1
|
Ombashi S, Wagemans JCG, van Roey VR, Amiel J, Galliani E, Khonsari RH, Zafra Vallejo V, Krimmel M, Dowgierd K, Wolvius EB. From Fusion to Function: Clinical Insights and Therapeutic Strategies in Syngnathia. J Craniofac Surg 2025:00001665-990000000-02716. [PMID: 40358514 DOI: 10.1097/scs.0000000000011451] [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: 03/12/2025] [Accepted: 03/13/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Syngnathia is a very rare craniofacial anomaly, referring to different forms of fusion of the upper and lower jaw, with or without involvement of the temporomandibular joint (TMJ). Due to its rarity, limited knowledge is available about the clinical manifestation and, in specific, about treatment strategies. The aim of the study is to map out the clinical manifestation and the currently used treatment options by collecting information systematically in a case series. METHODS Within the European Reference Network of Craniofacial Anomalies and ENT disorders, a call was sent out to identify cases of syngnathia. In several meetings, a clinical checklist was developed and approved by all involved centers. Data was collected through the checklist, and a comparison with previous literature was made. RESULTS A total of 12 cases of syngnathia were identified. Cases included bony and fibrous syngnathia, as well as a combination. The majority of the cases were diagnosed directly after birth. Most cases were associated with additional (craniofacial) anomalies and syndromes occurred in 4 of the cases. In 6 cases, the TMJ was involved. Treatment strategies varied and timing of the first surgery differed between 4 days and 9 years. In all cases, feeding problems occurred. The need for a tracheal stoma was reported in 8 cases. CONCLUSION This study presents the largest case series of syngnathia so far, emphasizing the high occurrence of oral health problems in the population. Suggestions for adjustment of a clinical classification system are made. Concerning treatment decisions, caution should be taken with early release operations.
Collapse
Affiliation(s)
- Saranda Ombashi
- Department of Oral and Maxillofacial Surgery, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Jop C G Wagemans
- Department of Oral and Maxillofacial Surgery, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Victor R van Roey
- Department of Plastic and Reconstructive Surgery and Hand Surgery, Erasmus University Medical Center, Rotterdam, The Netherlands
| | | | - Eva Galliani
- Department of Maxillofacial Surgery and Plastic Surgery, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Roman H Khonsari
- Department of Maxillofacial Surgery and Plastic Surgery, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Víctor Zafra Vallejo
- Department of Oral and Maxillofacial Surgery, 12 de Octubre University Hospital, Madrid, Spain
| | - Michael Krimmel
- Department of Oral and Maxillofacial Surgery, University Hospital Tübingen, Tübingen, Germany
| | - Krzysztof Dowgierd
- Department of Clinical Pediatrics, Head and Neck Surgery Clinic for Children and Young Adults, University of Warmia and Mazury, Olsztyn, Poland
| | - Eppo B Wolvius
- Department of Oral and Maxillofacial Surgery, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
2
|
Marchini M, Keller G, Khan N, Shah R, Saliceti Galarza A, Starr KB, Apostopoulos A, Sanger TJ. Sonic hedgehog and fibroblast growth factor 8 regulate the evolution of amniote facial proportions. Commun Biol 2025; 8:84. [PMID: 39827295 PMCID: PMC11742871 DOI: 10.1038/s42003-025-07522-0] [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: 10/01/2024] [Accepted: 01/10/2025] [Indexed: 01/22/2025] Open
Abstract
Amniote skulls are diverse in shape and skeletal composition, which is the basis of much adaptive diversification within this clade. Major differences in skull shape are established early in development, at a critical developmental interval spanning the initial outgrowth and fusion of the facial processes. In birds, this is orchestrated by domains of Shh and Fgf8 expression, known as the frontonasal ectodermal zone (FEZ). It is unclear whether this model of facial development applies to species with diverse facial skeletons, especially species possessing a skull morphology representative of early amniotes. By investigating facial morphogenesis in the lizard, Anolis sagrei, we show that reptilian skull development is driven by the same genes as mammals and birds, but the manner in which those genes regulate facial development is clade-specific. These genes are not expressed in the frontal-nasal prominence, the region of the avian FEZ. Downregulating Shh and Fgf8 signaling disrupts normal facial development, but in pathway-specific ways. Our results demonstrate that early facial morphogenesis in lizards does not conform to the FEZ model. Lizard skull development may be more representative of the ancestral amniote than other model species with highly derived facial skeletons suggesting that the FEZ may be an avian-specific novelty.
Collapse
Affiliation(s)
- Marta Marchini
- Department of Biology, Loyola University Chicago, Chicago, IL, USA
| | - Greta Keller
- Department of Biology, Loyola University Chicago, Chicago, IL, USA
| | - Naaz Khan
- Department of Biology, Loyola University Chicago, Chicago, IL, USA
| | - Rushabh Shah
- Department of Biology, Loyola University Chicago, Chicago, IL, USA
| | | | | | | | - Thomas J Sanger
- Department of Biology, Loyola University Chicago, Chicago, IL, USA.
| |
Collapse
|
3
|
Agostini V, Tessier A, Djaziri N, Khonsari RH, Galliani E, Kurihara Y, Honda M, Kurihara H, Hidaka K, Tuncbilek G, Picard A, Konas E, Amiel J, Gordon CT. Biallelic truncating variants in VGLL2 cause syngnathia in humans. J Med Genet 2023; 60:1084-1091. [PMID: 37666660 DOI: 10.1136/jmg-2022-109059] [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: 11/16/2022] [Accepted: 04/24/2023] [Indexed: 09/06/2023]
Abstract
BACKGROUND Syngnathia is an ultrarare craniofacial malformation characterised by an inability to open the mouth due to congenital fusion of the upper and lower jaws. The genetic causes of isolated bony syngnathia are unknown. METHODS We used whole exome and Sanger sequencing and microsatellite analysis in six patients (from four families) presenting with syngnathia. We used CRISPR/Cas9 genome editing to generate vgll2a and vgll4l germline mutant zebrafish, and performed craniofacial cartilage analysis in homozygous mutants. RESULTS We identified homozygous truncating variants in vestigial-like family member 2 (VGLL2) in all six patients. Two alleles were identified: one in families of Turkish origin and the other in families of Moroccan origin, suggesting a founder effect for each. A shared haplotype was confirmed for the Turkish patients. The VGLL family of genes encode cofactors of TEAD transcriptional regulators. Vgll2 is regionally expressed in the pharyngeal arches of model vertebrate embryos, and morpholino-based knockdown of vgll2a in zebrafish has been reported to cause defects in development of pharyngeal arch cartilages. However, we did not observe craniofacial anomalies in vgll2a or vgll4l homozygous mutant zebrafish nor in fish with double knockout of vgll2a and vgll4l. In Vgll2 -/- mice, which are known to present a skeletal muscle phenotype, we did not identify defects of the craniofacial skeleton. CONCLUSION Our results suggest that although loss of VGLL2 leads to a striking jaw phenotype in humans, other vertebrates may have the capacity to compensate for its absence during craniofacial development.
Collapse
Affiliation(s)
- Valeria Agostini
- Laboratory of embryology and genetics of human malformations, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR 1163, Institut Imagine and Université Paris Cité, Paris, France
| | - Aude Tessier
- Laboratory of embryology and genetics of human malformations, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR 1163, Institut Imagine and Université Paris Cité, Paris, France
| | - Nabila Djaziri
- Laboratory of embryology and genetics of human malformations, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR 1163, Institut Imagine and Université Paris Cité, Paris, France
| | - Roman Hossein Khonsari
- Service de Chirurgie Maxillofaciale et Chirurgie Plastique, Centre de référence Fentes et Malformations Faciales (MAFACE), Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris Cité, Paris, France
| | - Eva Galliani
- Service de Chirurgie Maxillofaciale et Chirurgie Plastique, Centre de référence Fentes et Malformations Faciales (MAFACE), Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris Cité, Paris, France
| | - Yukiko Kurihara
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Masahiko Honda
- Department of Biochemistry, Faculty of Medicine, Kindai University, Osaka-Sayama, Japan
| | - Hiroki Kurihara
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kyoko Hidaka
- Department of Bioscience and Genetics, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan
| | | | - Arnaud Picard
- Service de Chirurgie Maxillofaciale et Chirurgie Plastique, Centre de référence Fentes et Malformations Faciales (MAFACE), Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris Cité, Paris, France
| | | | - Jeanne Amiel
- Laboratory of embryology and genetics of human malformations, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR 1163, Institut Imagine and Université Paris Cité, Paris, France
- Service de Médecine Génomique des Maladies Rares, Hôpital Necker-Enfants Malades, AP-HP, Paris, France
| | - Christopher T Gordon
- Laboratory of embryology and genetics of human malformations, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR 1163, Institut Imagine and Université Paris Cité, Paris, France
| |
Collapse
|
4
|
Rapid induction and long-term self-renewal of neural crest-derived ectodermal chondrogenic cells from hPSCs. NPJ Regen Med 2022; 7:69. [PMID: 36477591 PMCID: PMC9729200 DOI: 10.1038/s41536-022-00265-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 11/29/2022] [Indexed: 12/12/2022] Open
Abstract
Articular cartilage is highly specific and has limited capacity for regeneration if damaged. Human pluripotent stem cells (hPSCs) have the potential to generate any cell type in the body. Here, we report the dual-phase induction of ectodermal chondrogenic cells (ECCs) from hPSCs through the neural crest (NC). ECCs were able to self-renew long-term (over numerous passages) in a cocktail of growth factors and small molecules. The cells stably expressed cranial neural crest-derived mandibular condylar cartilage markers, such as MSX1, FOXC1 and FOXC2. Compared with chondroprogenitors from iPSCs via the paraxial mesoderm, ECCs had single-cell transcriptome profiles similar to condylar chondrocytes. After the removal of the cocktail sustaining self-renewal, the cells stopped proliferating and differentiated into a homogenous chondrocyte population. Remarkably, after transplantation, this cell lineage was able to form cartilage-like structures resembling mandibular condylar cartilage in vivo. This finding provides a framework to generate self-renewing cranial chondrogenic progenitors, which could be useful for developing cell-based therapy for cranial cartilage injury.
Collapse
|
5
|
Fitriasari S, Trainor PA. Gene-environment interactions in the pathogenesis of common craniofacial anomalies. Curr Top Dev Biol 2022; 152:139-168. [PMID: 36707210 DOI: 10.1016/bs.ctdb.2022.10.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Craniofacial anomalies often exhibit phenotype variability and non-mendelian inheritance due to their multifactorial origin, involving both genetic and environmental factors. A combination of epidemiologic studies, genome-wide association, and analysis of animal models have provided insight into the effects of gene-environment interactions on craniofacial and brain development and the pathogenesis of congenital disorders. In this chapter, we briefly summarize the etiology and pathogenesis of common craniofacial anomalies, focusing on orofacial clefts, hemifacial microsomia, and microcephaly. We then discuss how environmental risk factors interact with genes to modulate the incidence and phenotype severity of craniofacial anomalies. Identifying environmental risk factors and dissecting their interaction with different genes and modifiers is central to improved strategies for preventing craniofacial anomalies.
Collapse
Affiliation(s)
| | - Paul A Trainor
- Stowers Institute for Medical Research, Kansas City, MO, United States; Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, United States.
| |
Collapse
|
6
|
Ang PS, Matrongolo MJ, Zietowski ML, Nathan SL, Reid RR, Tischfield MA. Cranium growth, patterning and homeostasis. Development 2022; 149:dev201017. [PMID: 36408946 PMCID: PMC9793421 DOI: 10.1242/dev.201017] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Craniofacial development requires precise spatiotemporal regulation of multiple signaling pathways that crosstalk to coordinate the growth and patterning of the skull with surrounding tissues. Recent insights into these signaling pathways and previously uncharacterized progenitor cell populations have refined our understanding of skull patterning, bone mineralization and tissue homeostasis. Here, we touch upon classical studies and recent advances with an emphasis on developmental and signaling mechanisms that regulate the osteoblast lineage for the calvaria, which forms the roof of the skull. We highlight studies that illustrate the roles of osteoprogenitor cells and cranial suture-derived stem cells for proper calvarial growth and homeostasis. We also discuss genes and signaling pathways that control suture patency and highlight how perturbing the molecular regulation of these pathways leads to craniosynostosis. Finally, we discuss the recently discovered tissue and signaling interactions that integrate skull and cerebrovascular development, and the potential implications for both cerebrospinal fluid hydrodynamics and brain waste clearance in craniosynostosis.
Collapse
Affiliation(s)
- Phillip S. Ang
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
- University of Chicago Pritzker School of Medicine, Chicago, IL 60637, USA
| | - Matt J. Matrongolo
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
- Child Health Institute of New Jersey, New Brunswick, NJ 08901, USA
| | | | - Shelby L. Nathan
- Laboratory of Craniofacial Biology and Development, Section of Plastic Surgery, Department of Surgery, University of Chicago Medicine, Chicago, IL 60637, USA
| | - Russell R. Reid
- Laboratory of Craniofacial Biology and Development, Section of Plastic Surgery, Department of Surgery, University of Chicago Medicine, Chicago, IL 60637, USA
| | - Max A. Tischfield
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
- Child Health Institute of New Jersey, New Brunswick, NJ 08901, USA
| |
Collapse
|
7
|
Zbasnik N, Dolan K, Buczkowski SA, Green RM, Hallgrímsson B, Marcucio RS, Moon AM, Fish JL. Fgf8 dosage regulates jaw shape and symmetry through pharyngeal-cardiac tissue relationships. Dev Dyn 2022; 251:1711-1727. [PMID: 35618654 PMCID: PMC9529861 DOI: 10.1002/dvdy.501] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 05/17/2022] [Accepted: 05/17/2022] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Asymmetries in craniofacial anomalies are commonly observed. In the facial skeleton, the left side is more commonly and/or severely affected than the right. Such asymmetries complicate treatment options. Mechanisms underlying variation in disease severity between individuals as well as within individuals (asymmetries) are still relatively unknown. RESULTS Developmental reductions in fibroblast growth factor 8 (Fgf8) have a dosage dependent effect on jaw size, shape, and symmetry. Further, Fgf8 mutants have directionally asymmetric jaws with the left side being more affected than the right. Defects in lower jaw development begin with disruption to Meckel's cartilage, which is discontinuous. All skeletal elements associated with the proximal condensation are dysmorphic, exemplified by a malformed and misoriented malleus. At later stages, Fgf8 mutants exhibit syngnathia, which falls into two broad categories: bony fusion of the maxillary and mandibular alveolar ridges and zygomatico-mandibular fusion. All of these morphological defects exhibit both inter- and intra-specimen variation. CONCLUSIONS We hypothesize that these asymmetries are linked to heart development resulting in higher levels of Fgf8 on the right side of the face, which may buffer the right side to developmental perturbations. This mouse model may facilitate future investigations of mechanisms underlying human syngnathia and facial asymmetry.
Collapse
Affiliation(s)
- Nathaniel Zbasnik
- Department of Biological SciencesUniversity of Massachusetts LowellLowellMassachusettsUSA
| | - Katie Dolan
- Department of Biological SciencesUniversity of Massachusetts LowellLowellMassachusettsUSA
| | - Stephanie A. Buczkowski
- Department of Molecular and Functional GenomicsGeisinger Medical CenterDanvillePennsylvaniaUSA
| | - Rebecca M. Green
- Center for Craniofacial and Dental Genetics and Department of Oral and Craniofacial SciencesUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Benedikt Hallgrímsson
- Department of Cell Biology and AnatomyAlberta Chidren's Hospital Research Institute, University of CalgaryCalgaryAlbertaCanada
| | - Ralph S. Marcucio
- Orthopaedic Surgery, Orthopaedic Trauma InstituteUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Anne M. Moon
- Department of Molecular and Functional GenomicsGeisinger Medical CenterDanvillePennsylvaniaUSA,Departments of Pediatrics and Human GeneticsUniversity of UtahSalt Lake CityUtahUSA
| | - Jennifer L. Fish
- Department of Biological SciencesUniversity of Massachusetts LowellLowellMassachusettsUSA
| |
Collapse
|
8
|
Van Otterloo E, Milanda I, Pike H, Thompson JA, Li H, Jones KL, Williams T. AP-2α and AP-2β cooperatively function in the craniofacial surface ectoderm to regulate chromatin and gene expression dynamics during facial development. eLife 2022; 11:e70511. [PMID: 35333176 PMCID: PMC9038197 DOI: 10.7554/elife.70511] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 03/22/2022] [Indexed: 11/13/2022] Open
Abstract
The facial surface ectoderm is essential for normal development of the underlying cranial neural crest cell populations, providing signals that direct appropriate growth, patterning, and morphogenesis. Despite the importance of the ectoderm as a signaling center, the molecular cues and genetic programs implemented within this tissue are understudied. Here, we show that removal of two members of the AP-2 transcription factor family, AP-2α and AP-2ß, within the early embryonic ectoderm of the mouse leads to major alterations in the craniofacial complex. Significantly, there are clefts in both the upper face and mandible, accompanied by fusion of the upper and lower jaws in the hinge region. Comparison of ATAC-seq and RNA-seq analyses between controls and mutants revealed significant changes in chromatin accessibility and gene expression centered on multiple AP-2 binding motifs associated with enhancer elements within these ectodermal lineages. In particular, loss of these AP-2 proteins affects both skin differentiation as well as multiple signaling pathways, most notably the WNT pathway. We also determined that the mutant clefting phenotypes that correlated with reduced WNT signaling could be rescued by Wnt1 ligand overexpression in the ectoderm. Collectively, these findings highlight a conserved ancestral function for AP-2 transcription factors in ectodermal development and signaling, and provide a framework from which to understand the gene regulatory network operating within this tissue that directs vertebrate craniofacial development.
Collapse
Affiliation(s)
- Eric Van Otterloo
- Iowa Institute for Oral Health Research, College of Dentistry & Dental Clinics, University of IowaIowa CityUnited States
- Department of Periodontics, College of Dentistry & Dental Clinics, University of IowaIowa CityUnited States
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of IowaIowa CityUnited States
- Department of Craniofacial Biology, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Isaac Milanda
- Department of Craniofacial Biology, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Hamish Pike
- Department of Craniofacial Biology, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Jamie A Thompson
- Iowa Institute for Oral Health Research, College of Dentistry & Dental Clinics, University of IowaIowa CityUnited States
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of IowaIowa CityUnited States
| | - Hong Li
- Department of Craniofacial Biology, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Kenneth L Jones
- Department of Pediatrics, Section of Hematology, Oncology, and Bone Marrow Transplant, University of Colorado School of Medicine, University of Colorado Anschutz Medical CampusAuroraUnited States
| | - Trevor Williams
- Department of Craniofacial Biology, University of Colorado Anschutz Medical CampusAuroraUnited States
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical CampusAuroraUnited States
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Children's Hospital ColoradoAuroraUnited States
| |
Collapse
|
9
|
Wang Y, Lu L, Niu Y, Zhang Q, Cheng C, Huang H, Huang X, Huang Q. The osteoporosis risk variant rs9820407 at 3p22.1 acts as an allele-specific enhancer to regulate CTNNB1 expression by long-range chromatin loop formation. Bone 2021; 153:116165. [PMID: 34461284 DOI: 10.1016/j.bone.2021.116165] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 08/25/2021] [Accepted: 08/25/2021] [Indexed: 11/26/2022]
Abstract
Previous powerful genome-wide association studies (GWASs) and whole-genome sequencing have identified multiple single-nucleotide polymorphisms (SNPs) located over 69 kb upstream of CTNNB1 at 3p22.1 locus associated with osteoporosis. The CTNNB1 gene encodes β-catenin that is an integral part of adherens junctions and the primary mediator of the canonical Wnt signaling pathway. The causal variants and underlying molecular mechanisms of the osteoporosis susceptibility locus 3p22.1 remains unknown. Through comprehensive computational analyses, including expression quantitative trait locus (eQTL), high-throughput chromatin interaction (Hi-C), epigenomic and functional annotation, four enhancer SNPs (rs9820407, rs9878224, rs454690 and rs9832204) were prioritized as potential causal SNPs at 3p22.1 for osteoporosis. Rs9820407 displayed the strongest enhancer activity in dual-luciferase assays. Specifically, the minor rs9820407-A can preferentially bind transcription factor FOXC1, elevate the enhancer activity and increase CTNNB1 expression. The architectural protein CTCF was presumably involved in long-range chromatin interaction between rs9820407 and CTNNB1. Our study provided a mechanistic insight into how noncoding enhancer SNP rs9820407 distally regulates CTNNB1 expression and modulates osteoporosis risk.
Collapse
Affiliation(s)
- Ya Wang
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, China
| | - Li Lu
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, China
| | - Yajing Niu
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, China
| | - Qiongdan Zhang
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, China
| | - Chen Cheng
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, China
| | - Han Huang
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, China
| | - Xinyao Huang
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, China
| | - Qingyang Huang
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan 430079, China.
| |
Collapse
|
10
|
Abe M, Cox TC, Firulli AB, Kanai SM, Dahlka J, Lim KC, Engel JD, Clouthier DE. GATA3 is essential for separating patterning domains during facial morphogenesis. Development 2021; 148:dev199534. [PMID: 34383890 PMCID: PMC8451945 DOI: 10.1242/dev.199534] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 08/02/2021] [Indexed: 11/20/2022]
Abstract
Neural crest cells (NCCs) within the mandibular and maxillary prominences of the first pharyngeal arch are initially competent to respond to signals from either region. However, mechanisms that are only partially understood establish developmental tissue boundaries to ensure spatially correct patterning. In the 'hinge and caps' model of facial development, signals from both ventral prominences (the caps) pattern the adjacent tissues whereas the intervening region, referred to as the maxillomandibular junction (the hinge), maintains separation of the mandibular and maxillary domains. One cap signal is GATA3, a member of the GATA family of zinc-finger transcription factors with a distinct expression pattern in the ventral-most part of the mandibular and maxillary portions of the first arch. Here, we show that disruption of Gata3 in mouse embryos leads to craniofacial microsomia and syngnathia (bony fusion of the upper and lower jaws) that results from changes in BMP4 and FGF8 gene regulatory networks within NCCs near the maxillomandibular junction. GATA3 is thus a crucial component in establishing the network of factors that functionally separate the upper and lower jaws during development.
Collapse
Affiliation(s)
- Makoto Abe
- Department of Craniofacial Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Oral Anatomy and Developmental Biology, Osaka University Graduate School of Dentistry, Suita, Osaka, 565-0871, Japan
| | - Timothy C. Cox
- Departments of Oral & Craniofacial Sciences and Pediatrics, University of Missouri-Kansas City, Kansas City, MO 64108, USA
| | - Anthony B. Firulli
- Herman B Wells Center for Pediatric Research, Departments of Pediatrics, Anatomy and Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Stanley M. Kanai
- Department of Craniofacial Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Jacob Dahlka
- Department of Craniofacial Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Kim-Chew Lim
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - James Douglas Engel
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - David E. Clouthier
- Department of Craniofacial Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
11
|
Takenoshita M, Takechi M, Vu Hoang T, Furutera T, Akagawa C, Namangkalakul W, Aoto K, Kume T, Miyashin M, Iwamoto T, Iseki S. Cell lineage- and expression-based inference of the roles of forkhead box transcription factor Foxc2 in craniofacial development. Dev Dyn 2021; 250:1125-1139. [PMID: 33667029 DOI: 10.1002/dvdy.324] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 02/08/2021] [Accepted: 02/24/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Foxc2 is a member of the winged helix/forkhead (Fox) box family of transcription factors. Loss of function of Foxc2 causes craniofacial abnormalities such as cleft palate and deformed cranial base, but its role during craniofacial development remains to be elucidated. RESULTS The contributions of Foxc2-positive and its descendant cells to the craniofacial structure at E18.5 were examined using a tamoxifen-inducible Cre driver mouse (Foxc2-CreERT2) crossed with the R26R-LacZ reporter mouse. Foxc2 expression at E8.5 is restricted to the cranial mesenchyme, contributing to specific components including the cranial base, sensory capsule, tongue, upper incisor, and middle ear. Expression at E10.5 was still positively regulated in most of those regions. In situ hybridization analysis of Foxc2 and its closely related gene, Foxc1, revealed that expression domains of these genes largely overlap in the cephalic mesenchyme. Meanwhile, the tongue expressed Foxc2 but not Foxc1, and its development was affected by the neural crest-specific deletion of Foxc2 in mice (Wnt1-Cre; Foxc2fl/fl ). CONCLUSIONS Foxc2 is expressed in cranial mesenchyme that contributes to specific craniofacial tissue components from an early stage, and it seems to be involved in their development in cooperation with Foxc1. Foxc2 also has its own role in tongue development.
Collapse
Affiliation(s)
- Manami Takenoshita
- Department of Molecular Craniofacial Embryology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan.,Department of Pediatric Dentistry and Special Needs Dentistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Masaki Takechi
- Department of Molecular Craniofacial Embryology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Tri Vu Hoang
- Department of Molecular Craniofacial Embryology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Toshiko Furutera
- Department of Molecular Craniofacial Embryology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Chisaki Akagawa
- Department of Molecular Craniofacial Embryology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Worachat Namangkalakul
- Department of Molecular Craniofacial Embryology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Kazushi Aoto
- Department of Biochemistry, Hamamatsu University School of Medicine, Tokyo, Japan
| | - Tsutomu Kume
- Feinberg Cardiovascular Research Institute, Development of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Michiyo Miyashin
- Department of Pediatric Dentistry and Special Needs Dentistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Tsutomu Iwamoto
- Department of Pediatric Dentistry and Special Needs Dentistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Sachiko Iseki
- Department of Molecular Craniofacial Embryology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| |
Collapse
|
12
|
Xu P, Yu HV, Tseng KC, Flath M, Fabian P, Segil N, Crump JG. Foxc1 establishes enhancer accessibility for craniofacial cartilage differentiation. eLife 2021; 10:63595. [PMID: 33501917 PMCID: PMC7891931 DOI: 10.7554/elife.63595] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 01/26/2021] [Indexed: 12/15/2022] Open
Abstract
The specification of cartilage requires Sox9, a transcription factor with broad roles for organogenesis outside the skeletal system. How Sox9 and other factors gain access to cartilage-specific cis-regulatory regions during skeletal development was unknown. By analyzing chromatin accessibility during the differentiation of neural crest cells into chondrocytes of the zebrafish head, we find that cartilage-associated chromatin accessibility is dynamically established. Cartilage-associated regions that become accessible after neural crest migration are co-enriched for Sox9 and Fox transcription factor binding motifs. In zebrafish lacking Foxc1 paralogs, we find a global decrease in chromatin accessibility in chondrocytes, consistent with a later loss of dorsal facial cartilages. Zebrafish transgenesis assays confirm that many of these Foxc1-dependent elements function as enhancers with region- and stage-specific activity in facial cartilages. These results show that Foxc1 promotes chondrogenesis in the face by establishing chromatin accessibility at a number of cartilage-associated gene enhancers.
Collapse
Affiliation(s)
- Pengfei Xu
- Eli and Edythe Broad Center for Regenerative Medicine, Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Haoze V Yu
- Eli and Edythe Broad Center for Regenerative Medicine, Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Kuo-Chang Tseng
- Eli and Edythe Broad Center for Regenerative Medicine, Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Mackenzie Flath
- Eli and Edythe Broad Center for Regenerative Medicine, Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Peter Fabian
- Eli and Edythe Broad Center for Regenerative Medicine, Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - Neil Segil
- Eli and Edythe Broad Center for Regenerative Medicine, Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| | - J Gage Crump
- Eli and Edythe Broad Center for Regenerative Medicine, Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| |
Collapse
|
13
|
Drtikolová Kaupová S, Schamall D, Cvrček J, Půtová L, Velemínský P, Teschler-Nicola M. The dietary behavior of two early medieval individuals with temporomandibular ankylosis. INTERNATIONAL JOURNAL OF PALEOPATHOLOGY 2020; 31:1-6. [PMID: 32805633 DOI: 10.1016/j.ijpp.2020.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 07/27/2020] [Accepted: 07/30/2020] [Indexed: 06/11/2023]
Abstract
OBJECTIVES This study aimed to reconstruct the dietary behavior of two early medieval individuals who display gnathic malformation. MATERIAL Two skeletons affected by temporomandibular ankylosis were analyzed, one from the Great Moravian burial site of Rajhradice (9th century AD, Czech Republic), and the other from the Avar burial site of Schӧnkirchen (8th century AD, Austria). METHODS Carbon and nitrogen isotopic values were measured from the bone collagen of both individuals. In the Rajhradice case, where the childhood origin of ankylosis is deduced, isotopic analysis of dentine sections was performed. RESULTS Both individuals show isotopic values within the range of variation of a contemporaneous population sample. There was no observable dietary change in the Rajhradice individual that could be linked to the occurrence of ankylosis. CONCLUSIONS Both individuals consumed diets typical for their populations. They appear to not have restricted access to foodstuffs, namely animal protein, which would likely have had to be served in liquid (e.g. milk) or in a highly mashed form to compensate for insufficient mastication. SIGNIFICANCE This finding provides specific evidence of care provided to these two afflicted members of past populations. LIMITATIONS Though the proportion of animal protein is an important indicator of the quality of diet, many other aspects of diet - such as micronutrient content - elude stable isotope analysis. SUGGESTIONS FOR FURTHER RESEARCH Amino acid compound specific isotope analyses of collagen would provide deeper insight into both the diet and physiology of the affected individuals.
Collapse
Affiliation(s)
- Sylva Drtikolová Kaupová
- Department of Anthropology, National Museum, Václavské náměstí 68, 11579 Praha 1, Czech Republic.
| | - Doris Schamall
- Department of Evolutionary Anthropology, University of Vienna, Althanstraße 14, 1090 Vienna, Austria; Department of Anthropology, Natural History Museum Vienna, Burgring 7, A 1010 Vienna, Austria.
| | - Jan Cvrček
- Department of Anthropology, National Museum, Václavské náměstí 68, 11579 Praha 1, Czech Republic; Department of Anthropology and Human Genetics, Faculty of Science, Charles University, Viničná 7, 128 43, Praha 2, Czech Republic.
| | - Lenka Půtová
- Department of Anthropology, National Museum, Václavské náměstí 68, 11579 Praha 1, Czech Republic; Institute for History of Medicine and Foreign Languages, First Faculty of Medicine, Charles University, U Nemocnice 4, 121 08, Prague, Czech Republic.
| | - Petr Velemínský
- Department of Anthropology, National Museum, Václavské náměstí 68, 11579 Praha 1, Czech Republic.
| | - Maria Teschler-Nicola
- Department of Evolutionary Anthropology, University of Vienna, Althanstraße 14, 1090 Vienna, Austria; Department of Anthropology, Natural History Museum Vienna, Burgring 7, A 1010 Vienna, Austria.
| |
Collapse
|
14
|
Dash S, Bhatt S, Sandell LL, Seidel CW, Ahn Y, Krumlauf RE, Trainor PA. The Mediator Subunit, Med23 Is Required for Embryonic Survival and Regulation of Canonical WNT Signaling During Cranial Ganglia Development. Front Physiol 2020; 11:531933. [PMID: 33192541 PMCID: PMC7642510 DOI: 10.3389/fphys.2020.531933] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Accepted: 09/16/2020] [Indexed: 11/13/2022] Open
Abstract
Development of the vertebrate head is a complex and dynamic process, which requires integration of all three germ layers and their derivatives. Of special importance are ectoderm-derived cells that form the cranial placodes, which then differentiate into the cranial ganglia and sensory organs. Critical to a fully functioning head, defects in cranial placode and sensory organ development can result in congenital craniofacial anomalies. In a forward genetic screen aimed at identifying novel regulators of craniofacial development, we discovered an embryonically lethal mouse mutant, snouty, which exhibits malformation of the facial prominences, cranial nerves and vasculature. The snouty mutation was mapped to a single nucleotide change in a ubiquitously expressed gene, Med23, which encodes a subunit of the global transcription co-factor complex, Mediator. Phenotypic analyses revealed that the craniofacial anomalies, particularly of the cranial ganglia, were caused by a failure in the proper specification of cranial placode neuronal precursors. Molecular analyses determined that defects in cranial placode neuronal differentiation in Med23 sn/sn mutants were associated with elevated WNT/β-catenin signaling, which can be partially rescued through combined Lrp6 and Wise loss-of-function. Our work therefore reveals a surprisingly tissue specific role for the ubiquitously expressed mediator complex protein Med23 in placode differentiation during cranial ganglia development. This highlights the importance of coupling general transcription to the regulation of WNT signaling during embryogenesis.
Collapse
Affiliation(s)
- Soma Dash
- Stowers Institute for Medical Research, Kansas City, MO, United States
| | - Shachi Bhatt
- Stowers Institute for Medical Research, Kansas City, MO, United States.,Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Lisa L Sandell
- Department of Oral Immunology and Infectious Diseases, School of Dentistry, University of Louisville, Louisville, KY, United States
| | | | - Youngwook Ahn
- Stowers Institute for Medical Research, Kansas City, MO, United States
| | - Robb E Krumlauf
- Stowers Institute for Medical Research, Kansas City, MO, United States.,Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Paul A Trainor
- Stowers Institute for Medical Research, Kansas City, MO, United States.,Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, United States
| |
Collapse
|
15
|
Moazzeni H, Khani M, Elahi E. Insights into the regulatory molecules involved in glaucoma pathogenesis. AMERICAN JOURNAL OF MEDICAL GENETICS PART C-SEMINARS IN MEDICAL GENETICS 2020; 184:782-827. [PMID: 32935930 DOI: 10.1002/ajmg.c.31833] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/26/2020] [Accepted: 07/28/2020] [Indexed: 12/19/2022]
Abstract
Glaucoma is an important cause of irreversible blindness, characterized by optic nerve anomalies. Increased intraocular pressure (IOP) and aging are major risk factors. Retinal ganglion cells and trabecular meshwork cells are certainly involved in the etiology of glaucoma. Glaucoma is usually a complex disease, and various genes and functions may contribute to its etiology. Among these may be genes that encode regulatory molecules. In this review, regulatory molecules including 18 transcription factors (TFs), 195 microRNAs (miRNAs), 106 long noncoding RNAs (lncRNAs), and two circular RNAs (circRNAs) that are reasonable candidates for having roles in glaucoma pathogenesis are described. The targets of the regulators are reported. Glaucoma-related features including apoptosis, stress responses, immune functions, ECM properties, IOP, and eye development are affected by the targeted genes. The targeted genes that are frequently targeted by multiple regulators most often affect apoptosis and the related features of cell death and cell survival. BCL2, CDKN1A, and TP53 are among the frequent targets of three types of glaucoma-relevant regulators, TFs, miRNAs, and lncRNAs. TP53 was itself identified as a glaucoma-relevant TF. Several of the glaucoma-relevant TFs are themselves among frequent targets of regulatory molecules, which is consistent with existence of a complex network involved in glaucoma pathogenesis.
Collapse
Affiliation(s)
- Hamidreza Moazzeni
- School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Marzieh Khani
- School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Elahe Elahi
- School of Biology, College of Science, University of Tehran, Tehran, Iran
| |
Collapse
|
16
|
Caddy JC, Luoma LM, Berry FB. FOXC1 negatively regulates BMP‐SMAD activity and Id1 expression during osteoblast differentiation. J Cell Biochem 2020; 121:3266-3277. [DOI: 10.1002/jcb.29595] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 12/09/2019] [Indexed: 12/15/2022]
Affiliation(s)
- Jordan C. Caddy
- Department of Medical GeneticsUniversity of Alberta Edmonton Alberta Canada
| | - Leiah M. Luoma
- Department of Medical GeneticsUniversity of Alberta Edmonton Alberta Canada
- Department of SurgeryUniversity of Alberta Edmonton Alberta Canada
| | - Fred B. Berry
- Department of Medical GeneticsUniversity of Alberta Edmonton Alberta Canada
- Department of SurgeryUniversity of Alberta Edmonton Alberta Canada
| |
Collapse
|
17
|
Ouyang N, Li H, Wang M, Shen H, Si J, Shen G. The Transcription Factor Foxc1 Promotes Osteogenesis by Directly Regulating Runx2 in Response of Intermittent Parathyroid Hormone (1-34) Treatment. Front Pharmacol 2020; 11:592. [PMID: 32431614 PMCID: PMC7216818 DOI: 10.3389/fphar.2020.00592] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Accepted: 04/17/2020] [Indexed: 01/23/2023] Open
Abstract
Parathyroid hormone (PTH) is crucial for bone remodeling. Intermittent PTH (1–34) administration stimulates osteogenesis and promotes bone formation; however, the possible targets and underlying mechanisms still remain unclear. In this study, functional links between PTH and Foxc1, a transcription factor reported to be predominant in skeletal development and formation, were indicated. We determined the impacts of Foxc1 on in vitro osteogenic differentiation and in vivo bone regeneration under intermittent PTH induction, and further explored its possible targets. We found that the expression level of Foxc1 was upregulated during osteogenic induction by intermittent PTH treatment, and the elevated expression of Foxc1 induced by PTH was inhibited by PTH1R silencing, while rescued by intermittent PTH supplement. By gain- and loss-of-function strategies targeting Foxc1 in MC3T3-E1 cells, we demonstrated that Foxc1 could promote in vitro osteogenic differentiation by intermittent PTH induction. Moreover, immunofluorescence analysis indicated the nuclear co-localization of Foxc1 with Runx2. Luciferase-reporter and chromatin immunoprecipitation analysis further confirmed that Foxc1 could bind to the P1 promoter region of Runx2 directly, which plays an indispensable part in osteogenic differentiation and bone mineralization. Meanwhile, we also revealed that Foxc1 could promote bone regeneration induced by intermittent PTH treatment in vivo. Taken together, this study revealed the role and mechanism of Foxc1 on in vitro osteogenic differentiation and in vivo bone regeneration in response of intermittent PTH treatment.
Collapse
Affiliation(s)
- Ningjuan Ouyang
- Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Hongliang Li
- Department of Oral and Cranio-maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Minjiao Wang
- Department of Oral and Cranio-maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Hongzhou Shen
- Department of Oral and Cranio-maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Jiawen Si
- Department of Oral and Cranio-maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai, China
| | - Guofang Shen
- Department of Oral and Cranio-maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai, China
| |
Collapse
|
18
|
Childhood glaucoma genes and phenotypes: Focus on FOXC1 mutations causing anterior segment dysgenesis and hearing loss. Exp Eye Res 2019; 190:107893. [PMID: 31836490 DOI: 10.1016/j.exer.2019.107893] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 10/16/2019] [Accepted: 12/04/2019] [Indexed: 12/27/2022]
Abstract
Childhood glaucoma is an important cause of blindness world-wide. Eleven genes are currently known to cause inherited forms of glaucoma with onset before age 20. While all the early-onset glaucoma genes cause severe disease, considerable phenotypic variability is observed among mutations carriers. In particular, FOXC1 genetic variants are associated with a broad range of phenotypes including multiple forms of glaucoma and also systemic abnormalities, especially hearing loss. FOXC1 is a member of the forkhead family of transcription factors and is involved in neural crest development necessary for formation of anterior eye structures and also pharyngeal arches that form the middle ear bones. In this study we review the clinical phenotypes reported for known FOXC1 mutations and show that mutations in patients with reported ocular anterior segment abnormalities and hearing loss primarily disrupt the critically important forkhead domain. These results suggest that optimal care for patients affected with anterior segment dysgenesis should include screening for FOXC1 mutations and also testing for hearing loss.
Collapse
|
19
|
Moazzeni H, Mirrahimi M, Moghadam A, Banaei-Esfahani A, Yazdani S, Elahi E. Identification of genes involved in glaucoma pathogenesis using combined network analysis and empirical studies. Hum Mol Genet 2019; 28:3637-3663. [PMID: 31518395 DOI: 10.1093/hmg/ddz222] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 08/30/2019] [Accepted: 09/04/2019] [Indexed: 12/25/2022] Open
Abstract
Glaucoma is a leading cause of blindness. We aimed in this study to identify genes that may make subtle and cumulative contributions to glaucoma pathogenesis. To this end, we identified molecular interactions and pathways that include transcription factors (TFs) FOXC1, PITX2, PAX6 and NFKB1 and various microRNAs including miR-204 known to have relevance to trabecular meshwork (TM) functions and/or glaucoma. TM tissue is involved in glaucoma pathogenesis. In-house microarray transcriptome results and data sources were used to identify target genes of the regulatory molecules. Bioinformatics analyses were done to filter TM and glaucoma relevant genes. These were submitted to network-creating softwares to define interactions, pathways and a network that would include the genes. The network was stringently scrutinized and minimized, then expanded by addition of microarray data and data on TF and microRNA-binding sites. Selected features of the network were confirmed by empirical studies such as dual luciferase assays, real-time PCR and western blot experiments and apoptosis assays. MYOC, WDR36, LTPBP2, RHOA, CYP1B1, OPA1, SPARC, MEIS2, PLEKHG5, RGS5, BBS5, ALDH1A1, NOMO2, CXCL6, FMNL2, ADAMTS5, CLOCK and DKK1 were among the genes included in the final network. Pathways identified included those that affect ECM properties, IOP, ciliary body functions, retinal ganglion cell viability, apoptosis, focal adhesion and oxidative stress response. The identification of many genes potentially involved in glaucoma pathology is consistent with its being a complex disease. The inclusion of several known glaucoma-related genes validates the approach used.
Collapse
Affiliation(s)
- Hamidreza Moazzeni
- School of Biology, College of Science, University of Tehran, Tehran, Iran
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mehraban Mirrahimi
- School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Abolfazl Moghadam
- School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Amir Banaei-Esfahani
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran
| | - Shahin Yazdani
- Ophthalmic Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Elahe Elahi
- School of Biology, College of Science, University of Tehran, Tehran, Iran
| |
Collapse
|
20
|
Wang Q, Kurosaka H, Kikuchi M, Nakaya A, Trainor PA, Yamashiro T. Perturbed development of cranial neural crest cells in association with reduced sonic hedgehog signaling underlies the pathogenesis of retinoic-acid-induced cleft palate. Dis Model Mech 2019; 12:dmm040279. [PMID: 31591086 PMCID: PMC6826016 DOI: 10.1242/dmm.040279] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 08/29/2019] [Indexed: 12/19/2022] Open
Abstract
Cleft palate (CP) is one of the most common congenital craniofacial anomalies in humans and can be caused by either single or multiple genetic and environmental factor(s). With respect to environmental factors, excessive intake of vitamin A during early pregnancy is associated with increased incidence of CP in offspring both in humans and in animal models. Vitamin A is metabolized to retinoic acid (RA); however, the pathogenetic mechanism of CP caused by altered RA signaling during early embryogenesis is not fully understood. To investigate the detailed cellular and molecular mechanism of RA-induced CP, we administered all-trans RA to pregnant mice at embryonic day (E)8.5. In the RA-treated group, we observed altered expression of Sox10, which marks cranial neural crest cells (CNCCs). Disruption of Sox10 expression was also observed at E10.5 in the maxillary component of the first branchial arch, which gives rise to secondary palatal shelves. Moreover, we found significant elevation of CNCC apoptosis in RA-treated embryos. RNA-sequencing comparisons of RA-treated embryos compared to controls revealed alterations in Sonic hedgehog (Shh) signaling. More specifically, the expression of Shh and its downstream genes Ptch1 and Gli1 was spatiotemporally downregulated in the developing face of RA-treated embryos. Consistent with these findings, the incidence of CP in association with excessive RA signaling was reduced by administration of the Shh signaling agonist SAG (Smoothened agonist). Altogether, our results uncovered a novel mechanistic association between RA-induced CP with decreased Shh signaling and elevated CNCC apoptosis.
Collapse
Affiliation(s)
- Qi Wang
- Department of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Osaka University, Suita 565-0871, Japan
| | - Hiroshi Kurosaka
- Department of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Osaka University, Suita 565-0871, Japan
| | - Masataka Kikuchi
- Department of Genome Informatics, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan
| | - Akihiro Nakaya
- Department of Genome Informatics, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan
| | - Paul A Trainor
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Takashi Yamashiro
- Department of Orthodontics and Dentofacial Orthopedics, Graduate School of Dentistry, Osaka University, Suita 565-0871, Japan
| |
Collapse
|
21
|
Whitesell TR, Chrystal PW, Ryu JR, Munsie N, Grosse A, French CR, Workentine ML, Li R, Zhu LJ, Waskiewicz A, Lehmann OJ, Lawson ND, Childs SJ. foxc1 is required for embryonic head vascular smooth muscle differentiation in zebrafish. Dev Biol 2019; 453:34-47. [PMID: 31199900 DOI: 10.1016/j.ydbio.2019.06.005] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 05/29/2019] [Accepted: 06/09/2019] [Indexed: 11/15/2022]
Abstract
Vascular smooth muscle of the head derives from neural crest, but developmental mechanisms and early transcriptional drivers of the vSMC lineage are not well characterized. We find that in early development, the transcription factor foxc1b is expressed in mesenchymal cells that associate with the vascular endothelium. Using timelapse imaging, we observe that foxc1b expressing mesenchymal cells differentiate into acta2 expressing vascular mural cells. We show that in zebrafish, while foxc1b is co-expressed in acta2 positive smooth muscle cells that associate with large diameter vessels, it is not co-expressed in capillaries where pdgfrβ positive pericytes are located. In addition to being an early marker of the lineage, foxc1 is essential for vSMC differentiation; we find that foxc1 loss of function mutants have defective vSMC differentiation and that early genetic ablation of foxc1b or acta2 expressing populations blocks vSMC differentiation. Furthermore, foxc1 is expressed upstream of acta2 and is required for acta2 expression in vSMCs. Using RNA-Seq we determine an enriched intersectional gene expression profile using dual expression of foxc1b and acta2 to identify novel vSMC markers. Taken together, our data suggests that foxc1 is a marker of vSMCs and plays a critical functional role in promoting their differentiation.
Collapse
Affiliation(s)
- Thomas R Whitesell
- Alberta Children's Hospital Research Institute, University of Calgary, Canada; Department of Biochemistry and Molecular Biology, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, Canada, T2N 4N1
| | - Paul W Chrystal
- Departments of Ophthalmology, and Medical Genetics, University of Alberta, Edmonton, Alberta, Canada; Department of Biological Sciences, CW405, Biological Sciences Bldg., 11455, Saskatchewan Dr., University of Alberta, Edmonton, AB, T6G 2E9, Canada; Women & Children's Health Research Institute, ECHA 4-081, 11405 87, Ave NW, University of Alberta, Edmonton, AB, T6G 1C9, Canada; Neurosciences and Mental Health Institute, 4-120 Katz Group Centre, University of Alberta, Edmonton, AB, T6G 2E1, Canada
| | - Jae-Ryeon Ryu
- Alberta Children's Hospital Research Institute, University of Calgary, Canada; Department of Biochemistry and Molecular Biology, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, Canada, T2N 4N1
| | - Nicole Munsie
- Alberta Children's Hospital Research Institute, University of Calgary, Canada; Department of Biochemistry and Molecular Biology, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, Canada, T2N 4N1
| | - Ann Grosse
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Medical School, 364 Plantation Street, Worcester, MA, USA, 01605
| | - Curtis R French
- Department of Biological Sciences, CW405, Biological Sciences Bldg., 11455, Saskatchewan Dr., University of Alberta, Edmonton, AB, T6G 2E9, Canada; Women & Children's Health Research Institute, ECHA 4-081, 11405 87, Ave NW, University of Alberta, Edmonton, AB, T6G 1C9, Canada; Neurosciences and Mental Health Institute, 4-120 Katz Group Centre, University of Alberta, Edmonton, AB, T6G 2E1, Canada
| | - Matthew L Workentine
- Faculty of Veterinary Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, Canada, T2N 4N1
| | - Rui Li
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Medical School, 364 Plantation Street, Worcester, MA, USA, 01605
| | - Lihua Julie Zhu
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Medical School, 364 Plantation Street, Worcester, MA, USA, 01605; Program in Bioinformatics and Integrative Biology, Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, USA, 01605
| | - Andrew Waskiewicz
- Department of Biological Sciences, CW405, Biological Sciences Bldg., 11455, Saskatchewan Dr., University of Alberta, Edmonton, AB, T6G 2E9, Canada; Women & Children's Health Research Institute, ECHA 4-081, 11405 87, Ave NW, University of Alberta, Edmonton, AB, T6G 1C9, Canada; Neurosciences and Mental Health Institute, 4-120 Katz Group Centre, University of Alberta, Edmonton, AB, T6G 2E1, Canada
| | - Ordan J Lehmann
- Departments of Ophthalmology, and Medical Genetics, University of Alberta, Edmonton, Alberta, Canada
| | - Nathan D Lawson
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Medical School, 364 Plantation Street, Worcester, MA, USA, 01605
| | - Sarah J Childs
- Alberta Children's Hospital Research Institute, University of Calgary, Canada; Department of Biochemistry and Molecular Biology, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta, Canada, T2N 4N1.
| |
Collapse
|
22
|
Lin LQ, Bai SS, Wei M. Application of computer-assisted navigation in treating congenital maxillomandibular syngnathia: A case report. World J Clin Cases 2019; 7:650-655. [PMID: 30863765 PMCID: PMC6406194 DOI: 10.12998/wjcc.v7.i5.650] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 01/28/2019] [Accepted: 01/30/2019] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Congenital maxillomandibular syngnathia is an extremely rare disorder characterized by craniofacial malformations and inability to open the mouth adequately, which leads to problems with feeding, swallowing, and breathing as well as temporomandibular joint ankylosis. The main goal of the surgery is to release the ankylosis, establish functioning mandible, and prevent re-fusion. However, surgical procedures for this disease are rarely reported.
CASE SUMMARY Here, we report a 7-mo-old girl with bilateral maxillomandibular syngnathia. The patient presented with difficulty in feeding, breathing, sounding, and swallowing and had developmental dysplasia. For treatment, we performed bone isolation by computer-assisted navigation and used silicone to fix the wound surface to prevent refusion of bone. To our knowledge, this is the only syngnathia case in the literature treated using computer-assisted navigation. With the guidance of precise navigation, we were able to minimize operation time by at least one hour, the patient's blood vessels, nerves, and tooth germs were well protected, and excessive bleeding was avoided. After six weeks, the patient showed improvement in mouth opening and no major issues of feeding.
CONCLUSION Application of computer-assisted navigation can significantly improve accuracy, effectiveness, and surgical safety in correcting congenital maxillomandibular syngnathia.
Collapse
Affiliation(s)
- Li-Qin Lin
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Shan-Shan Bai
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Min Wei
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| |
Collapse
|
23
|
Manocha S, Farokhnia N, Khosropanah S, Bertol JW, Santiago J, Fakhouri WD. Systematic review of hormonal and genetic factors involved in the nonsyndromic disorders of the lower jaw. Dev Dyn 2019; 248:162-172. [PMID: 30576023 DOI: 10.1002/dvdy.8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 11/30/2018] [Accepted: 12/14/2018] [Indexed: 12/14/2022] Open
Abstract
Mandibular disorders are among the most common birth defects in humans, yet the etiological factors are largely unknown. Most of the neonates affected by mandibular abnormalities have a sequence of secondary anomalies, including airway obstruction and feeding problems, that reduce the quality of life. In the event of lacking corrective surgeries, patients with mandibular congenital disorders suffer from additional lifelong problems such as sleep apnea and temporomandibular disorders, among others. The goal of this systematic review is to gather evidence on hormonal and genetic factors that are involved in signaling pathways and interactions that are potentially associated with the nonsyndromic mandibular disorders. We found that members of FGF and BMP pathways, including FGF8/10, FGFR2/3, BMP2/4/7, BMPR1A, ACVR1, and ACVR2A/B, have a prominent number of gene-gene interactions among all identified genes in this review. Gene ontology of the 154 genes showed that the functional gene sets are involved in all aspects of cellular processes and organogenesis. Some of the genes identified by the genome-wide association studies of common mandibular disorders are involved in skeletal formation and growth retardation based on animal models, suggesting a potential direct role as genetic risk factors in the common complex jaw disorders. Developmental Dynamics 248:162-172, 2019. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Srishti Manocha
- Center for Craniofacial Research, Department of Diagnostic and Biomedical Sciences, School of Dentistry, University of Texas Health Science Center at Houston, Houston, Texas
| | - Nadia Farokhnia
- Center for Craniofacial Research, Department of Diagnostic and Biomedical Sciences, School of Dentistry, University of Texas Health Science Center at Houston, Houston, Texas
| | - Sepideh Khosropanah
- Ostrow School of Dentistry, University of Southern California, California, Los Angeles
| | - Jessica W Bertol
- Center for Craniofacial Research, Department of Diagnostic and Biomedical Sciences, School of Dentistry, University of Texas Health Science Center at Houston, Houston, Texas
| | - Joel Santiago
- Pró-Reitoria de Pesquisa e Pós-graduação (PRPPG), Universidade do Sagrado Coração, Jardim Brasil, Bauru, Sao Paulo, Brazil
| | - Walid D Fakhouri
- Center for Craniofacial Research, Department of Diagnostic and Biomedical Sciences, School of Dentistry, University of Texas Health Science Center at Houston, Houston, Texas.,Department of Pediatrics, McGovern Medical School, University of Texas Health Science Center, Houston, Texas
| |
Collapse
|
24
|
Lu P, Yin B, Liu L. MicroRNA-138 Suppresses Osteoblastic Differentiation of Valvular Interstitial Cells in Degenerative Calcific Aortic Valve Disease. Int Heart J 2018; 60:136-144. [PMID: 30464116 DOI: 10.1536/ihj.18-086] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The aim of this study was to explore the function of miR-138 in the pathogenesis of degenerative calcific aortic valve disease (DCAVD).Aortic valve calcification tissue and normal tissue from DCAVD patients were collected to detect the expression of miR-138 by qRT-PCR, and immunohistochemical staining was performed to identify the phenotype of valve interstitial cells. QRT-PCR was performed to analyze the expression of miR-138, Runx2, MSX2, and ALP at day 7 after osteogenic differentiation. Alkaline phosphatase activity assay was performed at day 14 after osteogenic differentiation. Alizarin red staining was used to analyze the calcium nodule formation. TargetScan was used to predict potential targets of miR-138. QRT-PCR and Western blotting were performed to analyze the expression of FOXC1 in valve interstitial cells (VICs). The aortic valve calcification was evaluated by quantitative analysis of the velocity in the aortic annulus and transvalvular pressure gradients.In this study, we demonstrated the role of miR-138 in VIC osteogenesis. QRT-PCR results revealed miR-138 was significantly down-regulated in calcified aortic valves compared with non-calcified valves. MiR-138 overexpression inhibited VIC osteogenic differentiation in vitro, while down-regulation of miR-138 enhanced the process. Target prediction analysis and dual-luciferase reporter assay confirmed FOXC1 was a direct target of miR-138. Further research found FOXC1 overexpression promoted VIC osteogenic differentiation. In addition, animal experiments validated indirectly miR-138 could suppress aortic valve calcification.Our findings suggest miR-138 could function as a new inhibitor of VIC osteogenic differentiation, which may act by targeting FOXC1.
Collapse
Affiliation(s)
- Ping Lu
- Department of Cardiac Surgery, Qianfoshan Hospital, Shandong University
| | - Beibei Yin
- Department of Oncology, Qianfoshan Hospital, Shandong University
| | - Luqi Liu
- Department of Cardiac Surgery, Qianfoshan Hospital, Shandong University
| |
Collapse
|
25
|
Transcriptome analysis of Xenopus orofacial tissues deficient in retinoic acid receptor function. BMC Genomics 2018; 19:795. [PMID: 30390632 PMCID: PMC6215681 DOI: 10.1186/s12864-018-5186-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 10/18/2018] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Development of the face and mouth is orchestrated by a large number of transcription factors, signaling pathways and epigenetic regulators. While we know many of these regulators, our understanding of how they interact with each other and implement changes in gene expression during orofacial development is still in its infancy. Therefore, this study focuses on uncovering potential cooperation between transcriptional regulators and one important signaling pathway, retinoic acid, during development of the midface. RESULTS Transcriptome analyses was performed on facial tissues deficient for retinoic acid receptor function at two time points in development; early (35 hpf) just after the neural crest migrates and facial tissues are specified and later (60 hpf) when the mouth has formed and facial structures begin to differentiate. Functional and network analyses revealed that retinoic acid signaling could cooperate with novel epigenetic factors and calcium-NFAT signaling during early orofacial development. At the later stage, retinoic acid may work with WNT and BMP and regulate homeobox containing transcription factors. Finally, there is an overlap in genes dysregulated in Xenopus embryos with median clefts with human genes associated with similar orofacial defects. CONCLUSIONS This study uncovers novel signaling pathways required for orofacial development as well as pathways that could interact with retinoic acid signaling during the formation of the face. We show that frog faces are an important tool for studying orofacial development and birth defects.
Collapse
|
26
|
Davidson AJ, Lewis P, Przepiorski A, Sander V. Turning mesoderm into kidney. Semin Cell Dev Biol 2018; 91:86-93. [PMID: 30172050 DOI: 10.1016/j.semcdb.2018.08.016] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 08/24/2018] [Accepted: 08/28/2018] [Indexed: 02/07/2023]
Abstract
The intermediate mesoderm is located between the somites and the lateral plate mesoderm and gives rise to renal progenitors that contribute to the three mammalian kidney types (pronephros, mesonephros and metanephros). In this review, focusing largely on murine kidney development, we examine how the intermediate mesoderm forms during gastrulation/axis elongation and how it progressively gives rise to distinct renal progenitors along the rostro-caudal axis. We highlight some of the potential signalling cues and core transcription factor circuits that direct these processes, up to the point of early metanephric kidney formation.
Collapse
Affiliation(s)
- Alan J Davidson
- Department of Molecular Medicine & Pathology, School of Medical Sciences, Faculty of Medical & Health Sciences, The University of Auckland, Private Bag 921019, Auckland 1142, New Zealand.
| | - Paula Lewis
- Department of Molecular Medicine & Pathology, School of Medical Sciences, Faculty of Medical & Health Sciences, The University of Auckland, Private Bag 921019, Auckland 1142, New Zealand
| | - Aneta Przepiorski
- Department of Molecular Medicine & Pathology, School of Medical Sciences, Faculty of Medical & Health Sciences, The University of Auckland, Private Bag 921019, Auckland 1142, New Zealand
| | - Veronika Sander
- Department of Molecular Medicine & Pathology, School of Medical Sciences, Faculty of Medical & Health Sciences, The University of Auckland, Private Bag 921019, Auckland 1142, New Zealand
| |
Collapse
|
27
|
Flach H, Krieg J, Hoffmeister M, Dietmann P, Reusch A, Wischmann L, Kernl B, Riegger R, Oess S, Kühl SJ. Nosip functions during vertebrate eye and cranial cartilage development. Dev Dyn 2018; 247:1070-1082. [PMID: 30055071 DOI: 10.1002/dvdy.24659] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 07/01/2018] [Accepted: 07/13/2018] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND The nitric oxide synthase interacting protein (Nosip) has been associated with diverse human diseases including psychological disorders. In line, early neurogenesis of mouse and Xenopus is impaired upon Nosip deficiency. Nosip knockout mice show craniofacial defects and the down-regulation of Nosip in the mouse and Xenopus leads to microcephaly. Until now, the exact underlying molecular mechanisms of these malformations were still unknown. RESULTS Here, we show that nosip is expressed in the developing ocular system as well as the anterior neural crest cells of Xenopus laevis. Furthermore, Nosip inhibition causes severe defects in eye formation in the mouse and Xenopus. Retinal lamination as well as dorso-ventral patterning of the retina were affected in Nosip-depleted Xenopus embryos. Marker gene analysis using rax, pax6 and otx2 reveals an interference with the eye field induction and differentiation. A closer look on Nosip-deficient Xenopus embryos furthermore reveals disrupted cranial cartilage structures and an inhibition of anterior neural crest cell induction and migration shown by twist, snai2, and egr2. Moreover, foxc1 as downstream factor of retinoic acid signalling is affected upon Nosip deficiency. CONCLUSIONS Nosip is a crucial factor for the development of anterior neural tissue such the eyes and neural crest cells. Developmental Dynamics 247:1070-1082, 2018. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Hannah Flach
- Institute of Biochemistry and Molecular Biology, Ulm University, Ulm, Germany
| | - Julia Krieg
- Institute of Biochemistry and Molecular Biology, Ulm University, Ulm, Germany
| | - Meike Hoffmeister
- Institute of Biochemistry II, Goethe University, Frankfurt Medical School, Frankfurt/Main, Germany.,Institute of Biochemistry, Brandenburg Medical School (MHB) Theodor Fontane, Neuruppin, Germany
| | - Petra Dietmann
- Institute of Biochemistry and Molecular Biology, Ulm University, Ulm, Germany
| | - Adrian Reusch
- Institute of Biochemistry and Molecular Biology, Ulm University, Ulm, Germany
| | - Lisa Wischmann
- Institute of Biochemistry and Molecular Biology, Ulm University, Ulm, Germany
| | - Bianka Kernl
- Institute of Biochemistry and Molecular Biology, Ulm University, Ulm, Germany
| | - Ricarda Riegger
- Institute of Biochemistry and Molecular Biology, Ulm University, Ulm, Germany
| | - Stefanie Oess
- Institute of Biochemistry II, Goethe University, Frankfurt Medical School, Frankfurt/Main, Germany.,Institute of Biochemistry, Brandenburg Medical School (MHB) Theodor Fontane, Neuruppin, Germany
| | - Susanne J Kühl
- Institute of Biochemistry and Molecular Biology, Ulm University, Ulm, Germany
| |
Collapse
|
28
|
Xu P, Balczerski B, Ciozda A, Louie K, Oralova V, Huysseune A, Crump JG. Fox proteins are modular competency factors for facial cartilage and tooth specification. Development 2018; 145:dev.165498. [PMID: 29777011 DOI: 10.1242/dev.165498] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 05/11/2018] [Indexed: 12/30/2022]
Abstract
Facial form depends on the precise positioning of cartilage, bone, and tooth fields in the embryonic pharyngeal arches. How complex signaling information is integrated to specify these cell types remains a mystery. We find that modular expression of Forkhead domain transcription factors (Fox proteins) in the zebrafish face arises through integration of Hh, Fgf, Bmp, Edn1 and Jagged-Notch pathways. Whereas loss of C-class Fox proteins results in reduced upper facial cartilages, loss of F-class Fox proteins results in distal jaw truncations and absent midline cartilages and teeth. We show that Fox proteins are required for Sox9a to promote chondrogenic gene expression. Fox proteins are sufficient in neural crest-derived cells for cartilage development, and neural crest-specific misexpression of Fox proteins expands the cartilage domain but inhibits bone. These results support a modular role for Fox proteins in establishing the competency of progenitors to form cartilage and teeth in the face.
Collapse
Affiliation(s)
- Pengfei Xu
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California Keck School of Medicine, Los Angeles, CA 90033, USA
| | - Bartosz Balczerski
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California Keck School of Medicine, Los Angeles, CA 90033, USA
| | - Amanda Ciozda
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California Keck School of Medicine, Los Angeles, CA 90033, USA
| | - Kristin Louie
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California Keck School of Medicine, Los Angeles, CA 90033, USA
| | - Veronika Oralova
- Evolutionary Developmental Biology, Ghent University, B-9000 Ghent, Belgium
| | - Ann Huysseune
- Evolutionary Developmental Biology, Ghent University, B-9000 Ghent, Belgium
| | - J Gage Crump
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California Keck School of Medicine, Los Angeles, CA 90033, USA
| |
Collapse
|
29
|
Rubio-Palau J, Prieto-Gundin A, de Abreu Graterol LM, Vercruysse H. Maxillomandibular Syngnathia: 3D Planning and Review of the Literature. Craniomaxillofac Trauma Reconstr 2018; 11:124-130. [PMID: 29892327 DOI: 10.1055/s-0037-1606248] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 04/29/2017] [Indexed: 10/19/2022] Open
Abstract
Congenital maxillomandibular fusion or syngnathia is a rare craniofacial disorder with only 26 cases reported in the literature. We present a case of a congenital complex zygomatico-mandibular syngnathia associated with a palatal cleft, posterior maxilla and turbinate agenesia, mild hemifacial microsomia, and a disordered dental eruption. The patient has the highest age (15 years) at diagnosis described in the literature. 3D planning of the surgery was performed to study the patient's anatomy and design the necessary osteotomies to separate the jaws. En bloc removal of the fused fragment with bilateral coronoidectomy and aggressive long-term physiotherapy for 3 months led to a stable increase in mouth opening from 0 to 21 mm inter-incisor distance. The patient reported an improvement in speech, was able to eat without restriction regarding food consistency, and could maintain a good oral hygiene.
Collapse
Affiliation(s)
- Josep Rubio-Palau
- Division of Maxillofacial Surgery, Department of Pediatric Surgery, Hospital Sant Joan de Deu, Esplugues de Llobregat, Barcelona, Spain.,Department of Oral Medicine and Oral Public Health, Universitat Internacional de Catalunya, Barcelona, Catalunya, Spain
| | - Alejandra Prieto-Gundin
- Department of Pediatric Anesthesiology, Hospital Sant Joan de Deu, Esplugues de Llobregat, Barcelona, Spain
| | | | - Herman Vercruysse
- Department of Craniofacial Surgery, Great Ormond Street Hospital for Children, NHS Trust, London, United Kingdom
| |
Collapse
|
30
|
Van Otterloo E, Li H, Jones KL, Williams T. AP-2α and AP-2β cooperatively orchestrate homeobox gene expression during branchial arch patterning. Development 2018; 145:dev157438. [PMID: 29229773 PMCID: PMC5825845 DOI: 10.1242/dev.157438] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 12/05/2017] [Indexed: 12/19/2022]
Abstract
The evolution of a hinged moveable jaw with variable morphology is considered a major factor behind the successful expansion of the vertebrates. DLX homeobox transcription factors are crucial for establishing the positional code that patterns the mandible, maxilla and intervening hinge domain, but how the genes encoding these proteins are regulated remains unclear. Herein, we demonstrate that the concerted action of the AP-2α and AP-2β transcription factors within the mouse neural crest is essential for jaw patterning. In the absence of these two proteins, the hinge domain is lost and there are alterations in the size and patterning of the jaws correlating with dysregulation of homeobox gene expression, with reduced levels of Emx, Msx and Dlx paralogs accompanied by an expansion of Six1 expression. Moreover, detailed analysis of morphological features and gene expression changes indicate significant overlap with various compound Dlx gene mutants. Together, these findings reveal that the AP-2 genes have a major function in mammalian neural crest development, influencing patterning of the craniofacial skeleton via the DLX code, an effect that has implications for vertebrate facial evolution, as well as for human craniofacial disorders.
Collapse
Affiliation(s)
- Eric Van Otterloo
- Department of Craniofacial Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Hong Li
- Department of Craniofacial Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Kenneth L Jones
- Department of Pediatrics, Section of Hematology, Oncology, and Bone Marrow Transplant, University of Colorado School of Medicine, Aurora, CO 80045 USA
| | - Trevor Williams
- Department of Craniofacial Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Children's Hospital Colorado, Aurora, CO 80045, USA
| |
Collapse
|
31
|
Tavares ALP, Cox TC, Maxson RM, Ford HL, Clouthier DE. Negative regulation of endothelin signaling by SIX1 is required for proper maxillary development. Development 2017; 144:2021-2031. [PMID: 28455376 DOI: 10.1242/dev.145144] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 04/18/2017] [Indexed: 12/20/2022]
Abstract
Jaw morphogenesis is a complex event mediated by inductive signals that establish and maintain the distinct developmental domains required for formation of hinged jaws, the defining feature of gnathostomes. The mandibular portion of pharyngeal arch 1 is patterned dorsally by Jagged-Notch signaling and ventrally by endothelin receptor A (EDNRA) signaling. Loss of EDNRA signaling disrupts normal ventral gene expression, the result of which is homeotic transformation of the mandible into a maxilla-like structure. However, loss of Jagged-Notch signaling does not result in significant changes in maxillary development. Here we show in mouse that the transcription factor SIX1 regulates dorsal arch development not only by inducing dorsal Jag1 expression but also by inhibiting endothelin 1 (Edn1) expression in the pharyngeal endoderm of the dorsal arch, thus preventing dorsal EDNRA signaling. In the absence of SIX1, but not JAG1, aberrant EDNRA signaling in the dorsal domain results in partial duplication of the mandible. Together, our results illustrate that SIX1 is the central mediator of dorsal mandibular arch identity, thus ensuring separation of bone development between the upper and lower jaws.
Collapse
Affiliation(s)
- Andre L P Tavares
- Department of Craniofacial Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Timothy C Cox
- Department of Pediatrics (Craniofacial Medicine), University of Washington, and Center for Developmental Biology & Regenerative Medicine, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Robert M Maxson
- Department of Biochemistry and Molecular Biology and Norris Cancer Center, University of Southern California, Los Angeles, CA 87654, USA
| | - Heide L Ford
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - David E Clouthier
- Department of Craniofacial Biology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
32
|
Fontana P, Tortora C, Petillo R, Malacarne M, Cavani S, Miniero M, D'Ambrosio P, De Brasi D, Pisanti MA. Brachydactyly type E in an Italian family with 6p25 trisomy. Eur J Med Genet 2017; 60:195-199. [PMID: 28111183 DOI: 10.1016/j.ejmg.2017.01.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 01/10/2017] [Accepted: 01/18/2017] [Indexed: 11/19/2022]
Abstract
Brachydactyly type E is a congenital limb malformation characterized by small hands and feet as a result of shortened metacarpals and metatarsals. Genetic causes of this anomaly are heterogeneous and only partially characterized. In this report we describe an Italian family in which four subjects share brachydactyly type E and a 3 Mb microduplication in region 6p25. The duplication involves the gene FOXC1, expressed during the osteoblast differentiation, which appears a potential candidate gene for brachydactyly.
Collapse
Affiliation(s)
- Paolo Fontana
- Department of Molecular Medicine and Medical Biotechnologies, Federico II University of Naples, Naples, Italy.
| | - Cristina Tortora
- Department of Molecular Medicine and Medical Biotechnologies, Federico II University of Naples, Naples, Italy
| | - Roberta Petillo
- Cardiomiology and Medical Genetics, Department of Experimental Medicine, Second University of Naples, Naples, Italy
| | | | - Simona Cavani
- Division of Medical Genetics, Galliera Hospital, Genoa, Italy
| | - Martina Miniero
- Department of Molecular Medicine and Medical Biotechnologies, Federico II University of Naples, Naples, Italy
| | - Paola D'Ambrosio
- Cardiomiology and Medical Genetics, Department of Experimental Medicine, Second University of Naples, Naples, Italy
| | - Davide De Brasi
- Service of Medical Genetics, Cardarelli Hospital, Naples, Italy
| | | |
Collapse
|
33
|
Smeeton J, Askary A, Crump JG. Building and maintaining joints by exquisite local control of cell fate. WILEY INTERDISCIPLINARY REVIEWS. DEVELOPMENTAL BIOLOGY 2017; 6:10.1002/wdev.245. [PMID: 27581688 PMCID: PMC5877473 DOI: 10.1002/wdev.245] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 06/30/2016] [Accepted: 07/01/2016] [Indexed: 12/18/2022]
Abstract
We owe the flexibility of our bodies to sophisticated articulations between bones. Establishment of these joints requires the integration of multiple tissue types: permanent cartilage that cushions the articulating bones, synovial membranes that enclose a lubricating fluid-filled cavity, and a fibrous capsule and ligaments that provide structural support. Positioning the prospective joint region involves establishment of an "interzone" region of joint progenitor cells within a nascent cartilage condensation, which is achieved through the interplay of activators and inhibitors of multiple developmental signaling pathways. Within the interzone, tight regulation of BMP and TGFβ signaling prevents the hypertrophic maturation of joint chondrocytes, in part through downstream transcriptional repressors and epigenetic modulators. Synovial cells then acquire further specializations through expression of genes that promote lubrication, as well as the formation of complex structures such as cavities and entheses. Whereas genetic investigations in mice and humans have uncovered a number of regulators of joint development and homeostasis, recent work in zebrafish offers a complementary reductionist approach toward understanding joint positioning and the regulation of chondrocyte fate at joints. The complexity of building and maintaining joints may help explain why there are still few treatments for osteoarthritis, one of the most common diseases in the human population. A major challenge will be to understand how developmental abnormalities in joint structure, as well as postnatal roles for developmental genes in joint homeostasis, contribute to birth defects and degenerative diseases of joints. WIREs Dev Biol 2017, 6:e245. doi: 10.1002/wdev.245 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Joanna Smeeton
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California Keck School of Medicine, Los Angeles, CA, USA
| | - Amjad Askary
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California Keck School of Medicine, Los Angeles, CA, USA
| | - J. Gage Crump
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California Keck School of Medicine, Los Angeles, CA, USA
| |
Collapse
|
34
|
Abstract
The mammalian ear is a complex structure divided into three main parts: the outer; middle; and inner ear. These parts are formed from all three germ layers and neural crest cells, which have to integrate successfully in order to form a fully functioning organ of hearing. Any defect in development of the outer and middle ear leads to conductive hearing loss, while defects in the inner ear can lead to sensorineural hearing loss. This review focuses on the development of the parts of the ear involved with sound transduction into the inner ear, and the parts largely ignored in the world of hearing research: the outer and middle ear. The published data on the embryonic origin, signalling, genetic control, development and timing of the mammalian middle and outer ear are reviewed here along with new data showing the Eustachian tube cartilage is of dual embryonic origin. The embryonic origin of some of these structures has only recently been uncovered (Science, 339, 2013, 1453; Development, 140, 2013, 4386), while the molecular mechanisms controlling the growth, structure and integration of many outer and middle ear components are hardly known. The genetic analysis of outer and middle ear development is rather limited, with a small number of genes often affecting either more than one part of the ear or having only very small effects on development. This review therefore highlights the necessity for further research into the development of outer and middle ear structures, which will be important for the understanding and treatment of conductive hearing loss.
Collapse
Affiliation(s)
- Neal Anthwal
- Craniofacial Development and Stem Cell BiologyKing's College LondonLondonUK
| | - Hannah Thompson
- Craniofacial Development and Stem Cell BiologyKing's College LondonLondonUK
| |
Collapse
|
35
|
Hopkins A, Mirzayans F, Berry F. Foxc1 Expression in Early Osteogenic Differentiation Is Regulated by BMP4-SMAD Activity. J Cell Biochem 2016; 117:1707-17. [DOI: 10.1002/jcb.25464] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 12/11/2015] [Indexed: 01/01/2023]
Affiliation(s)
- Alexander Hopkins
- Department of Surgery; University of Alberta; Edmonton Alberta Canada
| | - Freda Mirzayans
- Department of Medical Genetics; University of Alberta; Edmonton Alberta Canada
| | - Fred Berry
- Department of Surgery; University of Alberta; Edmonton Alberta Canada
- Department of Medical Genetics; University of Alberta; Edmonton Alberta Canada
| |
Collapse
|
36
|
Fish JL. Developmental mechanisms underlying variation in craniofacial disease and evolution. Dev Biol 2015; 415:188-197. [PMID: 26724698 DOI: 10.1016/j.ydbio.2015.12.019] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 12/17/2015] [Accepted: 12/21/2015] [Indexed: 01/14/2023]
Abstract
Craniofacial disease phenotypes exhibit significant variation in penetrance and severity. Although many genetic contributions to phenotypic variation have been identified, genotype-phenotype correlations remain imprecise. Recent work in evolutionary developmental biology has exposed intriguing developmental mechanisms that potentially explain incongruities in genotype-phenotype relationships. This review focuses on two observations from work in comparative and experimental animal model systems that highlight how development structures variation. First, multiple genetic inputs converge on relatively few developmental processes. Investigation of when and how variation in developmental processes occurs may therefore help predict potential genetic interactions and phenotypic outcomes. Second, genetic mutation is typically associated with an increase in phenotypic variance. Several models outlining developmental mechanisms underlying mutational increases in phenotypic variance are discussed using Satb2-mediated variation in jaw size as an example. These data highlight development as a critical mediator of genotype-phenotype correlations. Future research in evolutionary developmental biology focusing on tissue-level processes may help elucidate the "black box" between genotype and phenotype, potentially leading to novel treatment, earlier diagnoses, and better clinical consultations for individuals affected by craniofacial anomalies.
Collapse
Affiliation(s)
- Jennifer L Fish
- University of Massachusetts Lowell, Department of Biological Sciences, 198 Riverside Street, Olsen Hall, Room 619, Lowell, MA 01854, United States.
| |
Collapse
|
37
|
Suzuki A, Sangani DR, Ansari A, Iwata J. Molecular mechanisms of midfacial developmental defects. Dev Dyn 2015; 245:276-93. [PMID: 26562615 DOI: 10.1002/dvdy.24368] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 10/19/2015] [Accepted: 11/01/2015] [Indexed: 12/22/2022] Open
Abstract
The morphogenesis of midfacial processes requires the coordination of a variety of cellular functions of both mesenchymal and epithelial cells to develop complex structures. Any failure or delay in midfacial development as well as any abnormal fusion of the medial and lateral nasal and maxillary prominences will result in developmental defects in the midface with a varying degree of severity, including cleft, hypoplasia, and midline expansion. Despite the advances in human genome sequencing technology, the causes of nearly 70% of all birth defects, which include midfacial development defects, remain unknown. Recent studies in animal models have highlighted the importance of specific signaling cascades and genetic-environmental interactions in the development of the midfacial region. This review will summarize the current understanding of the morphogenetic processes and molecular mechanisms underlying midfacial birth defects based on mouse models with midfacial developmental abnormalities.
Collapse
Affiliation(s)
- Akiko Suzuki
- Department of Diagnostic & Biomedical Sciences, School of Dentistry, The University of Texas Health Science Center at Houston, Houston, Texas.,Center for Craniofacial Research, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Dhruvee R Sangani
- Center for Craniofacial Research, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Afreen Ansari
- Center for Craniofacial Research, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Junichi Iwata
- Department of Diagnostic & Biomedical Sciences, School of Dentistry, The University of Texas Health Science Center at Houston, Houston, Texas.,Center for Craniofacial Research, The University of Texas Health Science Center at Houston, Houston, Texas.,The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas
| |
Collapse
|
38
|
Mapping of Craniofacial Traits in Outbred Mice Identifies Major Developmental Genes Involved in Shape Determination. PLoS Genet 2015; 11:e1005607. [PMID: 26523602 PMCID: PMC4629907 DOI: 10.1371/journal.pgen.1005607] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 09/24/2015] [Indexed: 02/05/2023] Open
Abstract
The vertebrate cranium is a prime example of the high evolvability of complex traits. While evidence of genes and developmental pathways underlying craniofacial shape determination is accumulating, we are still far from understanding how such variation at the genetic level is translated into craniofacial shape variation. Here we used 3D geometric morphometrics to map genes involved in shape determination in a population of outbred mice (Carworth Farms White, or CFW). We defined shape traits via principal component analysis of 3D skull and mandible measurements. We mapped genetic loci associated with shape traits at ~80,000 candidate single nucleotide polymorphisms in ~700 male mice. We found that craniofacial shape and size are highly heritable, polygenic traits. Despite the polygenic nature of the traits, we identified 17 loci that explain variation in skull shape, and 8 loci associated with variation in mandible shape. Together, the associated variants account for 11.4% of skull and 4.4% of mandible shape variation, however, the total additive genetic variance associated with phenotypic variation was estimated in ~45%. Candidate genes within the associated loci have known roles in craniofacial development; this includes 6 transcription factors and several regulators of bone developmental pathways. One gene, Mn1, has an unusually large effect on shape variation in our study. A knockout of this gene was previously shown to affect negatively the development of membranous bones of the cranial skeleton, and evolutionary analysis shows that the gene has arisen at the base of the bony vertebrates (Eutelostomi), where the ossified head first appeared. Therefore, Mn1 emerges as a key gene for both skull formation and within-population shape variation. Our study shows that it is possible to identify important developmental genes through genome-wide mapping of high-dimensional shape features in an outbred population. Formation of the face, mandible, and skull is determined in part by genetic factors, but the relationship between genetic variation and craniofacial development is not well understood. We demonstrate how recent advances in mouse genomics and statistical methods can be used to identify genes involved in craniofacial development. We use outbred mice together with a dense panel of genetic markers to identify genetic loci affecting craniofacial shape. Some of the loci we identify are also known from past studies to contribute to craniofacial development and bone formation. For example, the top candidate gene identified in this study, Mn1, is a gene that appeared at a time when animals started to form bony skulls, suggesting that it may be a key gene in this evolutionary innovation. This further suggests that Mn1 and other genes involved in head formation are also responsible for more fine-grained regulation of its shape. Our results confirm that the outbred mouse population used in this study is suitable to identify single genetic factors even under conditions where many genes cooperate to generate a complex phenotype.
Collapse
|
39
|
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
|
40
|
Hinton RJ, Jing J, Feng JQ. Genetic Influences on Temporomandibular Joint Development and Growth. Curr Top Dev Biol 2015; 115:85-109. [PMID: 26589922 DOI: 10.1016/bs.ctdb.2015.07.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The temporomandibular joint (TMJ) is a small synovial joint at which the mandible articulates with the skull during movements involved in speaking and mastication. However, the secondary cartilage lining its joint surfaces is indicative of a very different developmental history than limb cartilages. This review summarizes our current knowledge of genes that regulate the formation of primary components of the TMJ, as well as genes that regulate postnatal growth of the TMJ. Although the TMJ is regulated by some of the same genes that are important in limb joints, others appear unique to the TMJ or have different actions. Runx2, Sox9, and members of the TGF-β/BMP family are critical drivers of chondrogenesis during condylar cartilage morphogenesis, and Indian hedgehog (Ihh) is important for formation of the articular disc and cavitation. Osterix (Osx) is a critical regulator of endochondral bone formation during postnatal TMJ growth.
Collapse
Affiliation(s)
- Robert J Hinton
- Department of Biomedical Sciences, Texas A&M Baylor College of Dentistry, Dallas, Texas, USA.
| | - Junjun Jing
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Sichuan, PR China
| | - Jian Q Feng
- Department of Biomedical Sciences, Texas A&M Baylor College of Dentistry, Dallas, Texas, USA
| |
Collapse
|
41
|
Tavares ALP, Artinger KB, Clouthier DE. Regulating Craniofacial Development at the 3' End: MicroRNAs and Their Function in Facial Morphogenesis. Curr Top Dev Biol 2015; 115:335-75. [PMID: 26589932 DOI: 10.1016/bs.ctdb.2015.08.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Defects in craniofacial development represent a majority of observed human birth defects, occurring at a rate as high as 1:800 live births. These defects often occur due to changes in neural crest cell (NCC) patterning and development and can affect non-NCC-derived structures due to interactions between NCCs and the surrounding cell types. Proper craniofacial development requires an intricate array of gene expression networks that are tightly controlled spatiotemporally by a number of regulatory mechanisms. One of these mechanisms involves the action of microRNAs (miRNAs), a class of noncoding RNAs that repress gene expression by binding to miRNA recognition sequences typically located in the 3' UTR of target mRNAs. Recent evidence illustrates that miRNAs are crucial for vertebrate facial morphogenesis, with changes in miRNA expression leading to facial birth defects, including some in complex human syndromes such as 22q11 (DiGeorge Syndrome). In this review, we highlight the current understanding of miRNA biogenesis, the roles of miRNAs in overall craniofacial development, the impact that loss of miRNAs has on normal development and the requirement for miRNAs in the development of specific craniofacial structures, including teeth. From these studies, it is clear that miRNAs are essential for normal facial development and morphogenesis, and a potential key in establishing new paradigms for repair and regeneration of facial defects.
Collapse
Affiliation(s)
- Andre L P Tavares
- Department of Craniofacial Biology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Kristin B Artinger
- Department of Craniofacial Biology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - David E Clouthier
- Department of Craniofacial Biology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.
| |
Collapse
|
42
|
Affiliation(s)
- P Purcell
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - P A Trainor
- Stowers Institute for Medical Research, Kansas City, MO, USA Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
43
|
Liu CF, Lefebvre V. The transcription factors SOX9 and SOX5/SOX6 cooperate genome-wide through super-enhancers to drive chondrogenesis. Nucleic Acids Res 2015; 43:8183-203. [PMID: 26150426 PMCID: PMC4787819 DOI: 10.1093/nar/gkv688] [Citation(s) in RCA: 200] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 06/24/2015] [Indexed: 12/21/2022] Open
Abstract
SOX9 is a transcriptional activator required for chondrogenesis, and SOX5 and SOX6 are closely related DNA-binding proteins that critically enhance its function. We use here genome-wide approaches to gain novel insights into the full spectrum of the target genes and modes of action of this chondrogenic trio. Using the RCS cell line as a faithful model for proliferating/early prehypertrophic growth plate chondrocytes, we uncover that SOX6 and SOX9 bind thousands of genomic sites, frequently and most efficiently near each other. SOX9 recognizes pairs of inverted SOX motifs, whereas SOX6 favors pairs of tandem SOX motifs. The SOX proteins primarily target enhancers. While binding to a small fraction of typical enhancers, they bind multiple sites on almost all super-enhancers (SEs) present in RCS cells. These SEs are predominantly linked to cartilage-specific genes. The SOX proteins effectively work together to activate these SEs and are required for in vivo expression of their associated genes. These genes encode key regulatory factors, including the SOX trio proteins, and all essential cartilage extracellular matrix components. Chst11, Fgfr3, Runx2 and Runx3 are among many other newly identified SOX trio targets. SOX9 and SOX5/SOX6 thus cooperate genome-wide, primarily through SEs, to implement the growth plate chondrocyte differentiation program.
Collapse
Affiliation(s)
- Chia-Feng Liu
- Department of Cellular & Molecular Medicine, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| | - Véronique Lefebvre
- Department of Cellular & Molecular Medicine, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| |
Collapse
|
44
|
Sangani D, Suzuki A, VonVille H, Hixson JE, Iwata J. Gene Mutations Associated with Temporomandibular Joint Disorders: A Systematic Review. ACTA ACUST UNITED AC 2015; 2. [PMID: 27695703 PMCID: PMC5045035 DOI: 10.4236/oalib.1101583] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Background The temporomandibular joint (TMJ) is a bilateral synovial joint between the mandible and the temporal bone of the skull. TMJ disorders (TMDs) are a set of complicated and poorly understood clinical conditions, in which TMDs are associated with a number of symptoms including pain and limited jaw movement. The increasing scientific evidence suggests that genetic factors play a significant role in the pathology of TMDs. However, the underlying mechanism of TMDs remains largely unknown. Objective The study aimed to determine the associated genes to TMDs in humans and animals. Methods The literature search was conducted through databases including Medline (Ovid), EMBASE (Ovid), and PubMed (NLM) by using scientific terms for TMDs and genetics in March 2015. Additional studies were identified by searching bibliographies of highly relevant articles and Scopus (Elsevier). Results Our systematic analyses identified 31 articles through literature searches. A total of 112 genes were identified to be significantly and specifically associated with TMDs. Conclusion Our systematic review provides a list of accurate genes associated with TMDs and suggests a genetic contribution to the pathology of TMDs.
Collapse
Affiliation(s)
- Dhruvee Sangani
- Department of Epidemiology, Human Genetics & Environmental Sciences, The University of Texas School of Public Health, Houston, TX, USA; Department of Diagnostic & Biomedical Sciences, The University of Texas Health Science Center at Houston School of Dentistry, Houston, TX, USA
| | - Akiko Suzuki
- Department of Diagnostic & Biomedical Sciences, The University of Texas Health Science Center at Houston School of Dentistry, Houston, TX, USA; Center for Craniofacial Research, The University of Texas Health Science Center at Houston School of Dentistry, Houston, TX, USA
| | - Helena VonVille
- The University of Texas School of Public Health Library, Houston, TX, USA
| | - James E Hixson
- Department of Epidemiology, Human Genetics & Environmental Sciences, The University of Texas School of Public Health, Houston, TX, USA
| | - Junichi Iwata
- Department of Diagnostic & Biomedical Sciences, The University of Texas Health Science Center at Houston School of Dentistry, Houston, TX, USA; Center for Craniofacial Research, The University of Texas Health Science Center at Houston School of Dentistry, Houston, TX, USA; The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX, USA
| |
Collapse
|
45
|
The ribosome biogenesis factor Nol11 is required for optimal rDNA transcription and craniofacial development in Xenopus. PLoS Genet 2015; 11:e1005018. [PMID: 25756904 PMCID: PMC4354908 DOI: 10.1371/journal.pgen.1005018] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 01/22/2015] [Indexed: 01/30/2023] Open
Abstract
The production of ribosomes is ubiquitous and fundamental to life. As such, it is surprising that defects in ribosome biogenesis underlie a growing number of symptomatically distinct inherited disorders, collectively called ribosomopathies. We previously determined that the nucleolar protein, NOL11, is essential for optimal pre-rRNA transcription and processing in human tissue culture cells. However, the role of NOL11 in the development of a multicellular organism remains unknown. Here, we reveal a critical function for NOL11 in vertebrate ribosome biogenesis and craniofacial development. Nol11 is strongly expressed in the developing cranial neural crest (CNC) of both amphibians and mammals, and knockdown of Xenopus nol11 results in impaired pre-rRNA transcription and processing, increased apoptosis, and abnormal development of the craniofacial cartilages. Inhibition of p53 rescues this skeletal phenotype, but not the underlying ribosome biogenesis defect, demonstrating an evolutionarily conserved control mechanism through which ribosome-impaired craniofacial cells are removed. Excessive activation of this mechanism impairs craniofacial development. Together, our findings reveal a novel requirement for Nol11 in craniofacial development, present the first frog model of a ribosomopathy, and provide further insight into the clinically important relationship between specific ribosome biogenesis proteins and craniofacial cell survival.
Collapse
|
46
|
Edlund RK, Birol O, Groves AK. The role of foxi family transcription factors in the development of the ear and jaw. Curr Top Dev Biol 2015; 111:461-95. [PMID: 25662269 DOI: 10.1016/bs.ctdb.2014.11.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The mammalian outer, middle, and inner ears have different embryonic origins and evolved at different times in the vertebrate lineage. The outer ear is derived from first and second branchial arch ectoderm and mesoderm, the middle ear ossicles are derived from neural crest mesenchymal cells that invade the first and second branchial arches, whereas the inner ear and its associated vestibule-acoustic (VIIIth) ganglion are derived from the otic placode. In this chapter, we discuss recent findings in the development of these structures and describe the contributions of members of a Forkhead transcription factor family, the Foxi family to their formation. Foxi transcription factors are critical for formation of the otic placode, survival of the branchial arch neural crest, and developmental remodeling of the branchial arch ectoderm.
Collapse
Affiliation(s)
- Renée K Edlund
- Program in Developmental Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Onur Birol
- Program in Developmental Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Andrew K Groves
- Program in Developmental Biology, Baylor College of Medicine, Houston, Texas, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA; Department of Neuroscience, Baylor College of Medicine, Houston, Texas, USA.
| |
Collapse
|
47
|
Abstract
A rare disease is defined as a condition that affects less than 1 in 2000 individuals. Currently more than 7000 rare diseases have been documented, and most are thought to be of genetic origin. Rare diseases primarily affect children, and congenital craniofacial syndromes and disorders constitute a significant proportion of rare diseases, with over 700 having been described to date. Modeling craniofacial disorders in animal models has been instrumental in uncovering the etiology and pathogenesis of numerous conditions and in some cases has even led to potential therapeutic avenues for their prevention. In this chapter, we focus primarily on two general classes of rare disorders, ribosomopathies and ciliopathies, and the surprising finding that the disruption of fundamental, global processes can result in tissue-specific craniofacial defects. In addition, we discuss recent advances in understanding the pathogenesis of an extremely rare and specific craniofacial condition known as syngnathia, based on the first mouse models for this condition. Approximately 1% of all babies are born with a minor or major developmental anomaly, and individuals suffering from rare diseases deserve the same quality of treatment and care and attention to their disease as other patients.
Collapse
Affiliation(s)
- Annita Achilleos
- Stowers Institute for Medical Research, Kansas City, Missouri, USA
| | - Paul A Trainor
- Stowers Institute for Medical Research, Kansas City, Missouri, USA; Department of Anatomy & Cell Biology, University of Kansas Medical Center, Kansas City, Kansas, USA.
| |
Collapse
|
48
|
Green RM, Feng W, Phang T, Fish JL, Li H, Spritz RA, Marcucio RS, Hooper J, Jamniczky H, Hallgrímsson B, Williams T. Tfap2a-dependent changes in mouse facial morphology result in clefting that can be ameliorated by a reduction in Fgf8 gene dosage. Dis Model Mech 2015; 8:31-43. [PMID: 25381013 PMCID: PMC4283648 DOI: 10.1242/dmm.017616] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Accepted: 11/02/2014] [Indexed: 12/20/2022] Open
Abstract
Failure of facial prominence fusion causes cleft lip and palate (CL/P), a common human birth defect. Several potential mechanisms can be envisioned that would result in CL/P, including failure of prominence growth and/or alignment as well as a failure of fusion of the juxtaposed epithelial seams. Here, using geometric morphometrics, we analyzed facial outgrowth and shape change over time in a novel mouse model exhibiting fully penetrant bilateral CL/P. This robust model is based upon mutations in Tfap2a, the gene encoding transcription factor AP-2α, which has been implicated in both syndromic and non-syndromic human CL/P. Our findings indicate that aberrant morphology and subsequent misalignment of the facial prominences underlies the inability of the mutant prominences to fuse. Exencephaly also occured in some of the Tfap2a mutants and we observed additional morphometric differences that indicate an influence of neural tube closure defects on facial shape. Molecular analysis of the CL/P model indicates that Fgf signaling is misregulated in the face, and that reducing Fgf8 gene dosage can attenuate the clefting pathology by generating compensatory changes. Furthermore, mutations in either Tfap2a or Fgf8 increase variance in facial shape, but the combination of these mutations restores variance to normal levels. The alterations in variance provide a potential mechanistic link between clefting and the evolution and diversity of facial morphology. Overall, our findings suggest that CL/P can result from small gene-expression changes that alter the shape of the facial prominences and uncouple their coordinated morphogenesis, which is necessary for normal fusion.
Collapse
Affiliation(s)
- Rebecca M Green
- Department of Craniofacial Biology, University of Colorado Denver, 12801 East 17th Avenue, Aurora, CO 80045, USA
| | - Weiguo Feng
- Department of Craniofacial Biology, University of Colorado Denver, 12801 East 17th Avenue, Aurora, CO 80045, USA
| | - Tzulip Phang
- Department of Pharmacology, University of Colorado Denver, 12801 East 17th Avenue, Aurora, CO 80045, USA
| | - Jennifer L Fish
- University of California San Francisco, Orthopaedic Trauma Institute, Department of Orthopaedic Surgery, San Francisco, CA 94110, USA
| | - Hong Li
- Department of Craniofacial Biology, University of Colorado Denver, 12801 East 17th Avenue, Aurora, CO 80045, USA
| | - Richard A Spritz
- Human Medical Genetics and Genomics Program, University of Colorado School of Medicine, 12800 East 17th Avenue, Aurora, CO 80045, USA
| | - Ralph S Marcucio
- University of California San Francisco, Orthopaedic Trauma Institute, Department of Orthopaedic Surgery, San Francisco, CA 94110, USA
| | - Joan Hooper
- Department of Cell and Developmental Biology, University of Colorado Denver, 12801 East 17th Avenue, Aurora, CO 80045, USA
| | - Heather Jamniczky
- McCaig Institute for Bone and Joint Health, Department of Cell Biology & Anatomy, University of Calgary, 3280 Hospital Drive NW, Calgary, AB T2N3Z6, Canada
| | - Benedikt Hallgrímsson
- McCaig Institute for Bone and Joint Health, Department of Cell Biology & Anatomy, University of Calgary, 3280 Hospital Drive NW, Calgary, AB T2N3Z6, Canada. Alberta Children's Hospital Research Institute, University of Calgary, 3280 Hospital Drive NW, Calgary, AB T2N3Z6, Canada
| | - Trevor Williams
- Department of Craniofacial Biology, University of Colorado Denver, 12801 East 17th Avenue, Aurora, CO 80045, USA. Human Medical Genetics and Genomics Program, University of Colorado School of Medicine, 12800 East 17th Avenue, Aurora, CO 80045, USA. Department of Cell and Developmental Biology, University of Colorado Denver, 12801 East 17th Avenue, Aurora, CO 80045, USA.
| |
Collapse
|
49
|
Directed Bmp4 expression in neural crest cells generates a genetic model for the rare human bony syngnathia birth defect. Dev Biol 2014; 391:170-81. [PMID: 24785830 DOI: 10.1016/j.ydbio.2014.04.013] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Revised: 04/15/2014] [Accepted: 04/18/2014] [Indexed: 01/01/2023]
Abstract
Congenital bony syngnathia, a rare but severe human birth defect, is characterized by bony fusion of the mandible to the maxilla. However, the genetic mechanisms underlying this birth defect are poorly understood, largely due to limitation of available animal models. Here we present evidence that transgenic expression of Bmp4 in neural crest cells causes a series of craniofacial malformations in mice, including a bony fusion between the maxilla and hypoplastic mandible, resembling the bony syngnathia syndrome in humans. In addition, the anterior portion of the palatal shelves emerged from the mandibular arch instead of the maxilla in the mutants. Gene expression assays showed an altered expression of several facial patterning genes, including Hand2, Dlx2, Msx1, Barx1, Foxc2 and Fgf8, in the maxillary and mandibular processes of the mutants, indicating mis-patterned cranial neural crest (CNC) derived cells in the facial region. However, despite of formation of cleft palate and ectopic cartilage, forced expression of a constitutively active form of BMP receptor-Ia (caBmprIa) in CNC lineage did not produce the syngnathia phenotype, suggesting a non-cell autonomous effect of the augmented BMP4 signaling. Our studies demonstrate that aberrant BMP4-mediated signaling in CNC cells leads to mis-patterned facial skeleton and congenital bony syngnathia, and suggest an implication of mutations in BMP signaling pathway in human bony syngnathia.
Collapse
|