Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 843
Filter
Add more filters

Publication year range
1.
Orthod Craniofac Res ; 27(4): 589-597, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38409951

ABSTRACT

OBJECTIVES: This study aimed to assess the relative growth rates (RGRs) of the maxilla and mandible at varying fusion stages of the spheno-occipital synchondrosis (SOS), thereby elucidating the potential of SOS stages in predicting maxillomandibular growth. MATERIALS AND METHODS: A total of 320 subjects (171 boys and 149 girls), aged 6 to 18 years, were retrospectively included. Each subject had a minimum of two longitudinal cone-beam computed tomography (CBCT) images, with no more than one interval of SOS fusion stage change between the two scans. Subjects were categorized based on their SOS fusion stages and genders. The RGRs of the maxilla and mandible at various SOS fusion stages were measured and compared using longitudinal CBCT images. RESULTS: Significant statistical differences were observed in maxillomandibular RGRs across various SOS fusion stages. In girls, the sagittal growth of the maxilla remained stable and active until SOS 3, subsequently exhibited deceleration in SOS 4-5 (compared to SOS 3-4, P < .05) and continued to decrease in SOS 5-6. Whereas in boys, the sagittal growth of the maxilla remained stable until SOS 4, and a deceleration trend emerged starting from SOS 5 to 6 (P < .01 compared to SOS 4-5). Mandibular growth patterns in both genders exhibited a progression of increasing-accelerating-decelerating rates from SOS 2 to 6. The highest RGRs for total mandibular length were observed in SOS 3-4 and SOS 4-5. CONCLUSION: Spheno-occipital synchondrosis fusion stages can serve as a valid indicator of maxillomandibular growth maturation.


Subject(s)
Cone-Beam Computed Tomography , Mandible , Maxilla , Occipital Bone , Sphenoid Bone , Humans , Male , Female , Child , Adolescent , Cone-Beam Computed Tomography/methods , Longitudinal Studies , Mandible/diagnostic imaging , Mandible/growth & development , Occipital Bone/diagnostic imaging , Occipital Bone/growth & development , Maxilla/growth & development , Maxilla/diagnostic imaging , Retrospective Studies , Sphenoid Bone/diagnostic imaging , Sphenoid Bone/growth & development , Feasibility Studies , Maxillofacial Development/physiology , Cephalometry/methods , Sex Factors
2.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 55(2): 469-474, 2024 Mar 20.
Article in Zh | MEDLINE | ID: mdl-38645865

ABSTRACT

Craniomaxillofacial development involves a series of highly ordered temporal-spatial cellular differentiation processes in which a variety of cell signaling factors, such as fibroblast growth factors, play important regulatory roles. As a classic fibroblast growth factor, fibroblast growth factor 7 (FGF7) serves a wide range of regulatory functions. Previous studies have demonstrated that FGF7 regulates the proliferation and migration of epithelial cells, protects them, and promotes their repair. Furthermore, recent findings indicate that epithelial cells are not the only ones subjected to the broad and powerful regulatory capacity of FGF7. It has potential effects on skeletal system development as well. In addition, FGF7 plays an important role in the development of craniomaxillofacial organs, such as the palate, the eyes, and the teeth. Nonetheless, the role of FGF7 in oral craniomaxillofacial development needs to be further elucidated. In this paper, we summarized the published research on the role of FGF7 in oral craniomaxillofacial development to demonstrate the overall understanding of FGF7 and its potential functions in oral craniomaxillofacial development.


Subject(s)
Fibroblast Growth Factor 7 , Humans , Fibroblast Growth Factor 7/metabolism , Fibroblast Growth Factor 7/genetics , Animals , Skull/growth & development , Skull/metabolism , Maxillofacial Development/physiology , Tooth/metabolism , Tooth/growth & development
3.
Development ; 147(11)2020 06 11.
Article in English | MEDLINE | ID: mdl-32439763

ABSTRACT

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


Subject(s)
Blood Vessels/metabolism , Insulin-Like Growth Factor I/metabolism , Maxillofacial Development/physiology , Animals , Antigens, CD/genetics , Cadherins/deficiency , Cadherins/genetics , Cartilage/cytology , Cartilage/metabolism , Cartilage/pathology , Cell Line , Cell Proliferation , Chondrocytes/cytology , Chondrocytes/metabolism , Embryo, Mammalian/metabolism , Embryonic Development , Insulin-Like Growth Factor I/genetics , Mandible/cytology , Mandible/metabolism , Mice , Mice, Knockout , Signal Transduction , Tissue Culture Techniques , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Wnt1 Protein/deficiency , Wnt1 Protein/genetics
4.
Am J Phys Anthropol ; 174(2): 224-231, 2021 02.
Article in English | MEDLINE | ID: mdl-33098083

ABSTRACT

OBJECTIVES: Fetal environmental conditions are crucial for life-long health. Direct measurements of developmental conditions are limited in humans; thus, several biomarkers of those conditions have been proposed: that is, finger ridge-counts, level of facial fluctuating asymmetry (FA), and digit ratio (2D:4D). Since all of these biomarkers share a similar gestational time of formation, we hypothesize that their values are significantly correlated. MATERIALS AND METHODS: Data were collected at the Mogielica Human Ecology Study Site in southern Poland among 234 women. Finger ridge-counts, level of facial FA, and 2D:4D have been measured. The two-step analyses included Pearson's correlations of simple values of the biomarkers and correlations of composite variables calculated based on principal component analysis. RESULTS: We did not find any statistically significant correlations between finger ridge-counts, FA, and 2D:4D in women. Similarly, we did not observe any correlations between three composites created from the biomarkers. DISCUSSION: Our results indicate that there are no relationships between the biomarkers, suggested as proxies of the quality of prenatal conditions, in a single population. This is the first study analyzing three different markers simultaneously. The lack of correlations may indicate that the tested biomarkers reflect, in fact, different environmental conditions, occurring in separate "critical windows" of development, or that the biomarkers are not valid as proxies of developmental conditions.


Subject(s)
Facial Asymmetry/pathology , Fetal Development/physiology , Fetus/pathology , Fingers/anatomy & histology , Adult , Anthropology, Physical , Anthropometry , Biomarkers , Female , Fetus/anatomy & histology , Fingers/growth & development , Humans , Maxillofacial Development/physiology , Poland
5.
PLoS Genet ; 14(10): e1007675, 2018 10.
Article in English | MEDLINE | ID: mdl-30286078

ABSTRACT

The mechanisms that regulate post-natal growth of the craniofacial complex and that ultimately determine the size and shape of our faces are not well understood. Hippo signaling is a general mechanism to control tissue growth and organ size, and although it is known that Hippo signaling functions in neural crest specification and patterning during embryogenesis and before birth, its specific role in postnatal craniofacial growth remains elusive. We have identified the transcription factor FoxO6 as an activator of Hippo signaling regulating neonatal growth of the face. During late stages of mouse development, FoxO6 is expressed specifically in craniofacial tissues and FoxO6-/- mice undergo expansion of the face, frontal cortex, olfactory component and skull. Enlargement of the mandible and maxilla and lengthening of the incisors in FoxO6-/- mice are associated with increases in cell proliferation. In vitro and in vivo studies demonstrated that FoxO6 activates Lats1 expression, thereby increasing Yap phosphorylation and activation of Hippo signaling. FoxO6-/- mice have significantly reduced Hippo Signaling caused by a decrease in Lats1 expression and decreases in Shh and Runx2 expression, suggesting that Shh and Runx2 are also linked to Hippo signaling. In vitro, FoxO6 activates Hippo reporter constructs and regulates cell proliferation. Furthermore PITX2, a regulator of Hippo signaling is associated with Axenfeld-Rieger Syndrome causing a flattened midface and we show that PITX2 activates FoxO6 expression. Craniofacial specific expression of FoxO6 postnatally regulates Hippo signaling and cell proliferation. Together, these results identify a FoxO6-Hippo regulatory pathway that controls skull growth, odontogenesis and face morphology.


Subject(s)
Forkhead Transcription Factors/metabolism , Maxillofacial Development/physiology , Protein Serine-Threonine Kinases/metabolism , Skull/growth & development , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Differentiation/physiology , Cell Proliferation/physiology , Hippo Signaling Pathway , Homeodomain Proteins/metabolism , Maxillofacial Development/genetics , Mice , Neural Crest/cytology , Organ Size , Phosphorylation , Signal Transduction , Skull/metabolism , Transcription Factors/metabolism , Homeobox Protein PITX2
6.
Proc Natl Acad Sci U S A ; 114(35): 9403-9408, 2017 08 29.
Article in English | MEDLINE | ID: mdl-28808032

ABSTRACT

Much of the basic information about individual organ development comes from studies using model species. Whereas conservation of gene regulatory networks across higher taxa supports generalizations made from a limited number of species, generality of mechanistic inferences remains to be tested in tissue culture systems. Here, using mammalian tooth explants cultured in isolation, we investigate self-regulation of patterning by comparing developing molars of the mouse, the model species of mammalian research, and the bank vole. A distinct patterning difference between the vole and the mouse molars is the alternate cusp offset present in the vole. Analyses of both species using 3D reconstructions of developing molars and jaws, computational modeling of cusp patterning, and tooth explants cultured with small braces show that correct cusp offset requires constraints on the lateral expansion of the developing tooth. Vole molars cultured without the braces lose their cusp offset, and mouse molars cultured with the braces develop a cusp offset. Our results suggest that cusp offset, which changes frequently in mammalian evolution, is more dependent on the 3D support of the developing jaw than other aspects of tooth shape. This jaw-tooth integration of a specific aspect of the tooth phenotype indicates that organs may outsource specific aspects of their morphology to be regulated by adjacent body parts or organs. Comparative studies of morphologically different species are needed to infer the principles of organogenesis.


Subject(s)
Biological Evolution , Jaw , Maxillofacial Development/physiology , Tooth/anatomy & histology , Animals , Arvicolinae/embryology , Biomechanical Phenomena , Computer Simulation , Embryo, Mammalian , Embryonic Development , Mice , Models, Biological
7.
Dev Biol ; 440(2): 53-63, 2018 08 15.
Article in English | MEDLINE | ID: mdl-29792855

ABSTRACT

The Hippo signaling pathway is implicated in key aspects of cell proliferation, control of organ size, stem cell functions and tumor suppression. Its functions are primarily mediated either through direct effects on transcription factors to influence target gene expression or through crosstalk with other signaling pathways that regulate multiple physiological activities. Studies are revealing Hippo pathway involvement in diverse functions including renewal of intestinal epithelium, promotion of myocardial cell proliferation, cancer suppression, etc. In this review we discuss Hippo pathway signaling in oral-maxillofacial development and bone remodeling under normal and pathological conditions and highlight promising future research directions.


Subject(s)
Bone Remodeling/physiology , Maxillofacial Development/physiology , Protein Serine-Threonine Kinases/physiology , Animals , Cell Proliferation , Drosophila Proteins/metabolism , Drosophila Proteins/physiology , Hippo Signaling Pathway , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/physiology , Mice , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/physiology , Stem Cells/metabolism , Transcription Factors/metabolism
8.
Evol Dev ; 21(5): 231-246, 2019 09.
Article in English | MEDLINE | ID: mdl-31374588

ABSTRACT

Protrusile jaws are a highly useful innovation that has been linked to extensive diversification in fish feeding ecology. Jaw protrusion can enhance the performance of multiple functions, such as suction production and capturing elusive prey. Identifying the developmental factors that alter protrusion ability will improve our understanding of fish diversification. In the zebrafish protrusion arises postmetamorphosis. Fish metamorphosis typically includes significant changes in trophic morphology, accompanies a shift in feeding niche and coincides with increased thyroid hormone production. We tested whether thyroid hormone affects the development of zebrafish feeding mechanics. We found that it affected all developmental stages examined, but that effects were most pronounced after metamorphosis. Thyroid hormone levels affected the development of jaw morphology, feeding mechanics, shape variation, and cranial ossification. Adult zebrafish utilize protrusile jaws, but an absence of thyroid hormone impaired development of the premaxillary bone, which is critical to jaw protrusion. Premaxillae from early juvenile zebrafish and hypothyroid adult zebrafish resemble those from adults in the genera Danionella, Devario, and Microdevario that show little to no jaw protrusion. Our findings suggest that evolutionary changes in how the developing skulls of danionin minnows respond to thyroid hormone may have promoted diversification into different feeding niches.


Subject(s)
Jaw/physiology , Thyroid Hormones/metabolism , Zebrafish/physiology , Animals , Biological Evolution , Biomechanical Phenomena , Feeding Behavior , Maxillofacial Development/physiology , Zebrafish/growth & development
9.
J Anat ; 234(1): 89-105, 2019 01.
Article in English | MEDLINE | ID: mdl-30456781

ABSTRACT

Understanding how skeleton changes shape in ontogeny is fundamental to understanding how its shape diversifies in phylogeny. Amphibians pose a special case because their jaw and throat skeleton consists of cartilages that are dramatically reshaped midway through life to support new feeding and breathing styles. Although amphibian metamorphosis is commonly studied by immersing larvae in thyroid hormones (TH), how individual cartilages respond to TH is poorly understood. This study documents the effects of larval stage and TH type (T4 vs. T3), dose and deprivation on the size, shape and morphogenesis of the lower jaw and ceratohyal cartilages in the frog Xenopus laevis. It uses thyroid inhibitors to isolate the effects of each hormone at specific concentrations. It also deconstructs the TH responses into the effects on individual dimensions, and uses measures of percent change to eliminate the effects of body size and growth rate variation. As stage increases, T4 and T3 responses become increasingly similar to each other and to natural remodeling; the differences at low and intermediate stages result largely from abnormal responses to T3. Most notably, the beak-like lower jaw commonly observed at the lowest stage in other studies results largely from arrested growth of cartilage. TH responses are superimposed upon the growth typical for each stage so that cartilages can attain postmetamorphic shapes through dimensional changes that exceed those of natural metamorphosis. Using thyroid inhibitors alters the outcome of TH-induced remodeling, and T4 has almost the same capacity to induce metamorphic shape changes as T3. The results have implications for understanding how the starting shapes of larval elements affect morphogenesis, how chondrocytes behave to change cartilage shape, and how intracellular processing of TH might contribute to interspecific differences in shape change. Also, the data on animal mortality and which stages and doses most closely replicate natural remodeling have practical value for researchers who treat Xenopus tadpoles with TH.


Subject(s)
Cartilage/anatomy & histology , Cartilage/growth & development , Maxillofacial Development/physiology , Morphogenesis/physiology , Thyroxine/physiology , Triiodothyronine/physiology , Animals , Jaw/anatomy & histology , Jaw/physiology , Thyroid Hormones/physiology , Xenopus laevis
10.
Dev Biol ; 426(1): 97-114, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28363736

ABSTRACT

The rapid increase in gene-centric biological knowledge coupled with analytic approaches for genomewide data integration provides an opportunity to develop systems-level understanding of facial development. Experimental analyses have demonstrated the importance of signaling between the surface ectoderm and the underlying mesenchyme are coordinating facial patterning. However, current transcriptome data from the developing vertebrate face is dominated by the mesenchymal component, and the contributions of the ectoderm are not easily identified. We have generated transcriptome datasets from critical periods of mouse face formation that enable gene expression to be analyzed with respect to time, prominence, and tissue layer. Notably, by separating the ectoderm and mesenchyme we considerably improved the sensitivity compared to data obtained from whole prominences, with more genes detected over a wider dynamic range. From these data we generated a detailed description of ectoderm-specific developmental programs, including pan-ectodermal programs, prominence- specific programs and their temporal dynamics. The genes and pathways represented in these programs provide mechanistic insights into several aspects of ectodermal development. We also used these data to identify co-expression modules specific to facial development. We then used 14 co-expression modules enriched for genes involved in orofacial clefts to make specific mechanistic predictions about genes involved in tongue specification, in nasal process patterning and in jaw development. Our multidimensional gene expression dataset is a unique resource for systems analysis of the developing face; our co-expression modules are a resource for predicting functions of poorly annotated genes, or for predicting roles for genes that have yet to be studied in the context of facial development; and our analytic approaches provide a paradigm for analysis of other complex developmental programs.


Subject(s)
Ectoderm/embryology , Face/embryology , Gene Expression Regulation, Developmental/genetics , Maxillofacial Development/physiology , Mesoderm/embryology , Systems Biology , Animals , Jaw/embryology , Mice , Mice, Inbred C57BL , Nose/embryology , Tongue/embryology
11.
J Oral Maxillofac Surg ; 76(11): 2388-2397, 2018 11.
Article in English | MEDLINE | ID: mdl-29679588

ABSTRACT

PURPOSE: Submucous cleft palate (SMCP) is a particular subtype of cleft palate deformity; research related to the craniofacial features of patients with SMCP is comparatively rare. The study objective was to perform a cephalometric comparison of the craniofacial features of patients with SMCP and non-cleft controls at different ages. MATERIALS AND METHODS: The sample in this cross-sectional study was composed of 2 groups: SMCP patients and non-cleft controls. The primary predictor variables were study group (cleft and non-cleft) and age. Age was divided into 3 groups. The outcome variables of interest were craniofacial measurements. The measurements used reflect cranial length, cranial angle, maxillary sagittal length and protrusion, maxillary vertical height, pharyngeal depth, facial height, mandibular length and protrusion, mandibular plane angle, and intermaxillary relation. Adjusted cephalometric craniofacial measurements between the groups were compared in 3 age groups using generalized linear models after being adjusted for age and gender. RESULTS: The study included 60 SMCP patients and 60 non-cleft controls. SMCP patients and non-cleft controls were divided into 3 subgroups: those aged 5 to 7 years, those aged 9 to 11 years, and those aged 18 to 30 years. Patients with SMCP at age 5 to 7 years showed a shortened cranial base length, maxillary sagittal length and height, and bony pharynx depth. Patients with SMCP at age 9 to 11 years showed a smaller maxillary sagittal length and bony pharynx depth and an inharmonious jaw relationship. Patients with SMCP at age 18 to 30 years showed a smaller maxillary sagittal length and height and an inharmonious jaw relationship. CONCLUSIONS: SMCP is associated with progressive maxillary retrognathism and reduced profile convexity from childhood to adulthood.


Subject(s)
Cleft Palate/physiopathology , Maxilla/growth & development , Maxillofacial Development/physiology , Pharynx/growth & development , Retrognathia/physiopathology , Skull Base/growth & development , Adolescent , Adult , Age Factors , Cephalometry , Child , Child, Preschool , Cleft Palate/diagnostic imaging , Cross-Sectional Studies , Female , Humans , Male , Maxilla/abnormalities , Maxilla/diagnostic imaging , Pharynx/abnormalities , Pharynx/diagnostic imaging , Retrognathia/diagnostic imaging , Skull Base/abnormalities , Skull Base/diagnostic imaging
12.
J Craniofac Surg ; 29(6): 1495-1500, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30015744

ABSTRACT

The aim of this study was to investigate the effect of orthodontic treatment on the maxillofacial growth of patients with unilateral cleft lip and palate. The Great Ormond Street, London and Oslo (GOSLON) yardstick was used for a longitudinal evaluation of 24 patients with cleft lip and palate treated at the Cleft Center of the Lauro Wanderley University Hospital, Paraiba State, northeastern Brazil. Dental casts were evaluated by 3 orthodontists and classified according to the GOSLON yardstick. The evaluation was performed at 2 different stages: T1 (before orthodontic treatment) and T2 (follow-up evaluation) after a 6-year mean follow-up interval. The Kappa test was used to evaluate intra- and interexaminer agreement, and paired t-test was used to compare the differences between T1 and T2, with a 99% confidence interval. The average intraexaminer Kappa was 0.979, ranging from 0.971 to 0.990. The interexaminer Kappa value was 0.926 at T1, ranging from 0.885 to 0.964, and 0.896 at T2, ranging from 0.696 to 1.0. The mean GOSLON yardstick found at T1 was 2.5 ±â€Š1.18 with 50% in G1 + G2, 29.18% in G3, and 20.82% in G4 + G5. At T2, the GOSLON average was 1.71 ±â€Š1.12, with 79.18% in G1 + G2, 12.5% in G3, and 8.32% in G4 + G5. A statistically significant difference was found between T1 and T2. The results suggest that orthodontic treatment improves facial growth in patients with unilateral cleft lip and palate.


Subject(s)
Cleft Lip , Cleft Palate , Maxillofacial Development/physiology , Orthodontics, Corrective , Brazil , Cleft Lip/pathology , Cleft Lip/therapy , Cleft Palate/pathology , Cleft Palate/therapy , Face/anatomy & histology , Face/pathology , Humans , Longitudinal Studies
13.
J Oral Rehabil ; 45(3): 228-234, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29230834

ABSTRACT

Oro-facial dysfunctions (OFD) or oro-facial myofunctional disorders in children lead to severe problems in teeth and jaw position, articulation, chewing and swallowing. The forces of the tongue, the central muscle for articulation, chewing and swallowing are focused on in several studies. In this examination, isometric tongue protrusion forces (TPF) of children with OFD and controls were compared. Thirty participants with OFD and 30 controls were presented a target force level as a straight line on a monitor that they were supposed to match by generating an isometric tongue force for different target levels (0.25 N and 0.5 N). Correlations of the severity of OFD (symptom score) with the capacities of the TPF 0.25 N and 0.5 N were calculated. Statistical differences were obvious in TPF variability and the accuracy, depending on the weight. Tongue contact time, expressed as per cent (TCT, total contact: 100%), was significantly lower in children with OFD (P = .005). Mean and median TPF was not different between groups. The predictive value of TPF for OFD revealed a level of 58.6% for TPF 0.25 N and 74.5% for TPF 0.5 N. Correlations of the severity of OFD were seen for some parameters. Subjects with OFD show significantly lower competencies in accuracy and endurance of tongue protrusion forces. This may have a high impact on phenotyping children with OFD and influence therapeutical approaches.


Subject(s)
Articulation Disorders/physiopathology , Chronic Disease , Deglutition Disorders/physiopathology , Facial Muscles/physiopathology , Hypoglossal Nerve/physiopathology , Maxillofacial Development/physiology , Tongue/physiopathology , Adolescent , Articulation Disorders/diagnosis , Child , Deglutition/physiology , Deglutition Disorders/diagnosis , Disability Evaluation , Disease Progression , Electromyography , Evaluation Studies as Topic , Female , Humans , Male , Mastication/physiology , Patient Compliance/statistics & numerical data , Phenotype , Predictive Value of Tests , Reproducibility of Results , Severity of Illness Index
14.
Dev Biol ; 415(2): 278-295, 2016 07 15.
Article in English | MEDLINE | ID: mdl-26746790

ABSTRACT

The cranial base is a component of the neurocranium and has a central role in the structural integration of the face, brain and vertebral column. Consequently, alteration in the shape of the human cranial base has been intimately linked with primate evolution and defective development is associated with numerous human facial abnormalities. Here we describe a novel recessive mutant mouse strain that presented with a domed head and fully penetrant cleft secondary palate coupled with defects in the formation of the underlying cranial base. Mapping and non-complementation studies revealed a specific mutation in Memo1 - a gene originally associated with cell migration. Expression analysis of Memo1 identified robust expression in the perichondrium and periosteum of the developing cranial base, but only modest expression in the palatal shelves. Fittingly, although the palatal shelves failed to elevate in Memo1 mutants, expression changes were modest within the shelves themselves. In contrast, the cranial base, which forms via endochondral ossification had major reductions in the expression of genes responsible for bone formation, notably matrix metalloproteinases and markers of the osteoblast lineage, mirrored by an increase in markers of cartilage and extracellular matrix development. Concomitant with these changes, mutant cranial bases showed an increased zone of hypertrophic chondrocytes accompanied by a reduction in both vascular invasion and mineralization. Finally, neural crest cell-specific deletion of Memo1 caused a failure of anterior cranial base ossification indicating a cell autonomous role for MEMO1 in the development of these neural crest cell derived structures. However, palate formation was largely normal in these conditional mutants, suggesting a non-autonomous role for MEMO1 in palatal closure. Overall, these findings assign a new function to MEMO1 in driving endochondral ossification in the cranium, and also link abnormal development of the cranial base with more widespread effects on craniofacial shape relevant to human craniofacial dysmorphology.


Subject(s)
Cleft Palate/genetics , Intracellular Signaling Peptides and Proteins/physiology , Maxillofacial Development/physiology , Osteogenesis/physiology , Palate/embryology , Skull Base/embryology , Animals , Cartilage/embryology , Cartilage/pathology , Cleft Palate/embryology , Ethylnitrosourea , Exons , Gene Expression Regulation, Developmental , Genes, Recessive , Humans , Male , Mesoderm/cytology , Mesoderm/embryology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mutagenesis , Neural Crest/cytology , Neural Crest/embryology , Palate/metabolism , Palate/pathology , Point Mutation , Skull Base/metabolism , Skull Base/pathology , Species Specificity
15.
Dev Biol ; 415(2): 338-346, 2016 07 15.
Article in English | MEDLINE | ID: mdl-26719128

ABSTRACT

We have made great strides towards understanding the etiology of craniofacial disorders, especially for 'simple' Mendelian traits. However, the facial skeleton is a complex trait, and the full spectrum of genetic, developmental, and environmental factors that contribute to its final geometry remain unresolved. Forward genetic screens are constrained with respect to complex traits due to the types of genes and alleles commonly identified, developmental pleiotropy, and limited information about the impact of environmental interactions. Here, we discuss how studies in an evolutionary model - African cichlid fishes - can complement traditional approaches to understand the genetic and developmental origins of complex shape. Cichlids exhibit an unparalleled range of natural craniofacial morphologies that model normal human variation, and in certain instances mimic human facial dysmorphologies. Moreover, the evolutionary history and genomic architecture of cichlids make them an ideal system to identify the genetic basis of these phenotypes via quantitative trait loci (QTL) mapping and population genomics. Given the molecular conservation of developmental genes and pathways, insights from cichlids are applicable to human facial variation and disease. We review recent work in this system, which has identified lbh as a novel regulator of neural crest cell migration, determined the Wnt and Hedgehog pathways mediate species-specific bone morphologies, and examined how plastic responses to diet modulate adult facial shapes. These studies have not only revealed new roles for existing pathways in craniofacial development, but have identified new genes and mechanisms involved in shaping the craniofacial skeleton. In all, we suggest that combining work in traditional laboratory and evolutionary models offers significant potential to provide a more complete and comprehensive picture of the myriad factors that are involved in the development of complex traits.


Subject(s)
Cichlids/embryology , Craniofacial Abnormalities/embryology , Disease Models, Animal , Fish Diseases/embryology , Gene-Environment Interaction , Head/anatomy & histology , Maxillofacial Development/physiology , Animals , Biological Evolution , Cichlids/anatomy & histology , Cichlids/genetics , Craniofacial Abnormalities/genetics , Craniofacial Abnormalities/veterinary , Embryo, Nonmammalian/pathology , Embryonic Development , Feeding Behavior , Gene Expression Regulation, Developmental , Genetic Pleiotropy , Head/embryology , Humans , Maxillofacial Development/genetics , Neural Crest/embryology , Phenotype , Quantitative Trait Loci , Signal Transduction , Species Specificity , Trans-Activators/genetics
16.
Dev Biol ; 415(2): 306-313, 2016 07 15.
Article in English | MEDLINE | ID: mdl-26116174

ABSTRACT

Cleft palate is among the most common human birth defects. Submucous cleft palate (SMCP) is a subgroup of cleft palate, which may be as common as overt cleft palate. Despite the high frequency of SMCP in humans, only recently have several animal models of SMCP begun to provide insight into the mechanisms by which SMCP develops. In this study, we show that enhanced BMP signaling through constitutively active ACVR1 in palatal epithelium causes submucous cleft palate in mice. In these mutant mice, the fusion of both palatal mesenchyme in hard palate, and muscles in soft palate were hampered by epithelial tissue. During palatal fusion, enhanced SMAD-dependent BMP signaling impaired cell death and altered cell proliferation rate in medial edge epithelium (MEE), and resulted in MEE persistence. At the molecular level, downregulation of ΔNp63, which is crucial for normal palatal fusion, in MEE cells was impaired, leading to a reduction in caspase-3 activation. Our study provides a new insight into the etiology of SMCP caused by augmented BMP signaling.


Subject(s)
Activin Receptors, Type I/genetics , Bone Morphogenetic Proteins/physiology , Cleft Palate/genetics , Epithelium/embryology , Maxillofacial Development/physiology , Mouth Mucosa/embryology , Activin Receptors, Type I/physiology , Animals , Apoptosis , Caspase 3/physiology , Cleft Palate/embryology , Cleft Palate/metabolism , Enzyme Activation , Epithelium/pathology , Luminescent Proteins/analysis , Luminescent Proteins/genetics , Mesoderm/embryology , Mice , Mouth Mucosa/pathology , Mutation , Organ Culture Techniques , Phosphoproteins/biosynthesis , Phosphoproteins/genetics , Signal Transduction , Smad Proteins/physiology , Trans-Activators/biosynthesis , Trans-Activators/genetics , Up-Regulation
17.
Dev Biol ; 415(2): 188-197, 2016 07 15.
Article in English | MEDLINE | ID: mdl-26724698

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.


Subject(s)
Craniofacial Abnormalities/genetics , Genetic Association Studies , Maxillofacial Development/physiology , Animals , Biological Evolution , Evolution, Molecular , Face/embryology , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Genetic Variation , Head/embryology , Humans , Matrix Attachment Region Binding Proteins/genetics , Matrix Attachment Region Binding Proteins/physiology , Maxillofacial Development/genetics , Mesoderm/cytology , Mesoderm/embryology , Morphogenesis , Mutation , Neural Crest/cytology , Neural Crest/embryology , Skull/embryology , Transcription Factors/genetics , Transcription Factors/physiology , Vertebrates/embryology , Vertebrates/genetics
18.
Dev Biol ; 415(2): 296-305, 2016 07 15.
Article in English | MEDLINE | ID: mdl-26772999

ABSTRACT

The palate functions as the roof of the mouth in mammals, separating the oral and nasal cavities. Its complex embryonic development and assembly poses unique susceptibilities to intrinsic and extrinsic disruptions. Such disruptions may cause failure of the developing palatal shelves to fuse along the midline resulting in a cleft. In other cases the palate may fuse at an arch, resulting in a vaulted oral cavity, termed high-arched palate. There are many models available for studying the pathogenesis of cleft palate but a relative paucity for high-arched palate. One condition exhibiting either cleft palate or high-arched palate is Treacher Collins syndrome, a congenital disorder characterized by numerous craniofacial anomalies. We quantitatively analyzed palatal perturbations in the Tcof1(+/-) mouse model of Treacher Collins syndrome, which phenocopies the condition in humans. We discovered that 46% of Tcof1(+/-) mutant embryos and new born pups exhibit either soft clefts or full clefts. In addition, 17% of Tcof1(+/-) mutants were found to exhibit high-arched palate, defined as two sigma above the corresponding wild-type population mean for height and angular based arch measurements. Furthermore, palatal shelf length and shelf width were decreased in all Tcof1(+/-) mutant embryos and pups compared to controls. Interestingly, these phenotypes were subsequently ameliorated through genetic inhibition of p53. The results of our study therefore provide a simple, reproducible and quantitative method for investigating models of high-arched palate.


Subject(s)
Maxillofacial Development/physiology , Nuclear Proteins/genetics , Palate/abnormalities , Phosphoproteins/genetics , Animals , Cleft Palate/diagnostic imaging , Cleft Palate/embryology , Cleft Palate/genetics , Crosses, Genetic , Disease Models, Animal , Female , Gene Knockout Techniques , Genes, p53 , Heterozygote , Humans , Imaging, Three-Dimensional , Intracellular Signaling Peptides and Proteins , Male , Mandibulofacial Dysostosis/diagnostic imaging , Mandibulofacial Dysostosis/embryology , Mandibulofacial Dysostosis/genetics , Maxillofacial Development/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Microscopy, Confocal , Nuclear Proteins/physiology , Palate/diagnostic imaging , Palate/embryology , Phenotype , Phosphoproteins/physiology , Species Specificity
19.
Dev Biol ; 415(2): 198-215, 2016 07 15.
Article in English | MEDLINE | ID: mdl-26875496

ABSTRACT

The Hedgehog signalling pathway plays a fundamental role in orchestrating normal craniofacial development in vertebrates. In particular, Sonic hedgehog (Shh) is produced in three key domains during the early formation of the head; neuroectoderm of the ventral forebrain, facial ectoderm and the pharyngeal endoderm; with signal transduction evident in both ectodermal and mesenchymal tissue compartments. Shh signalling from the prechordal plate and ventral midline of the diencephalon is required for appropriate division of the eyefield and forebrain, with mutation in a number of pathway components associated with Holoprosencephaly, a clinically heterogeneous developmental defect characterized by a failure of the early forebrain vesicle to divide into distinct halves. In addition, signalling from the pharyngeal endoderm and facial ectoderm plays an essential role during development of the face, influencing cranial neural crest cells that migrate into the early facial processes. In recent years, the complexity of Shh signalling has been highlighted by the identification of multiple novel proteins that are involved in regulating both the release and reception of this protein. Here, we review the contributions of Shh signalling during early craniofacial development, focusing on Hedgehog receptor function and describing the consequences of disruption for inherited anomalies of this region in both mouse models and human populations.


Subject(s)
Craniofacial Abnormalities/embryology , Hedgehog Proteins/physiology , Maxillofacial Development/physiology , Patched Receptors/physiology , Signal Transduction , Animals , Cell Movement , Cilia/physiology , Ciliopathies/embryology , Ciliopathies/genetics , Ciliopathies/physiopathology , Craniofacial Abnormalities/genetics , Craniofacial Abnormalities/physiopathology , Diencephalon/embryology , Disease Models, Animal , Ectoderm/embryology , Endoderm/embryology , Face/abnormalities , Face/embryology , Gene Expression Regulation, Developmental , Holoprosencephaly/embryology , Holoprosencephaly/genetics , Holoprosencephaly/physiopathology , Humans , Maxillofacial Development/genetics , Membrane Proteins/physiology , Neural Crest/cytology , Neural Crest/embryology , Patched Receptors/genetics , Signal Transduction/genetics , Skull/abnormalities , Skull/embryology
20.
Dev Biol ; 415(2): 314-325, 2016 07 15.
Article in English | MEDLINE | ID: mdl-26777098

ABSTRACT

Prenatal exposure to ethanol results in fetal alcohol spectrum disorder (FASD), a syndrome characterised by a broad range of clinical manifestations including craniofacial dysmorphologies and neurological defects. The characterisation of the mechanisms by which ethanol exerts its teratogenic effects is difficult due to the pleiotropic nature of its actions. Different experimental model systems have been employed to investigate the aetiology of FASD. Here, I will review studies using these different model organisms that have helped to elucidate how ethanol causes the craniofacial abnormalities characteristic of FASD. In these studies, ethanol was found to impair the prechordal plate-an important embryonic signalling centre-during gastrulation and to negatively affect the induction, migration and survival of the neural crest, a cell population that generates the cartilage and most of the bones of the skull. At the cellular level, ethanol appears to inhibit Sonic hedgehog signalling, alter levels of retionoic acid activity, trigger a Ca(2+)-CamKII-dependent pathway that antagonises WNT signalling, affect cytoskeletal dynamics and increase oxidative stress. Embryos of the domestic chick Gallus gallus domesticus have played a central role in developing a working model for the effects of ethanol on craniofacial development because they are easily accessible and because key steps in craniofacial development are particularly well established in the avian embryo. I will finish this review by highlighting some potential future avenues of fetal alcohol research.


Subject(s)
Abnormalities, Drug-Induced/embryology , Chick Embryo/drug effects , Craniofacial Abnormalities/chemically induced , Disease Models, Animal , Ethanol/toxicity , Face/embryology , Fetal Alcohol Spectrum Disorders/physiopathology , Maxillofacial Development/drug effects , Skull/embryology , Animals , Calcium Signaling/drug effects , Craniofacial Abnormalities/embryology , Embryo, Mammalian/drug effects , Embryo, Nonmammalian/drug effects , Endoderm/drug effects , Face/abnormalities , Fetal Alcohol Spectrum Disorders/pathology , Gastrula/drug effects , Genetic Predisposition to Disease , Hedgehog Proteins/physiology , Holoprosencephaly/chemically induced , Holoprosencephaly/embryology , Humans , Maxillofacial Development/physiology , Neural Crest/drug effects , Neural Crest/pathology , Signal Transduction/drug effects , Skull/abnormalities , Species Specificity , Tretinoin/physiology , Tretinoin/toxicity , Wnt Signaling Pathway/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL