In addition to two patients in the literature with ToF, we describe five additional individuals with Myhre syndrome and classic ToF, ToF with pulmonary atresia and multiple aorto-pulmonary collaterals, and ToF with absent pulmonary valve. Aorta hypoplasia was documented in one patient and suspected in another two. In half of these individuals, postoperative cardiac dysfunction was thought to be more severe than classic postoperative ToF repair. There may be an increase in right ventricular pressure, and right ventricular dysfunction due to free pulmonic regurgitation. Noncardiac developmental abnormalities in our series and the literature, including corectopia, heterochromia iridis, and congenital miosis suggest an underlying defect of neural crest cell migration in Myhre syndrome. We advise clinicians that Myhre syndrome should be considered in the genetic evaluation of a child with ToF, short stature, unusual facial features, and developmental delay, as these children may be at risk for increased postoperative morbidity. Additional research is needed to investigate the hypothesis that postoperative hemodynamics in these patients may be consistent with restrictive myocardial physiology.
Collapse
Affiliation(s)
- Gerarda Cappuccio
- Department of Translational Medicine, Section of Pediatrics, Federico II University, Naples, Italy.,Telethon Institute of Genetics and Medicine, Pozzuoli (Naples), Italy
| | - Nicola Brunetti-Pierri
- Department of Translational Medicine, Section of Pediatrics, Federico II University, Naples, Italy.,Telethon Institute of Genetics and Medicine, Pozzuoli (Naples), Italy
| | - Paul Clift
- Adult Congenital Heart Disease Unit, University Hospitals Birmingham, Birmingham, UK
| | - Christopher Learn
- Adult Congenital Heart Disease Program, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - John C Dykes
- Departments of Pediatrics, Stanford, California, USA
| | - Catherine L Mercer
- Wessex Clinical Genetics Service, University Hospital Southampton NHS Foundation Trust, Princess Anne Hospital, Southampton, UK
| | - Bert Callewaert
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium.,Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
| | - Ilse Meerschaut
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium.,Department of Pediatrics, Ghent University Hospital, Ghent, Belgium
| | | | - Irene Bruno
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, Trieste, Italy
| | - Matthew J Gillespie
- Division of Cardiology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Aaron T Dorfman
- Division of Cardiology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Adda Grimberg
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Division of Endocrinology and Diabetes, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Mark E Lindsay
- Department of Pediatrics, Division of Pediatric Cardiology, Massachusetts General Hospital, Boston, Massachusetts, USA.,Cardiovascular Research Center, Division of Cardiology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Angela E Lin
- Genetics Unit, Department of Pediatrics, MassGeneral Hospital for Children, Boston, Massachusetts, USA
| |
Collapse
|
15
|
Fan X, Loebel DAF, Bildsoe H, Wilkie EE, Qin J, Wang J, Tam PPL. Tissue interactions, cell signaling and transcriptional control in the cranial mesoderm during craniofacial development. AIMS GENETICS 2021. [DOI: 10.3934/genet.2016.1.74] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
AbstractThe cranial neural crest and the cranial mesoderm are the source of tissues from which the bone and cartilage of the skull, face and jaws are constructed. The development of the cranial mesoderm is not well studied, which is inconsistent with its importance in craniofacial morphogenesis as a source of precursor tissue of the chondrocranium, muscles, vasculature and connective tissues, mechanical support for tissue morphogenesis, and the signaling activity that mediate interactions with the cranial neural crest. Phenotypic analysis of conditional knockout mouse mutants, complemented by the transcriptome analysis of differentially enriched genes in the cranial mesoderm and cranial neural crest, have identified signaling pathways that may mediate cross-talk between the two tissues. In the cranial mesenchyme, Bmp4 is expressed in the mesoderm cells while its signaling activity could impact on both the mesoderm and the neural crest cells. In contrast, Fgf8 is predominantly expressed in the cranial neural crest cells and it influences skeletal development and myogenesis in the cranial mesoderm. WNT signaling, which emanates from the cranial neural crest cells, interacts with BMP and FGF signaling in monitoring the switch between tissue progenitor expansion and differentiation. The transcription factor Twist1, a critical molecular regulator of many aspects of craniofacial development, coordinates the activity of the above pathways in cranial mesoderm and cranial neural crest tissue compartments.
Collapse
Affiliation(s)
- Xiaochen Fan
- Embryology Unit, Children's Medical Research Institute, Westmead NSW 2145, Australia
| | - David A F Loebel
- Embryology Unit, Children's Medical Research Institute, Westmead NSW 2145, Australia
| | - Heidi Bildsoe
- Embryology Unit, Children's Medical Research Institute, Westmead NSW 2145, Australia
| | - Emilie E Wilkie
- Embryology Unit, Children's Medical Research Institute, Westmead NSW 2145, Australia
- Bioinformatics Group, Children's Medical Research Institute, Westmead NSW 2145, Australia
| | - Jing Qin
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, NT, Hong Kong
| | - Junwen Wang
- Department of Health Sciences Research, Center for Individualized Medicine, Mayo Clinic, and Department of Biomedical Informatics, Arizona State University, Scottsdale AZ 85259, USA
| | - Patrick P L Tam
- Embryology Unit, Children's Medical Research Institute, Westmead NSW 2145, Australia
- School of Medical Sciences, Sydney Medical School, University of Sydney, NSW 2006, Australia
| |
Collapse
|
16
|
Prasad MS, Charney RM, Patel LJ, García-Castro MI. Distinct molecular profile and restricted stem cell potential defines the prospective human cranial neural crest from embryonic stem cell state. Stem Cell Res 2020; 49:102086. [PMID: 33370869 PMCID: PMC7932500 DOI: 10.1016/j.scr.2020.102086] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 06/14/2020] [Accepted: 11/04/2020] [Indexed: 12/18/2022] Open
Abstract
Neural crest cells are an embryonic multipotent stem cell population. Recent studies in model organisms have suggested that neural crest cells are specified earlier than previously thought, at blastula stages. However, the molecular dynamics of early neural crest specification, and functional changes from pluripotent precursors to early specified NC, remain to be elucidated. In this report, we utilized a robust human model of cranial neural crest formation to address the distinct molecular character of the earliest stages of neural crest specification and assess the functional differences from its embryonic stem cell precursor. Our human neural crest model reveals a rapid change in the epigenetic state of neural crest and pluripotency genes, accompanied by changes in gene expression upon Wnt-based induction from embryonic stem cells. These changes in gene expression are directly regulated by the transcriptional activity of β-catenin. Furthermore, prospective cranial neural crest cells are characterized by restricted stem cell potential compared to embryonic stem cells. Our results suggest that human neural crest induced by Wnt/β-catenin signaling from human embryonic stem cells rapidly acquire a prospective neural crest cell state defined by a unique molecular signature and endowed with limited potential compared to pluripotent stem cells.
Collapse
Affiliation(s)
- Maneeshi S Prasad
- School of Medicine Division of Biomedical Sciences, University of California, Riverside, USA.
| | - Rebekah M Charney
- School of Medicine Division of Biomedical Sciences, University of California, Riverside, USA
| | - Lipsa J Patel
- School of Medicine Division of Biomedical Sciences, University of California, Riverside, USA
| | - Martín I García-Castro
- School of Medicine Division of Biomedical Sciences, University of California, Riverside, USA.
| |
Collapse
|
17
|
Reynolds K, Zhang S, Sun B, Garland M, Ji Y, Zhou CJ. Genetics and signaling mechanisms of orofacial clefts. Birth Defects Res 2020; 112:1588-1634. [PMID: 32666711 PMCID: PMC7883771 DOI: 10.1002/bdr2.1754] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 06/11/2020] [Accepted: 06/15/2020] [Indexed: 12/31/2022]
Abstract
Craniofacial development involves several complex tissue movements including several fusion processes to form the frontonasal and maxillary structures, including the upper lip and palate. Each of these movements are controlled by many different factors that are tightly regulated by several integral morphogenetic signaling pathways. Subject to both genetic and environmental influences, interruption at nearly any stage can disrupt lip, nasal, or palate fusion and result in a cleft. Here, we discuss many of the genetic risk factors that may contribute to the presentation of orofacial clefts in patients, and several of the key signaling pathways and underlying cellular mechanisms that control lip and palate formation, as identified primarily through investigating equivalent processes in animal models, are examined.
Collapse
Affiliation(s)
- Kurt Reynolds
- Department of Biochemistry and Molecular Medicine, University of California at Davis, School of Medicine, Sacramento, CA 95817
- Institute for Pediatric Regenerative Medicine of Shriners Hospitals for Children, University of California at Davis, School of Medicine, Sacramento, CA 95817
- Biochemistry, Molecular, Cellular, and Developmental Biology (BMCDB) graduate group, University of California, Davis, CA 95616
| | - Shuwen Zhang
- Department of Biochemistry and Molecular Medicine, University of California at Davis, School of Medicine, Sacramento, CA 95817
- Institute for Pediatric Regenerative Medicine of Shriners Hospitals for Children, University of California at Davis, School of Medicine, Sacramento, CA 95817
| | - Bo Sun
- Department of Biochemistry and Molecular Medicine, University of California at Davis, School of Medicine, Sacramento, CA 95817
- Institute for Pediatric Regenerative Medicine of Shriners Hospitals for Children, University of California at Davis, School of Medicine, Sacramento, CA 95817
| | - Michael Garland
- Department of Biochemistry and Molecular Medicine, University of California at Davis, School of Medicine, Sacramento, CA 95817
- Institute for Pediatric Regenerative Medicine of Shriners Hospitals for Children, University of California at Davis, School of Medicine, Sacramento, CA 95817
| | - Yu Ji
- Department of Biochemistry and Molecular Medicine, University of California at Davis, School of Medicine, Sacramento, CA 95817
- Institute for Pediatric Regenerative Medicine of Shriners Hospitals for Children, University of California at Davis, School of Medicine, Sacramento, CA 95817
- Biochemistry, Molecular, Cellular, and Developmental Biology (BMCDB) graduate group, University of California, Davis, CA 95616
| | - Chengji J. Zhou
- Department of Biochemistry and Molecular Medicine, University of California at Davis, School of Medicine, Sacramento, CA 95817
- Institute for Pediatric Regenerative Medicine of Shriners Hospitals for Children, University of California at Davis, School of Medicine, Sacramento, CA 95817
- Biochemistry, Molecular, Cellular, and Developmental Biology (BMCDB) graduate group, University of California, Davis, CA 95616
| |
Collapse
|
18
|
Chen G, Xu H, Yao Y, Xu T, Yuan M, Zhang X, Lv Z, Wu M. BMP Signaling in the Development and Regeneration of Cranium Bones and Maintenance of Calvarial Stem Cells. Front Cell Dev Biol 2020; 8:135. [PMID: 32211409 PMCID: PMC7075941 DOI: 10.3389/fcell.2020.00135] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 02/18/2020] [Indexed: 12/13/2022] Open
Abstract
The bone morphogenetic protein (BMP) signaling pathway is highly conserved across many species, and its importance for the patterning of the skeletal system has been demonstrated. A disrupted BMP signaling pathway results in severe skeletal defects. Murine calvaria has been identified to have dual-tissue lineages, namely, the cranial neural-crest cells and the paraxial mesoderm. Modulations of the BMP signaling pathway have been demonstrated to be significant in determining calvarial osteogenic potentials and ossification in vitro and in vivo. More importantly, the BMP signaling pathway plays a role in the maintenance of the homeostasis of the calvarial stem cells, indicating a potential clinic significance in calvarial bone and in expediting regeneration. Following the inherent evidence of BMP signaling in craniofacial biology, we summarize recent discoveries relating to BMP signaling in the development of calvarial structures, functions of the suture stem cells and their niche and regeneration. This review will not only provide a better understanding of BMP signaling in cranial biology, but also exhibit the molecular targets of BMP signaling that possess clinical potential for tissue regeneration.
Collapse
Affiliation(s)
- Guiqian Chen
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Haodong Xu
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Yifeng Yao
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Tingting Xu
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Mengting Yuan
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Xingen Zhang
- Department of Orthopedics, Zhejiang Rongjun Hospital, Jiaxing, China
| | - Zhengbing Lv
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Mengrui Wu
- Institute of Genetics, Life Science College, Zhejiang University, Hangzhou, China
| |
Collapse
|
19
|
Zanella M, Vitriolo A, Andirko A, Martins PT, Sturm S, O’Rourke T, Laugsch M, Malerba N, Skaros A, Trattaro S, Germain PL, Mihailovic M, Merla G, Rada-Iglesias A, Boeckx C, Testa G. Dosage analysis of the 7q11.23 Williams region identifies BAZ1B as a major human gene patterning the modern human face and underlying self-domestication. SCIENCE ADVANCES 2019; 5:eaaw7908. [PMID: 31840056 PMCID: PMC6892627 DOI: 10.1126/sciadv.aaw7908] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 09/26/2019] [Indexed: 05/10/2023]
Abstract
We undertook a functional dissection of chromatin remodeler BAZ1B in neural crest (NC) stem cells (NCSCs) from a uniquely informative cohort of typical and atypical patients harboring 7q11.23 copy number variants. Our results reveal a key contribution of BAZ1B to NCSC in vitro induction and migration, coupled with a crucial involvement in NC-specific transcriptional circuits and distal regulation. By intersecting our experimental data with new paleogenetic analyses comparing modern and archaic humans, we found a modern-specific enrichment for regulatory changes both in BAZ1B and its experimentally defined downstream targets, thereby providing the first empirical validation of the human self-domestication hypothesis and positioning BAZ1B as a master regulator of the modern human face. In so doing, we provide experimental evidence that the craniofacial and cognitive/behavioral phenotypes caused by alterations of the Williams-Beuren syndrome critical region can serve as a powerful entry point into the evolution of the modern human face and prosociality.
Collapse
Affiliation(s)
- Matteo Zanella
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Laboratory of Stem Cell Epigenetics, IEO, European Institute of Oncology, IRCCS, Milan, Italy
| | - Alessandro Vitriolo
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Laboratory of Stem Cell Epigenetics, IEO, European Institute of Oncology, IRCCS, Milan, Italy
| | - Alejandro Andirko
- University of Barcelona, Barcelona, Spain
- University of Barcelona Institute of Complex Systems (UBICS), Barcelona, Spain
| | - Pedro Tiago Martins
- University of Barcelona, Barcelona, Spain
- University of Barcelona Institute of Complex Systems (UBICS), Barcelona, Spain
| | - Stefanie Sturm
- University of Barcelona, Barcelona, Spain
- University of Barcelona Institute of Complex Systems (UBICS), Barcelona, Spain
| | - Thomas O’Rourke
- University of Barcelona, Barcelona, Spain
- University of Barcelona Institute of Complex Systems (UBICS), Barcelona, Spain
| | - Magdalena Laugsch
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
- Institute of Human Genetics, University Hospital Cologne, Cologne, Germany
- Institute of Human Genetics, University Hospital Heidelberg, Heidelberg, Germany
| | - Natascia Malerba
- Division of Medical Genetics, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Foggia, Italy
| | - Adrianos Skaros
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Laboratory of Stem Cell Epigenetics, IEO, European Institute of Oncology, IRCCS, Milan, Italy
| | - Sebastiano Trattaro
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Laboratory of Stem Cell Epigenetics, IEO, European Institute of Oncology, IRCCS, Milan, Italy
| | - Pierre-Luc Germain
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Laboratory of Stem Cell Epigenetics, IEO, European Institute of Oncology, IRCCS, Milan, Italy
- D-HEST Institute for Neuroscience, ETH Zürich, Switzerland
| | - Marija Mihailovic
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Laboratory of Stem Cell Epigenetics, IEO, European Institute of Oncology, IRCCS, Milan, Italy
| | - Giuseppe Merla
- Division of Medical Genetics, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Foggia, Italy
| | - Alvaro Rada-Iglesias
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
- Cluster of Excellence Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Germany
- Institute of Biomedicine and Biotechnology of Cantabria, University of Cantabria, Cantabria, Spain
| | - Cedric Boeckx
- University of Barcelona, Barcelona, Spain
- University of Barcelona Institute of Complex Systems (UBICS), Barcelona, Spain
- Catalan Institute for Advanced Studies and Research (ICREA), Barcelona, Spain
| | - Giuseppe Testa
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Laboratory of Stem Cell Epigenetics, IEO, European Institute of Oncology, IRCCS, Milan, Italy
- Human Technopole, Center for Neurogenomics, Via Cristina Belgioioso 171, Milan, Italy
| |
Collapse
|
20
|
Ankamreddy H, Min H, Kim JY, Yang X, Cho ES, Kim UK, Bok J. Region-specific endodermal signals direct neural crest cells to form the three middle ear ossicles. Development 2019; 146:dev.167965. [PMID: 30630826 DOI: 10.1242/dev.167965] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 12/24/2018] [Indexed: 11/20/2022]
Abstract
Defects in the middle ear ossicles - malleus, incus and stapes - can lead to conductive hearing loss. During development, neural crest cells (NCCs) migrate from the dorsal hindbrain to specific locations in pharyngeal arch (PA) 1 and 2, to form the malleus-incus and stapes, respectively. It is unclear how migratory NCCs reach their proper destination in the PA and initiate mesenchymal condensation to form specific ossicles. We show that secreted molecules sonic hedgehog (SHH) and bone morphogenetic protein 4 (BMP4) emanating from the pharyngeal endoderm are important in instructing region-specific NCC condensation to form malleus-incus and stapes, respectively, in mouse. Tissue-specific knockout of Shh in the pharyngeal endoderm or Smo (a transducer of SHH signaling) in NCCs causes the loss of malleus-incus condensation in PA1 but only affects the maintenance of stapes condensation in PA2. By contrast, knockout of Bmp4 in the pharyngeal endoderm or Smad4 (a transducer of TGFβ/BMP signaling) in the NCCs disrupts NCC migration into the stapes region in PA2, affecting stapes formation. These results indicate that region-specific endodermal signals direct formation of specific middle ear ossicles.
Collapse
Affiliation(s)
- Harinarayana Ankamreddy
- Department of Anatomy, Yonsei University College of Medicine, Seoul, South Korea.,BK21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Hyehyun Min
- Department of Anatomy, Yonsei University College of Medicine, Seoul, South Korea
| | - Jae Yoon Kim
- Department of Anatomy, Yonsei University College of Medicine, Seoul, South Korea.,BK21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Xiao Yang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206, China
| | - Eui-Sic Cho
- Cluster for Craniofacial Development and Regeneration Research, Institute of Oral Biosciences, Chonbuk National University School of Dentistry, Jeonju, South Korea
| | - Un-Kyung Kim
- Department of Biology, College of Natural Sciences, Kyungpook National University, Daegu, South Korea.,School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, South Korea
| | - Jinwoong Bok
- Department of Anatomy, Yonsei University College of Medicine, Seoul, South Korea .,BK21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea.,Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
21
|
Yun CY, Choi H, You YJ, Yang JY, Baek JA, Cho ES. Requirement of Smad4-mediated signaling in odontoblast differentiation and dentin matrix formation. Anat Cell Biol 2016; 49:199-205. [PMID: 27722013 PMCID: PMC5052229 DOI: 10.5115/acb.2016.49.3.199] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 08/17/2016] [Accepted: 08/29/2016] [Indexed: 01/07/2023] Open
Abstract
Dentin is the major part of tooth and formed by odontoblasts. Under the influence of the inner enamel epithelium, odontoblasts differentiate from ectomesenchymal cells of the dental papilla and secrete pre-dentin which then undergo mineralization into dentin. Transforming growth factor-beta (TGF-β)/bone morphogenetic protein (BMP) signaling is essential for dentinogenesis; however, the precise molecular mechanisms remain unclear. To understand the role of TGF-β/BMP signaling in odontoblast differentiation and dentin formation, we generated mice with conditional ablation of Smad4, a key intracellular mediator of TGF-β/BMP signaling, using Osr2 or OC-Cre mice. Here we found the molars of Osr2CreSmad4 mutant mice exhibited impaired odontoblast differentiation, and normal dentin was replaced by ectopic bone-like structure. In Osr2CreSmad4 mutant mice, cell polarity of odontoblast was lost, and the thickness of crown dentin was decreased in later stage compared to wild type. Moreover, the root dentin was also impaired and showed ectopic bone-like structure similar to Osr2CreSmad4 mutant mice. Taken together, our results suggest that Smad4-dependent TGF-β/BMP signaling plays a critical role in odontoblast differentiation and dentin formation during tooth development.
Collapse
Affiliation(s)
- Chi-Young Yun
- Cluster for Craniofacial Development and Regeneration Research, Institute of Oral Biosciences, Chonbuk National University School of Dentistry, Jeonju, Korea
| | - Hwajung Choi
- Cluster for Craniofacial Development and Regeneration Research, Institute of Oral Biosciences, Chonbuk National University School of Dentistry, Jeonju, Korea
| | - Young-Jae You
- Cluster for Craniofacial Development and Regeneration Research, Institute of Oral Biosciences, Chonbuk National University School of Dentistry, Jeonju, Korea
| | - Jin-Young Yang
- Department of Dental Hygiene, Daejeon Institute of Science and Technology, Daejeon, Korea
| | - Jin-A Baek
- Cluster for Craniofacial Development and Regeneration Research, Institute of Oral Biosciences, Chonbuk National University School of Dentistry, Jeonju, Korea
| | - Eui-Sic Cho
- Cluster for Craniofacial Development and Regeneration Research, Institute of Oral Biosciences, Chonbuk National University School of Dentistry, Jeonju, Korea
| |
Collapse
|
22
|
Transferrin receptor facilitates TGF-β and BMP signaling activation to control craniofacial morphogenesis. Cell Death Dis 2016; 7:e2282. [PMID: 27362800 PMCID: PMC5108332 DOI: 10.1038/cddis.2016.170] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 03/20/2016] [Accepted: 03/30/2016] [Indexed: 02/05/2023]
Abstract
The Pierre Robin Sequence (PRS), consisting of cleft palate, glossoptosis and micrognathia, is a common human birth defect. However, how this abnormality occurs remains largely unknown. Here we report that neural crest cell (NCC)-specific knockout of transferrin receptor (Tfrc), a well known transferrin transporter protein, caused micrognathia, cleft palate, severe respiratory distress and inability to suckle in mice, which highly resemble human PRS. Histological and anatomical analysis revealed that the cleft palate is due to the failure of palatal shelves elevation that resulted from a retarded extension of Meckel's cartilage. Interestingly, Tfrc deletion dramatically suppressed both transforming growth factor-β (TGF-β) and bone morphogenetic protein (BMP) signaling in cranial NCCs-derived mandibular tissues, suggesting that Tfrc may act as a facilitator of these two signaling pathways during craniofacial morphogenesis. Together, our study uncovers an unknown function of Tfrc in craniofacial development and provides novel insight into the etiology of PRS.
Collapse
|
23
|
Gou Y, Zhang T, Xu J. Transcription Factors in Craniofacial Development: From Receptor Signaling to Transcriptional and Epigenetic Regulation. Curr Top Dev Biol 2015; 115:377-410. [PMID: 26589933 DOI: 10.1016/bs.ctdb.2015.07.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Craniofacial morphogenesis is driven by spatial-temporal terrains of gene expression, which give rise to stereotypical pattern formation. Transcription factors are key cellular components that control these gene expressions. They are information hubs that integrate inputs from extracellular factors and environmental cues, direct epigenetic modifications, and define transcriptional status. These activities allow transcription factors to confer specificity and potency to transcription regulation during development.
Collapse
Affiliation(s)
- Yongchao Gou
- State Key Laboratory of Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Center for Craniofacial Molecular Biology, School of Dentistry, University of Southern California, Los Angeles, USA
| | - Tingwei Zhang
- Center for Craniofacial Molecular Biology, School of Dentistry, University of Southern California, Los Angeles, USA; State Key Laboratory of Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jian Xu
- Center for Craniofacial Molecular Biology, School of Dentistry, University of Southern California, Los Angeles, USA.
| |
Collapse
|
24
|
Yu T, Volponi AA, Babb R, An Z, Sharpe PT. Stem Cells in Tooth Development, Growth, Repair, and Regeneration. Curr Top Dev Biol 2015; 115:187-212. [PMID: 26589926 DOI: 10.1016/bs.ctdb.2015.07.010] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Human teeth contain stem cells in all their mesenchymal-derived tissues, which include the pulp, periodontal ligament, and developing roots, in addition to the support tissues such as the alveolar bone. The precise roles of these cells remain poorly understood and most likely involve tissue repair mechanisms but their relative ease of harvesting makes teeth a valuable potential source of mesenchymal stem cells (MSCs) for therapeutic use. These dental MSC populations all appear to have the same developmental origins, being derived from cranial neural crest cells, a population of embryonic stem cells with multipotential properties. In rodents, the incisor teeth grow continuously throughout life, a feature that requires populations of continuously active mesenchymal and epithelial stem cells. The discrete locations of these stem cells in the incisor have rendered them amenable for study and much is being learnt about the general properties of these stem cells for the incisor as a model system. The incisor MSCs appear to be a heterogeneous population consisting of cells from different neural crest-derived tissues. The epithelial stem cells can be traced directly back in development to a Sox10(+) population present at the time of tooth initiation. In this review, we describe the basic biology of dental stem cells, their functions, and potential clinical uses.
Collapse
Affiliation(s)
- Tian Yu
- Craniofacial Development and Stem Cell Biology, Dental Institute, Kings College London, London, United Kingdom
| | - Ana Angelova Volponi
- Craniofacial Development and Stem Cell Biology, Dental Institute, Kings College London, London, United Kingdom
| | - Rebecca Babb
- Craniofacial Development and Stem Cell Biology, Dental Institute, Kings College London, London, United Kingdom
| | - Zhengwen An
- Craniofacial Development and Stem Cell Biology, Dental Institute, Kings College London, London, United Kingdom
| | - Paul T Sharpe
- Craniofacial Development and Stem Cell Biology, Dental Institute, Kings College London, London, United Kingdom.
| |
Collapse
|
25
|
Yang G, Zhou J, Teng Y, Xie J, Lin J, Guo X, Gao Y, He M, Yang X, Wang S. Mesenchymal TGF-β signaling orchestrates dental epithelial stem cell homeostasis through Wnt signaling. Stem Cells 2015; 32:2939-48. [PMID: 24964772 DOI: 10.1002/stem.1772] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Revised: 05/21/2014] [Accepted: 06/21/2014] [Indexed: 01/04/2023]
Abstract
In mouse, continuous growth of the postnatal incisor is coordinated by two populations of multipotent progenitor cells, the dental papilla mesenchymal cells and dental epithelial stem cells, residing at the proximal end of the incisor, yet the molecular mechanism underlying the cooperation between mesenchymal and epithelial cells is largely unknown. Here, transforming growth factor-β (TGF-β) type II receptor (Tgfbr2) was specifically deleted within the postnatal dental papilla mesenchyme. The Tgfbr2-deficient mice displayed malformed incisors with wavy mineralized structures at the labial side as a result of increased differentiation of dental epithelial stem cells. We found that mesenchymal Tgfbr2 disruption led to upregulated expression of Wnt5a and downregulated expression of Fgf3/10 in the mesenchyme, both of which synergistically enhanced Lrp5/6-β-catenin signaling in the cervical loop epithelium. In accord with these findings, mesenchyme-specific depletion of the Wnt transporter gene Wls abolished the aberrant mineralized structures caused by Tgfbr2 deletion. Thus, mesenchymal TGF-β signaling provides a unifying mechanism for the homeostasis of dental epithelial stem cells via a Wnt signaling-mediated mesenchymal-epithelial cell interaction.
Collapse
Affiliation(s)
- Guan Yang
- Molecular Laboratory for Gene Therapy and Tooth Regeneration, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, People's Republic of China; State Key Laboratory of Proteomics, Genetic Laboratory of Development and Diseases, Institute of Biotechnology, Beijing, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Windhausen T, Squifflet S, Renn J, Muller M. BMP Signaling Regulates Bone Morphogenesis in Zebrafish through Promoting Osteoblast Function as Assessed by Their Nitric Oxide Production. Molecules 2015; 20:7586-601. [PMID: 25919279 PMCID: PMC6272212 DOI: 10.3390/molecules20057586] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Revised: 04/16/2015] [Accepted: 04/21/2015] [Indexed: 01/21/2023] Open
Abstract
Bone morphogenetic proteins (BMPs) control many developmental and physiological processes, including skeleton formation and homeostasis. Previous studies in zebrafish revealed the crucial importance of proper BMP signaling before 48 h post-fertilization (hpf) for cartilage formation in the skull. Here, we focus on the involvement of the BMP pathway between 48 and 96 hpf in bone formation after 96 hpf. Using BMP inhibitors and the expression of a dominant-negative BMP receptor, we analyze whether the loss of BMP signaling affects osteoblastogenesis, osteoblast function and bone mineralization. To this end, we used the transgenic zebrafish line Tg(osterix:mCherry), detection of nitric oxide (NO) production, and alizarin red staining, respectively. We observed that inhibition of BMP signaling between 48 and 72 hpf led to a reduction of NO production and bone mineralization. Osteoblast maturation and chondrogenesis, on the other hand, seemed unchanged. Osteoblast function and bone formation were less affected when BMP signaling was inhibited between 72 and 96 hpf. These results suggest that for the onset of bone formation, proper BMP signaling between 48 and 72 hpf is crucial to ensure osteoblast function and ossification. Furthermore, detection of NO in developing zebrafish larvae appears as an early indicator of bone calcification activity.
Collapse
Affiliation(s)
- Thomas Windhausen
- Laboratory for Organogenesis and Regeneration, Université de Liège, GIGA-R B34, Sart Tilman, 4000 Liège, Belgium.
| | - Steeve Squifflet
- Laboratory for Organogenesis and Regeneration, Université de Liège, GIGA-R B34, Sart Tilman, 4000 Liège, Belgium.
| | - Jörg Renn
- Laboratory for Organogenesis and Regeneration, Université de Liège, GIGA-R B34, Sart Tilman, 4000 Liège, Belgium.
| | - Marc Muller
- Laboratory for Organogenesis and Regeneration, Université de Liège, GIGA-R B34, Sart Tilman, 4000 Liège, Belgium.
| |
Collapse
|
27
|
BMP-SHH signaling network controls epithelial stem cell fate via regulation of its niche in the developing tooth. Dev Cell 2015; 33:125-35. [PMID: 25865348 DOI: 10.1016/j.devcel.2015.02.021] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Revised: 12/17/2014] [Accepted: 02/24/2015] [Indexed: 11/22/2022]
Abstract
During embryogenesis, ectodermal stem cells adopt different fates and form diverse ectodermal organs, such as teeth, hair follicles, mammary glands, and salivary glands. Interestingly, these ectodermal organs differ in their tissue homeostasis, which leads to differential abilities for continuous growth postnatally. Mouse molars lose the ability to grow continuously, whereas incisors retain this ability. In this study, we found that a BMP-Smad4-SHH-Gli1 signaling network may provide a niche supporting transient Sox2+ dental epithelial stem cells in mouse molars. This mechanism also plays a role in continuously growing mouse incisors. The differential fate of epithelial stem cells in mouse molars and incisors is controlled by this BMP/SHH signaling network, which partially accounts for the different postnatal growth potential of molars and incisors. Collectively, our study highlights the importance of crosstalk between two signaling pathways, BMP and SHH, in regulating the fate of epithelial stem cells during organogenesis.
Collapse
|
28
|
Smad4 regulates ureteral smooth muscle cell differentiation during mouse embryogenesis. PLoS One 2014; 9:e104503. [PMID: 25127126 PMCID: PMC4134214 DOI: 10.1371/journal.pone.0104503] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Accepted: 07/14/2014] [Indexed: 12/03/2022] Open
Abstract
Proper formation of ureteral smooth muscle cells (SMCs) during embryogenesis is essential for ureter peristalsis that propels urine from the kidney to the bladder in mammals. Currently the molecular factors that regulate differentiation of ureteral mesenchymal cells into SMCs are incompletely understood. A recent study has reported that Smad4 deficiency reduces the number of ureteral SMCs. However, its precise role in the ureteral smooth muscle development remains largely unknown. Here, we used Tbx18:Cre knock-in mouse line to delete Smad4 to examine its requirement in the development of ureteral mesenchyme and SMC differentiation. We found that mice with specific deletion of Smad4 in Tbx18-expressing ureteral mesenchyme exhibited hydroureter and hydronephrosis at embryonic day (E) 16.5, and the mutant mesenchymal cells failed to differentiate into SMCs with increased apoptosis and decreased proliferation. Molecular markers for SMCs including alpha smooth muscle actin (α-SMA) and smooth muscle myosin heavy chain (SM-MHC) were absent in the mutant ureters. Moreover, disruption of Smad4 significantly reduced the expression of genes, including Sox9, Tbx18 and Myocardin associated with SMC differentiation. These findings suggest that Smad4 is essential for initiating the SMC differentiation program during ureter development.
Collapse
|
29
|
Abstract
AIMS Myhre syndrome is a rare disorder characterized by abnormal growth of the skeleton, muscles, and joints. The relationship of this syndrome to craniofacial growth and development is unknown. To the authors' knowledge, this is the first Japanese case ever studied. METHODOLOGY At 10 years and 7 months of age, the patient was referred to the Department of Pediatric Dentistry in the authors' hospital, complaining of a dental problem. RESULTS The craniofacial region exhibited a long lower face, high and narrowed palate with submucous cleft palate, maxillary constriction, prognathism, open bite, and crowding of the dental arch. Some of these morphological disorders could be affected by the functional manifestations of muscular hypertrophy of the cheek region, muscle tenseness, or the low position of the tongue. General disorders of muscular hypertrophy, thickened bones, and limited joint mobility are consistent with craniofacial findings of muscle tension in the cheek region, thickened calvarium, and limitation of temporomandibular joint movement. The submucous cleft palate and crown deformation of the mandibular central incisor may be affected by dysfunctions of SMAD4 signaling. CONCLUSIONS Craniofacial growth and development is affected by the general characteristics of Myhre syndrome, and could be important in its diagnosis.
Collapse
|
30
|
Biggs LC, Mikkola ML. Early inductive events in ectodermal appendage morphogenesis. Semin Cell Dev Biol 2014; 25-26:11-21. [DOI: 10.1016/j.semcdb.2014.01.007] [Citation(s) in RCA: 131] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Revised: 01/23/2014] [Accepted: 01/24/2014] [Indexed: 01/18/2023]
|
31
|
Wang Y, Cox MK, Coricor G, MacDougall M, Serra R. Inactivation of Tgfbr2 in Osterix-Cre expressing dental mesenchyme disrupts molar root formation. Dev Biol 2013; 382:27-37. [PMID: 23933490 DOI: 10.1016/j.ydbio.2013.08.003] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Accepted: 08/01/2013] [Indexed: 11/19/2022]
Abstract
It has been difficult to examine the role of TGF-ß in post-natal tooth development due to perinatal lethality in many of the signaling deficient mouse models. To address the role of Tgfbr2 in postnatal tooth development, we generated a mouse in which Tgfbr2 was deleted in odontoblast- and bone-producing mesenchyme. Osx-Cre;Tgfbr2(fl/fl) mice were generated (Tgfbr2(cko)) and post-natal tooth development was compared in Tgfbr2(cko) and control littermates. X-ray and μCT analysis showed that in Tgfbr2(cko) mice radicular dentin matrix density was reduced in the molars. Molar shape was abnormal and molar eruption was delayed in the mutant mice. Most significantly, defects in root formation, including failure of the root to elongate, were observed by postnatal day 10. Immunostaining for Keratin-14 (K14) was used to delineate Hertwig's epithelial root sheath (HERS). The results showed a delay in elongation and disorganization of the HERS in Tgfbr2(cko) mice. In addition, the HERS was maintained and the break up into epithelial rests was attenuated suggesting that Tgfbr2 acts on dental mesenchyme to indirectly regulate the formation and maintenance of the HERS. Altered odontoblast organization and reduced Dspp expression indicated that odontoblast differentiation was disrupted in the mutant mice likely contributing to the defect in root formation. Nevertheless, expression of Nfic, a key mesenchymal regulator of root development, was similar in Tgfbr2(cko) mice and controls. The number of osteoclasts in the bone surrounding the tooth was reduced and osteoblast differentiation was disrupted likely contributing to both root and eruption defects. We conclude that Tgfbr2 in dental mesenchyme and bone is required for tooth development particularly root formation.
Collapse
Affiliation(s)
- Ying Wang
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Megan K Cox
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - George Coricor
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Mary MacDougall
- Institute of Oral Health Research, School of Dentistry, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Rosa Serra
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
32
|
Hegarty SV, O'Keeffe GW, Sullivan AM. BMP-Smad 1/5/8 signalling in the development of the nervous system. Prog Neurobiol 2013; 109:28-41. [PMID: 23891815 DOI: 10.1016/j.pneurobio.2013.07.002] [Citation(s) in RCA: 126] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2012] [Revised: 07/16/2013] [Accepted: 07/16/2013] [Indexed: 02/07/2023]
Abstract
The transcription factors, Smad1, Smad5 and Smad8, are the pivotal intracellular effectors of the bone morphogenetic protein (BMP) family of proteins. BMPs and their receptors are expressed in the nervous system (NS) throughout its development. This review focuses on the actions of Smad 1/5/8 in the developing NS. The mechanisms by which these Smad proteins regulate the induction of the neuroectoderm, the central nervous system (CNS) primordium, and finally the neural crest, which gives rise to the peripheral nervous system (PNS), are reviewed herein. We describe how, following neural tube closure, the most dorsal aspect of the tube becomes a signalling centre for BMPs, which directs the pattern of the development of the dorsal spinal cord (SC), through the action of Smad1, Smad5 and Smad8. The direct effects of Smad 1/5/8 signalling on the development of neuronal and non-neuronal cells from various neural progenitor cell populations are then described. Finally, this review discusses the neurodevelopmental abnormalities associated with the knockdown of Smad 1/5/8.
Collapse
Affiliation(s)
- Shane V Hegarty
- Department of Anatomy & Neuroscience, University College Cork, Cork, Ireland
| | | | | |
Collapse
|
33
|
CTGF mediates Smad-dependent transforming growth factor β signaling to regulate mesenchymal cell proliferation during palate development. Mol Cell Biol 2013; 33:3482-93. [PMID: 23816882 DOI: 10.1128/mcb.00615-13] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Transforming growth factor β (TGF-β) signaling plays crucial functions in the regulation of craniofacial development, including palatogenesis. Here, we have identified connective tissue growth factor (Ctgf) as a downstream target of the TGF-β signaling pathway in palatogenesis. The pattern of Ctgf expression in wild-type embryos suggests that it may be involved in key processes during palate development. We found that Ctgf expression is downregulated in both Wnt1-Cre; Tgfbr2(fl/fl) and Osr2-Cre; Smad4(fl/fl) palates. In Tgfbr2 mutant embryos, downregulation of Ctgf expression is associated with p38 mitogen-activated protein kinase (MAPK) overactivation, whereas loss of function of Smad4 itself leads to downregulation of Ctgf expression. We also found that CTGF regulates its own expression via TGF-β signaling. Osr2-Cre; Smad4(fl/fl) mice exhibit a defect in cell proliferation similar to that of Tgfbr2 mutant mice, as well as cleft palate. We detected no alteration in bone morphogenetic protein (BMP) downstream targets in Smad4 mutant palates, suggesting that the reduction in cell proliferation is due to defective transduction of TGF-β signaling via decreased Ctgf expression. Significantly, an exogenous source of CTGF was able to rescue the cell proliferation defect in both Tgfbr2 and Smad4 mutant palates. Collectively, our data suggest that CTGF regulates proliferation as a mediator of the canonical pathway of TGF-β signaling during palatogenesis.
Collapse
|
34
|
Falk S, Joosten E, Kaartinen V, Sommer L. Smad4 and Trim33/Tif1γ redundantly regulate neural stem cells in the developing cortex. ACTA ACUST UNITED AC 2013; 24:2951-63. [PMID: 23765158 DOI: 10.1093/cercor/bht149] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
During central nervous system (CNS) development, proliferation and differentiation of neural stem cells (NSCs) have to be regulated in a spatio-temporal fashion. Here, we report different branches of the transforming growth factor β (TGFβ) signaling pathway to be required for the brain area-specific control of NSCs. In the midbrain, canonical TGFβ signaling via Smad4 regulates the balance between proliferation and differentiation of NSCs. Accordingly, Smad4 deletion resulted in horizontal expansion of NSCs due to increased proliferation, decreased differentiation, and decreased cell cycle exit. In the developing cortex, however, ablation of Smad4 alone did not have any effect on proliferation and differentiation of NSCs. In contrast, concomitant mutation of both Smad4 and Trim33 led to an increase in proliferative cells in the ventricular zone due to decreased cell cycle exit, revealing a functional redundancy of Smad4 and Trim33. Furthermore, in Smad4-Trim33 double mutant embryos, cortical NSCs generated an excess of deep layer neurons concurrent with a delayed and reduced production of upper layer neurons and, in addition, failed to undergo the neurogenic to gliogenic switch at the right developmental stage. Thus, our data disclose that in different regions of the developing CNS different aspects of the TGFβ signaling pathway are required to ensure proper development.
Collapse
Affiliation(s)
- Sven Falk
- Division of Cell and Developmental Biology, Institute of Anatomy, University of Zurich, Zurich, Switzerland, Current address: Helmholtz Center Munich, German Research Center for Environmental Health, Institute for Stem Cell Research, D-85764 Neuherberg, Germany
| | - Esméé Joosten
- Division of Cell and Developmental Biology, Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Vesa Kaartinen
- Department of Biologic and Materials Sciences, University of Michigan, Ann Arbor, MI 48109, USA and
| | - Lukas Sommer
- Division of Cell and Developmental Biology, Institute of Anatomy, University of Zurich, Zurich, Switzerland
| |
Collapse
|
35
|
Sheehan-Rooney K, Swartz ME, Lovely CB, Dixon MJ, Eberhart JK. Bmp and Shh signaling mediate the expression of satb2 in the pharyngeal arches. PLoS One 2013; 8:e59533. [PMID: 23555697 PMCID: PMC3605343 DOI: 10.1371/journal.pone.0059533] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Accepted: 02/15/2013] [Indexed: 12/16/2022] Open
Abstract
In human, mutation of the transcription factor SATB2 causes severe defects to the palate and jaw. The expression and sequence of SATB2 is highly conserved across vertebrate species, including zebrafish. We sought to understand the regulation of satb2 using the zebrafish model system. Due to the normal expression domains of satb2, we analyzed satb2 expression in mutants with disrupted Hh signaling or defective ventral patterning. While satb2 expression appears independent of Edn1 signaling, appropriate expression requires Shha, Smo, Smad5 and Hand2 function. Transplantation experiments show that neural crest cells receive both Bmp and Hh signaling to induce satb2 expression. Dorsomorphin- and cyclopamine-mediated inhibition of Bmp and Hh signaling, respectively, suggests that proper satb2 expression requires a relatively earlier Bmp signal and a later Hh signal. We propose that Bmp signaling establishes competence for the neural crest to respond to Hh signaling, thus inducing satb2 expression.
Collapse
Affiliation(s)
- Kelly Sheehan-Rooney
- Section of Molecular, Cell and Developmental Biology, Institute of Cellular and Molecular Biology, University of Texas at Austin, Austin, Texas, United States of America
| | - Mary E. Swartz
- Section of Molecular, Cell and Developmental Biology, Institute of Cellular and Molecular Biology, University of Texas at Austin, Austin, Texas, United States of America
| | - C. Ben Lovely
- Section of Molecular, Cell and Developmental Biology, Institute of Cellular and Molecular Biology, University of Texas at Austin, Austin, Texas, United States of America
| | - Michael J. Dixon
- Faculty of Life Sciences and Faculty of Medical and Human Sciences, Manchester Academic Health Sciences Centre, Michael Smith Building, University of Manchester, Manchester, United Kingdom
| | - Johann K. Eberhart
- Section of Molecular, Cell and Developmental Biology, Institute of Cellular and Molecular Biology, University of Texas at Austin, Austin, Texas, United States of America
- * E-mail:
| |
Collapse
|
36
|
Dalcq J, Pasque V, Ghaye A, Larbuisson A, Motte P, Martial JA, Muller M. RUNX3, EGR1 and SOX9B form a regulatory cascade required to modulate BMP-signaling during cranial cartilage development in zebrafish. PLoS One 2012; 7:e50140. [PMID: 23209659 PMCID: PMC3507947 DOI: 10.1371/journal.pone.0050140] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2012] [Accepted: 10/17/2012] [Indexed: 12/14/2022] Open
Abstract
The cartilaginous elements forming the pharyngeal arches of the zebrafish derive from cranial neural crest cells. Their proper differentiation and patterning are regulated by reciprocal interactions between neural crest cells and surrounding endodermal, ectodermal and mesodermal tissues. In this study, we show that the endodermal factors Runx3 and Sox9b form a regulatory cascade with Egr1 resulting in transcriptional repression of the fsta gene, encoding a BMP antagonist, in pharyngeal endoderm. Using a transgenic line expressing a dominant negative BMP receptor or a specific BMP inhibitor (dorsomorphin), we show that BMP signaling is indeed required around 30 hpf in the neural crest cells to allow cell differentiation and proper pharyngeal cartilage formation. Runx3, Egr1, Sox9b and BMP signaling are required for expression of runx2b, one of the key regulator of cranial cartilage maturation and bone formation. Finally, we show that egr1 depletion leads to increased expression of fsta and inhibition of BMP signaling in the pharyngeal region. In conclusion, we show that the successive induction of the transcription factors Runx3, Egr1 and Sox9b constitutes a regulatory cascade that controls expression of Follistatin A in pharyngeal endoderm, the latter modulating BMP signaling in developing cranial cartilage in zebrafish.
Collapse
Affiliation(s)
- Julia Dalcq
- Laboratory for Molecular Biology and Genetic Engineering, GIGA-R, Université de Liège, Liège, Belgium
| | - Vincent Pasque
- Laboratory for Molecular Biology and Genetic Engineering, GIGA-R, Université de Liège, Liège, Belgium
| | - Aurélie Ghaye
- Laboratory for Molecular Biology and Genetic Engineering, GIGA-R, Université de Liège, Liège, Belgium
| | - Arnaud Larbuisson
- Laboratory for Molecular Biology and Genetic Engineering, GIGA-R, Université de Liège, Liège, Belgium
| | - Patrick Motte
- Plant Functional Genomics and Molecular Imaging and Center for Assistance in Technology of Microscopy, University of Liège, Liège, Belgium
| | - Joseph A. Martial
- Laboratory for Molecular Biology and Genetic Engineering, GIGA-R, Université de Liège, Liège, Belgium
| | - Marc Muller
- Laboratory for Molecular Biology and Genetic Engineering, GIGA-R, Université de Liège, Liège, Belgium
- * E-mail:
| |
Collapse
|
37
|
Jeong J, Cesario J, Zhao Y, Burns L, Westphal H, Rubenstein JLR. Cleft palate defect of Dlx1/2-/- mutant mice is caused by lack of vertical outgrowth in the posterior palate. Dev Dyn 2012; 241:1757-69. [PMID: 22972697 DOI: 10.1002/dvdy.23867] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/28/2012] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Mice lacking the activities of Dlx1 and Dlx2 (Dlx1/2-/-) exhibit cleft palate, one of the most common human congenital defects, but the etiology behind this phenotype has been unknown. Therefore, we analyzed the morphological, cellular, and molecular changes caused by inactivation of Dlx1 and Dlx2 as related to palate development. RESULTS Dlx1/2-/- mutants exhibited lack of vertical growth in the posterior palate during the earliest stage of palatogenesis. We attributed this growth deficiency to reduced cell proliferation. Expression of a cell cycle regulator Ccnd1 was specifically down-regulated in the same region. Previous studies established that the epithelial-mesenchymal signaling loop involving Shh, Bmp4, and Fgf10 is important for cell proliferation and tissue growth during palate development. This signaling loop was disrupted in Dlx1/2-/- palate. Interestingly, however, the decreases in Ccnd1 expression and mitosis in Dlx1/2-/- mutants were independent of this signaling loop. Finally, Dlx1/2 activity was required for normal expression of several transcription factor genes whose mutation results in palate defects. CONCLUSIONS The functions of Dlx1 and Dlx2 are crucial for the initial formation of the posterior palatal shelves, and that the Dlx genes lie upstream of multiple signaling molecules and transcription factors important for later stages of palatogenesis.
Collapse
Affiliation(s)
- Juhee Jeong
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY 10010, USA.
| | | | | | | | | | | |
Collapse
|
38
|
Wang Y, Li L, Zheng Y, Yuan G, Yang G, He F, Chen Y. BMP activity is required for tooth development from the lamina to bud stage. J Dent Res 2012; 91:690-5. [PMID: 22592126 PMCID: PMC3383849 DOI: 10.1177/0022034512448660] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2011] [Revised: 04/16/2012] [Accepted: 04/23/2012] [Indexed: 12/25/2022] Open
Abstract
Several Bmp genes are expressed in the developing mouse tooth germ from the initiation to the late-differentiation stages, and play pivotal roles in multiple steps of tooth development. In this study, we investigated the requirement of BMP activity in early tooth development by transgenic overexpression of the extracellular BMP antagonist Noggin. We show that overexpression of Noggin in the dental epithelium at the tooth initiation stage arrests tooth development at the lamina/early-bud stage. This phenotype is coupled with a significantly reduced level of cell proliferation rate and a down-regulation of Cyclin-D1 expression, specifically in the dental epithelium. Despite unaltered expression of genes known to be implicated in early tooth development in the dental mesenchyme and dental epithelium of transgenic embryos, the expression of Pitx2, a molecular marker for the dental epithelium, became down-regulated, suggesting the loss of odontogenic fate in the transgenic dental epithelium. Our results reveal a novel role for BMP signaling in the progression of tooth development from the lamina stage to the bud stage by regulating cell proliferation and by maintaining odontogenic fate of the dental epithelium.
Collapse
Affiliation(s)
- Y. Wang
- Department of Operative Dentistry and Endodontics, College of Stomatology, The Fourth Military Medical University, Xi’an, P.R. China
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118, USA
| | - L. Li
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118, USA
| | - Y. Zheng
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118, USA
- College of Stomatology, Fujian Medical University, Fuzhou, P.R. China
| | - G. Yuan
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118, USA
| | - G. Yang
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118, USA
| | - F. He
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118, USA
| | - Y. Chen
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118, USA
| |
Collapse
|
39
|
Parada C, Chai Y. Roles of BMP signaling pathway in lip and palate development. FRONTIERS OF ORAL BIOLOGY 2012; 16:60-70. [PMID: 22759670 DOI: 10.1159/000337617] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Cleft lip with or without cleft palate (CLP) and cleft palate only (CP) are severe disruptions affecting orofacial structures. Patients with orofacial clefts require complex interdisciplinary care, which includes nursing, plastic surgery, maxillofacial surgery, otolaryngology, speech therapy, audiology, psychological and genetic counseling, orthodontics and dental treatment, among others. Overall, treatment of clefts of the lip and palate entails a significant economic burden for families and society. Therefore, prevention is the ultimate objective and this will be facilitated by a complete understanding of the etiology of this condition. Here we review the current concepts regarding the genetic and environmental factors contributing to orofacial clefts and emphasize on the roles of BMP signaling pathway components in the normal and aberrant development of the lip and palate.
Collapse
Affiliation(s)
- Carolina Parada
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA, USA
| | | |
Collapse
|
40
|
Stuhlmiller TJ, García-Castro MI. Current perspectives of the signaling pathways directing neural crest induction. Cell Mol Life Sci 2012; 69:3715-37. [PMID: 22547091 PMCID: PMC3478512 DOI: 10.1007/s00018-012-0991-8] [Citation(s) in RCA: 167] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2011] [Revised: 03/12/2012] [Accepted: 04/02/2012] [Indexed: 01/05/2023]
Abstract
The neural crest is a migratory population of embryonic cells with a tremendous potential to differentiate and contribute to nearly every organ system in the adult body. Over the past two decades, an incredible amount of research has given us a reasonable understanding of how these cells are generated. Neural crest induction involves the combinatorial input of multiple signaling pathways and transcription factors, and is thought to occur in two phases from gastrulation to neurulation. In the first phase, FGF and Wnt signaling induce NC progenitors at the border of the neural plate, activating the expression of members of the Msx, Pax, and Zic families, among others. In the second phase, BMP, Wnt, and Notch signaling maintain these progenitors and bring about the expression of definitive NC markers including Snail2, FoxD3, and Sox9/10. In recent years, additional signaling molecules and modulators of these pathways have been uncovered, creating an increasingly complex regulatory network. In this work, we provide a comprehensive review of the major signaling pathways that participate in neural crest induction, with a focus on recent developments and current perspectives. We provide a simplified model of early neural crest development and stress similarities and differences between four major model organisms: Xenopus, chick, zebrafish, and mouse.
Collapse
Affiliation(s)
- Timothy J Stuhlmiller
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, CT 06520-8103, USA
| | | |
Collapse
|
41
|
Han D, Zhao H, Parada C, Hacia JG, Bringas P, Chai Y. A TGFβ-Smad4-Fgf6 signaling cascade controls myogenic differentiation and myoblast fusion during tongue development. Development 2012; 139:1640-50. [PMID: 22438570 DOI: 10.1242/dev.076653] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The tongue is a muscular organ and plays a crucial role in speech, deglutition and taste. Despite the important physiological functions of the tongue, little is known about the regulatory mechanisms of tongue muscle development. TGFβ family members play important roles in regulating myogenesis, but the functional significance of Smad-dependent TGFβ signaling in regulating tongue skeletal muscle development remains unclear. In this study, we have investigated Smad4-mediated TGFβ signaling in the development of occipital somite-derived myogenic progenitors during tongue morphogenesis through tissue-specific inactivation of Smad4 (using Myf5-Cre;Smad4(flox/flox) mice). During the initiation of tongue development, cranial neural crest (CNC) cells occupy the tongue buds before myogenic progenitors migrate into the tongue primordium, suggesting that CNC cells play an instructive role in guiding tongue muscle development. Moreover, ablation of Smad4 results in defects in myogenic terminal differentiation and myoblast fusion. Despite compromised muscle differentiation, tendon formation appears unaffected in the tongue of Myf5-Cre;Smad4(flox/flox) mice, suggesting that the differentiation and maintenance of CNC-derived tendon cells are independent of Smad4-mediated signaling in myogenic cells in the tongue. Furthermore, loss of Smad4 results in a significant reduction in expression of several members of the FGF family, including Fgf6 and Fgfr4. Exogenous Fgf6 partially rescues the tongue myoblast fusion defect of Myf5-Cre;Smad4(flox/flox) mice. Taken together, our study demonstrates that a TGFβ-Smad4-Fgf6 signaling cascade plays a crucial role in myogenic cell fate determination and lineage progression during tongue myogenesis.
Collapse
Affiliation(s)
- Dong Han
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA 90033, USA
| | | | | | | | | | | |
Collapse
|
42
|
Abstract
Cleft palate, a malformation of the secondary palate development, is one of the most common human congenital birth defects. Palate formation is a complex process resulting in the separation of the oral and nasal cavities that involves multiple events, including palatal growth, elevation, and fusion. Recent findings show that transforming growth factor beta (TGF-β) signaling plays crucial roles in regulating palate development in both the palatal epithelium and mesenchyme. Here, we highlight recent advances in our understanding of TGF-β signaling during palate development.
Collapse
Affiliation(s)
- J Iwata
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA 90033, USA
| | | | | |
Collapse
|
43
|
Alexander C, Zuniga E, Blitz IL, Wada N, Le Pabic P, Javidan Y, Zhang T, Cho KW, Crump JG, Schilling TF. Combinatorial roles for BMPs and Endothelin 1 in patterning the dorsal-ventral axis of the craniofacial skeleton. Development 2011; 138:5135-46. [PMID: 22031543 DOI: 10.1242/dev.067801] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Bone morphogenetic proteins (BMPs) play crucial roles in craniofacial development but little is known about their interactions with other signals, such as Endothelin 1 (Edn1) and Jagged/Notch, which pattern the dorsal-ventral (DV) axis of the pharyngeal arches. Here, we use transgenic zebrafish to monitor and perturb BMP signaling during arch formation. With a BMP-responsive transgene, Tg(Bre:GFP), we show active BMP signaling in neural crest (NC)-derived skeletal precursors of the ventral arches, and in surrounding epithelia. Loss-of-function studies using a heat shock-inducible, dominant-negative BMP receptor 1a [Tg(hs70I:dnBmpr1a-GFP)] to bypass early roles show that BMP signaling is required for ventral arch development just after NC migration, the same stages at which we detect Tg(Bre:GFP). Inhibition of BMP signaling at these stages reduces expression of the ventral signal Edn1, as well as ventral-specific genes such as hand2 and dlx6a in the arches, and expands expression of the dorsal signal jag1b. This results in a loss or reduction of ventral and intermediate skeletal elements and a mis-shapen dorsal arch skeleton. Conversely, ectopic BMP causes dorsal expansion of ventral-specific gene expression and corresponding reductions/transformations of dorsal cartilages. Soon after NC migration, BMP is required to induce Edn1 and overexpression of either signal partially rescues ventral skeletal defects in embryos deficient for the other. However, once arch primordia are established the effects of BMPs become restricted to more ventral and anterior (palate) domains, which do not depend on Edn1. This suggests that BMPs act upstream and in parallel to Edn1 to promote ventral fates in the arches during early DV patterning, but later acquire distinct roles that further subdivide the identities of NC cells to pattern the craniofacial skeleton.
Collapse
Affiliation(s)
- Courtney Alexander
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92697, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Urness LD, Bleyl SB, Wright TJ, Moon AM, Mansour SL. Redundant and dosage sensitive requirements for Fgf3 and Fgf10 in cardiovascular development. Dev Biol 2011; 356:383-97. [PMID: 21664901 PMCID: PMC3143275 DOI: 10.1016/j.ydbio.2011.05.671] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2010] [Revised: 05/06/2011] [Accepted: 05/20/2011] [Indexed: 11/29/2022]
Abstract
Heart development requires contributions from, and coordinated signaling interactions between, several cell populations, including splanchnic and pharyngeal mesoderm, postotic neural crest and the proepicardium. Here we report that Fgf3 and Fgf10, which are expressed dynamically in and near these cardiovascular progenitors, have redundant and dosage sensitive requirements in multiple aspects of early murine cardiovascular development. Embryos with Fgf3(-/+);Fgf10(-/-), Fgf3(-/-);Fgf10(-/+) and Fgf3(-/-);Fgf10(-/-) genotypes formed an allelic series of increasing severity with respect to embryonic survival, with double mutants dead by E11.5. Morphologic analysis of embryos with three mutant alleles at E11.5-E13.5 and double mutants at E9.5-E11.0 revealed multiple cardiovascular defects affecting the outflow tract, ventricular septum, atrioventricular cushions, ventricular myocardium, dorsal mesenchymal protrusion, pulmonary arteries, epicardium and fourth pharyngeal arch artery. Assessment of molecular markers in E8.0-E10.5 double mutants revealed abnormalities in each progenitor population, and suggests that Fgf3 and Fgf10 are not required for specification of cardiovascular progenitors, but rather for their normal developmental coordination. These results imply that coding or regulatory mutations in FGF3 or FGF10 could contribute to human congenital heart defects.
Collapse
Affiliation(s)
- Lisa D. Urness
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Steven B. Bleyl
- Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, Utah 84132, USA
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT 84132, USA
| | - Tracy J. Wright
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Anne M. Moon
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT 84132, USA
- Molecular Medicine Program, University of Utah, Salt Lake City, UT, 84112, USA
| | - Suzanne L. Mansour
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT 84132, USA
| |
Collapse
|
45
|
Abstract
The neural crest cell (NCC) lineage is often referred to as the fourth germ layer in embryos, as its wide range of migration and early colonization of multiple tissues and organ systems throughout the developing body is astounding. Many human birth defects are thought to have their origins within the NCC lineage. Exciting recent conditional mouse targeting and transgenic combinatorial suppression approaches have revealed that the Tgf-b superfamily is a key signaling pathway within the cardiac and cranial NCC subpopulations. Given the complexity of Tgf-b superfamily signaling and that multiple ligand and receptor combinations have already been shown to be expressed within the NCC subpopulations, and the difficulty in transgenically targeting entire signaling cascades, we review several up-to-date transgenic approaches that are revealing unexpected consequences.
Collapse
Affiliation(s)
- Simon J Conway
- HB Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA.
| | | |
Collapse
|
46
|
Li J, Huang X, Xu X, Mayo J, Bringas P, Jiang R, Wang S, Chai Y. SMAD4-mediated WNT signaling controls the fate of cranial neural crest cells during tooth morphogenesis. Development 2011; 138:1977-89. [PMID: 21490069 DOI: 10.1242/dev.061341] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
TGFβ/BMP signaling regulates the fate of multipotential cranial neural crest (CNC) cells during tooth and jawbone formation as these cells differentiate into odontoblasts and osteoblasts, respectively. The functional significance of SMAD4, the common mediator of TGFβ/BMP signaling, in regulating the fate of CNC cells remains unclear. In this study, we investigated the mechanism of SMAD4 in regulating the fate of CNC-derived dental mesenchymal cells through tissue-specific inactivation of Smad4. Ablation of Smad4 results in defects in odontoblast differentiation and dentin formation. Moreover, ectopic bone-like structures replaced normal dentin in the teeth of Osr2-IresCre;Smad4(fl/fl) mice. Despite the lack of dentin, enamel formation appeared unaffected in Osr2-IresCre;Smad4(fl/fl) mice, challenging the paradigm that the initiation of enamel development depends on normal dentin formation. At the molecular level, loss of Smad4 results in downregulation of the WNT pathway inhibitors Dkk1 and Sfrp1 and in the upregulation of canonical WNT signaling, including increased β-catenin activity. More importantly, inhibition of the upregulated canonical WNT pathway in Osr2-IresCre;Smad4(fl/fl) dental mesenchyme in vitro partially rescued the CNC cell fate change. Taken together, our study demonstrates that SMAD4 plays a crucial role in regulating the interplay between TGFβ/BMP and WNT signaling to ensure the proper CNC cell fate decision during organogenesis.
Collapse
Affiliation(s)
- Jingyuan Li
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, 2250 Alcazar Street, CSA 103, Los Angeles, CA 90033, USA
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Wise SB, Stock DW. bmp2b and bmp4 are dispensable for zebrafish tooth development. Dev Dyn 2011; 239:2534-46. [PMID: 21038444 DOI: 10.1002/dvdy.22411] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Bone morphogenetic protein (Bmp) signaling has been shown to play important roles in tooth development at virtually all stages from initiation to hard tissue formation. The specific ligands involved in these processes have not been directly tested by loss-of-function experiments, however. We used morpholino antisense oligonucleotides and mutant analysis in the zebrafish to reduce or eliminate the function of bmp2b and bmp4, two ligands known to be expressed in zebrafish teeth and whose mammalian orthologs are thought to play important roles in tooth development. Surprisingly, we found that elimination of function of these two genes singly and in combination did not prevent the formation of mature, attached teeth. The mostly likely explanation for this result is functional redundancy with other Bmp ligands, which may differ between the zebrafish and the mouse.
Collapse
Affiliation(s)
- Sarah B Wise
- Department of Ecology and Evolutionary Biology, University of Colorado, Boulder, Colorado 80309-0449, USA
| | | |
Collapse
|
48
|
Song L, Zhao M, Wu B, Zhou B, Wang Q, Jiao K. Cell autonomous requirement of endocardial Smad4 during atrioventricular cushion development in mouse embryos. Dev Dyn 2011; 240:211-20. [PMID: 21089072 PMCID: PMC3020975 DOI: 10.1002/dvdy.22493] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Atrioventricular (AV) cushions are the precursors of AV septum and valves. In this study, we examined roles of Smad4 during AV cushion development using a conditional gene inactivation approach. We found that endothelial/endocardial inactivation of Smad4 led to the hypocellular AV cushion defect and that both reduced cell proliferation and increased apoptosis contributed to the defect. Expression of multiple genes critical for cushion development was down-regulated in mutant embryos. In collagen gel assays, the number of mesenchymal cells formed is significantly reduced in mutant AV explants compared to that in control explants, suggesting that the reduction of cushion mesenchyme formation in mutants is unlikely secondary to their gross vasculature abnormalities. Using a previously developed immortal endocardial cell line, we showed that Smad4 is required for BMP signaling- stimulated upregulation of Tbx20 and Gata4. Therefore, our data collectively support the cell-autonomous requirement of endocardial Smad4 in regulating AV cushion development.
Collapse
Affiliation(s)
- Langying Song
- Research Division, Department of Genetics, University of Alabama at Birmingham, Birmingham, Alabama
| | - Mei Zhao
- Research Division, Department of Genetics, University of Alabama at Birmingham, Birmingham, Alabama
| | - Bingruo Wu
- Department of Genetics, Albert Einstein College of Medicine, New York, New York
| | - Bin Zhou
- Department of Genetics, Albert Einstein College of Medicine, New York, New York
| | - Qin Wang
- Department of Physiology and Biophysics, the University of Alabama at Birmingham, Birmingham, Alabama
| | - Kai Jiao
- Research Division, Department of Genetics, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
49
|
Tang S, Snider P, Firulli AB, Conway SJ. Trigenic neural crest-restricted Smad7 over-expression results in congenital craniofacial and cardiovascular defects. Dev Biol 2010; 344:233-47. [PMID: 20457144 PMCID: PMC2909335 DOI: 10.1016/j.ydbio.2010.05.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2010] [Revised: 04/30/2010] [Accepted: 05/03/2010] [Indexed: 01/10/2023]
Abstract
Smad7 is a negative regulator of TGFbeta superfamily signaling. Using a three-component triple transgenic system, expression of the inhibitory Smad7 was induced via doxycycline within the NCC lineages at pre- and post-migratory stages. Consistent with its role in negatively regulating both TGFbeta and BMP signaling in vitro, induction of Smad7 within the NCC significantly suppressed phosphorylation levels of both Smad1/5/8 and Smad2/3 in vivo, resulting in subsequent loss of NCC-derived craniofacial, pharyngeal and cardiac OFT cushion cells. At the cellular level, increased cell death was observed in pharyngeal arches. However, cell proliferation and NCC-derived smooth muscle differentiation were unaltered. NCC lineage mapping demonstrated that cardiac NCC emigration and initial migration were not affected, but subsequent colonization of the OFT was significantly reduced. Induction of Smad7 in post-migratory NCC resulted in interventricular septal chamber septation defects, suggesting that TGFbeta superfamily signaling is also essential for cardiac NCC at post-migratory stages to govern normal cardiac development. Taken together, the data illustrate that tightly regulated TGFbeta superfamily signaling plays an essential role during craniofacial and cardiac NCC colonization and cell survival in vivo.
Collapse
Affiliation(s)
- Sunyong Tang
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202 USA
- Department of Biochemistry & Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| | - Paige Snider
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| | - Antony B. Firulli
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| | - Simon J. Conway
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202 USA
- Department of Biochemistry & Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| |
Collapse
|
50
|
Greene RM, Pisano MM. Palate morphogenesis: current understanding and future directions. BIRTH DEFECTS RESEARCH. PART C, EMBRYO TODAY : REVIEWS 2010; 90:133-54. [PMID: 20544696 PMCID: PMC3138490 DOI: 10.1002/bdrc.20180] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
In the past, most scientists conducted their inquiries of nature via inductivism, the patient accumulation of "pieces of information" in the pious hope that the sum of the parts would clarify the whole. Increasingly, modern biology employs the tools of bioinformatics and systems biology in attempts to reveal the "big picture." Most successful laboratories engaged in the pursuit of the secrets of embryonic development, particularly those whose research focus is craniofacial development, pursue a middle road where research efforts embrace, rather than abandon, what some have called the "pedestrian" qualities of inductivism, while increasingly employing modern data mining technologies. The secondary palate has provided an excellent paradigm that has enabled examination of a wide variety of developmental processes. Examination of cellular signal transduction, as it directs embryogenesis, has proven exceptionally revealing with regard to clarification of the "facts" of palatal ontogeny-at least the facts as we currently understand them. Herein, we review the most basic fundamentals of orofacial embryology and discuss how functioning of TGFbeta, BMP, Shh, and Wnt signal transduction pathways contributes to palatal morphogenesis. Our current understanding of palate medial edge epithelial differentiation is also examined. We conclude with a discussion of how the rapidly expanding field of epigenetics, particularly regulation of gene expression by miRNAs and DNA methylation, is critical to control of cell and tissue differentiation, and how examination of these epigenetic processes has already begun to provide a better understanding of, and greater appreciation for, the complexities of palatal morphogenesis.
Collapse
Affiliation(s)
- Robert M Greene
- Department of Molecular, Cellular and Craniofacial Biology, University of Louisville, Birth Defects Center, ULSD, Louisville, Kentucky 40292, USA.
| | | |
Collapse
|