Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 652
Filter
1.
Early Hum Dev ; 193: 106021, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38701668

ABSTRACT

OBJECTIVE: Fetal face measurements in prenatal ultrasound can aid in identifying craniofacial abnormalities in the developing fetus. However, the accuracy and reliability of ultrasound measurements can be affected by factors such as fetal position, image quality, and the sonographer's expertise. This study assesses the accuracy and reliability of fetal facial measurements in prenatal ultrasound. Additionally, the temporal evolution of measurements is studied, comparing prenatal and postnatal measurements. METHODS: Three different experts located up to 23 facial landmarks in 49 prenatal 3D ultrasound scans from normal Caucasian fetuses at weeks 20, 26, and 35 of gestation. Intra- and inter-observer variability was obtained. Postnatal facial measurements were also obtained at 15 days and 1 month postpartum. RESULTS: Most facial landmarks exhibited low errors, with overall intra- and inter-observer errors of 1.01 mm and 1.60 mm, respectively. Landmarks on the nose were found to be the most reliable, while the most challenging ones were those located on the ears and eyes. Overall, scans obtained at 26 weeks of gestation presented the best trade-off between observer variability and landmark visibility. The temporal evolution of the measurements revealed that the lower face area had the highest rate of growth throughout the latest stages of pregnancy. CONCLUSIONS: Craniofacial landmarks can be evaluated using 3D fetal ultrasound, especially those located on the nose, mouth, and chin. Despite its limitations, this study provides valuable insights into prenatal and postnatal biometric changes over time, which could aid in developing predictive models for postnatal measurements based on prenatal data.


Subject(s)
Face , Ultrasonography, Prenatal , Humans , Female , Ultrasonography, Prenatal/methods , Ultrasonography, Prenatal/standards , Face/diagnostic imaging , Face/embryology , Face/anatomy & histology , Pregnancy , Imaging, Three-Dimensional/methods , Longitudinal Studies , Observer Variation , Reproducibility of Results , Adult
2.
Curr Top Dev Biol ; 157: 83-123, 2024.
Article in English | MEDLINE | ID: mdl-38556460

ABSTRACT

For almost a century, developmental biologists have appreciated that the ability of the embryonic organizer to induce and pattern the body plan is intertwined with its differentiation into axial mesoderm. Despite this, we still have a relatively poor understanding of the contribution of axial mesoderm to induction and patterning of different body regions, and the manner in which axial mesoderm-derived information is interpreted in tissues of changing competence. Here, with a particular focus on the nervous system, we review the evidence that axial mesoderm notochord and prechordal mesoderm/mesendoderm act as organizers, discuss how their influence extends through the different axes of the developing organism, and describe how the ability of axial mesoderm to direct morphogenesis impacts on its role as a local organizer.


Subject(s)
Brain/embryology , Face/embryology , Germ Layers , Mesoderm , Nervous System , Mesoderm/physiology , Morphogenesis , Body Patterning
3.
Dis Model Mech ; 17(6)2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38511331

ABSTRACT

CDK13-related disorder, also known as congenital heart defects, dysmorphic facial features and intellectual developmental disorder (CHDFIDD) is associated with mutations in the CDK13 gene encoding transcription-regulating cyclin-dependent kinase 13 (CDK13). Here, we focused on the development of craniofacial structures and analyzed early embryonic stages in CHDFIDD mouse models, with one model comprising a hypomorphic mutation in Cdk13 and exhibiting cleft lip/palate, and another model comprising knockout of Cdk13, featuring a stronger phenotype including midfacial cleft. Cdk13 was found to be physiologically expressed at high levels in the mouse embryonic craniofacial structures, namely in the forebrain, nasal epithelium and maxillary mesenchyme. We also uncovered that Cdk13 deficiency leads to development of hypoplastic branches of the trigeminal nerve including the maxillary branch. Additionally, we detected significant changes in the expression levels of genes involved in neurogenesis (Ache, Dcx, Mef2c, Neurog1, Ntn1, Pou4f1) within the developing palatal shelves. These results, together with changes in the expression pattern of other key face-specific genes (Fgf8, Foxd1, Msx1, Meis2 and Shh) at early stages in Cdk13 mutant embryos, demonstrate a key role of CDK13 in the regulation of craniofacial morphogenesis.


Subject(s)
Disease Models, Animal , Embryonic Development , Gene Expression Regulation, Developmental , Neurogenesis , Animals , Neurogenesis/genetics , Embryonic Development/genetics , Cyclin-Dependent Kinases/metabolism , Cyclin-Dependent Kinases/genetics , Skull/embryology , Skull/pathology , Mice , Cleft Palate/genetics , Cleft Palate/pathology , Cleft Palate/embryology , Cleft Lip/genetics , Cleft Lip/pathology , Cleft Lip/embryology , Trigeminal Nerve/embryology , Embryo, Mammalian/metabolism , Face/embryology , Face/abnormalities , Phenotype , Intellectual Disability/genetics , Mutation/genetics , Doublecortin Protein
4.
Development ; 148(17)2021 09 01.
Article in English | MEDLINE | ID: mdl-34383890

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.


Subject(s)
Body Patterning , Face/embryology , GATA3 Transcription Factor/metabolism , Animals , Branchial Region/cytology , Branchial Region/embryology , Branchial Region/metabolism , Cell Death , Cell Proliferation , Craniofacial Abnormalities/embryology , Craniofacial Abnormalities/genetics , Craniofacial Abnormalities/metabolism , Embryo, Mammalian , GATA3 Transcription Factor/genetics , Gene Expression Regulation, Developmental , Mandible/cytology , Mandible/embryology , Maxilla/cytology , Maxilla/embryology , Mice , Morphogenesis , Neural Crest/cytology , Neural Crest/embryology , Neural Crest/metabolism
5.
PLoS Genet ; 17(5): e1009528, 2021 05.
Article in English | MEDLINE | ID: mdl-33983923

ABSTRACT

The analysis of contemporary genomic data typically operates on one-dimensional phenotypic measurements (e.g. standing height). Here we report on a data-driven, family-informed strategy to facial phenotyping that searches for biologically relevant traits and reduces multivariate 3D facial shape variability into amendable univariate measurements, while preserving its structurally complex nature. We performed a biometric identification of siblings in a sample of 424 children, defining 1,048 sib-shared facial traits. Subsequent quantification and analyses in an independent European cohort (n = 8,246) demonstrated significant heritability for a subset of traits (0.17-0.53) and highlighted 218 genome-wide significant loci (38 also study-wide) associated with facial variation shared by siblings. These loci showed preferential enrichment for active chromatin marks in cranial neural crest cells and embryonic craniofacial tissues and several regions harbor putative craniofacial genes, thereby enhancing our knowledge on the genetic architecture of normal-range facial variation.


Subject(s)
Biometric Identification , Face/anatomy & histology , Genomics , Imaging, Three-Dimensional , Multifactorial Inheritance/genetics , Phenotype , Siblings , Adolescent , Child , Child, Preschool , Craniofacial Abnormalities/genetics , Datasets as Topic , Europe/ethnology , Face/abnormalities , Face/embryology , Female , Genetic Association Studies , Humans , Male , White People/genetics
6.
Head Neck Pathol ; 15(1): 1-15, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33723764

ABSTRACT

Craniofacial development, one of the most complex sequences of developmental events in embryology, features a uniquely transient, pluripotent stem cell-like population known as the neural crest (NC). Neural crest cells (NCCs) originate from the dorsal aspect of the neural tube and migrate along pre-determined routes into the developing branchial arches and frontonasal plate. The exceptional rates of proliferation and migration of NCCs enable their diverse contribution to a wide variety of craniofacial structures. Subsequent differentiation of these cells gives rise to cartilage, bones, and a number of mesenchymally-derived tissues. Deficiencies in any stage of differentiation can result in facial clefts and abnormalities associated with craniofacial syndromes. A small number of conserved signaling pathways are involved in controlling NC differentiation and craniofacial development. They are used in a reiterated fashion to help define precise temporospatial cell and tissue formation. Although many aspects of their cellular and molecular control have yet to be described, it is clear that together they form intricately integrated signaling networks required for spatial orientation and developmental stability and plasticity, which are hallmarks of craniofacial development. Mutations that affect the functions of these signaling pathways are often directly or indirectly identified in congenital syndromes. Clinical applications of NC-derived mesenchymal stem/progenitor cells, persistent into adulthood, hold great promise for tissue repair and regeneration. Realization of NCC potential for regenerative therapies motivates understanding of the intricacies of cell communication and differentiation that underlie the complexities of NC-derived tissues.


Subject(s)
Face/embryology , Neural Crest , Skull/embryology , Animals , Cell Differentiation/physiology , Embryology/methods , Embryonic Development/physiology , Humans
7.
Curr Top Dev Biol ; 141: 241-277, 2021.
Article in English | MEDLINE | ID: mdl-33602490

ABSTRACT

Amniotes, a clade of terrestrial vertebrates, which includes all of the descendants of the last common ancestor of the reptiles (including dinosaurs and birds) and mammals, is one of the most successful group of animals on our planet. In addition to having an egg equipped with an amnion, an adaptation to lay eggs on land, amniotes possess a number of other major morphological characteristics. Chief among them is the amniote skull, which can be classified into several major types distinguished by the presence and number of temporal fenestrae (windows) in the posterior part. Amniotes evolved from ancestors who possessed a skull composed of a complex mosaic of small bones separated by sutures. Changes in skull composition underlie much of the large-scale evolution of amniotes with many lineages showing a trend in reduction of cranial elements known as the "Williston's Law." The skull of amniotes is also arranged into a set of modules of closely co-evolving bones as revealed by modularity and integration tests. One of the most consistently recovered and at the same time most versatile modules is the "face," anatomically defined as the anterior portion of the head. The faces of amniotes display extraordinary amount of variation, with many adaptive radiations showing parallel tendencies in facial scaling, e.g., changes in length or width. This review explores the natural history of the amniote face and discusses how a better understanding of its anatomy and developmental biology helps to explain the outstanding scale of adaptive facial diversity. We propose a model for facial evolution in the amniotes, based on the differential rate of cranial neural crest cell proliferation and the timing of their skeletal differentiation.


Subject(s)
Biological Evolution , Face/anatomy & histology , Skull/anatomy & histology , Vertebrates , Animals , Body Patterning , Face/embryology , Gene Expression Regulation, Developmental , Neural Crest/cytology , Skull/embryology , Vertebrates/anatomy & histology
8.
Surg Radiol Anat ; 43(6): 961-973, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33492439

ABSTRACT

The embryological development of the cerebral vasculature is very complex. Historical and also more recent studies based on human embryos, comparative anatomy and cerebral angiographies allowed us to better understand this vasculature development. The knowledge and understanding of such embryological development are important for physicians interested in neurovascular pathologies. Indeed, all vascular variants and almost all vascular pathologies, such as aneurysms, dolichoectasia, atherosclerosis, and neurovascular conflicts could be explained by an alteration during the embryological life. There are also many variants of these vascular structures present in normal developed adults, which are variably associated with pathological entities. Understanding the process which leads to the development of the normal cerebral arterial system in humans is, therefore, very important to have a better knowledge of the possible clinical and surgical implications of these anomalies. In this paper, we review the embryological development of the cranio-facial arterial vasculature from its beginning at approximately days 21-50 of intrauterine life, with pictures illustrating each developmental phase.


Subject(s)
Cerebral Arteries/embryology , Embryo, Mammalian/blood supply , Embryonic Development , Face/blood supply , Medical Illustration , Anatomy, Comparative , Cerebral Angiography , Cerebral Arteries/diagnostic imaging , Face/embryology , Humans
9.
J Clin Ultrasound ; 49(4): 315-321, 2021 May.
Article in English | MEDLINE | ID: mdl-33314194

ABSTRACT

PURPOSE: To compare the reliability of measurements of five fetal facial profile parameters by operators with different levels of experience, at 15 to 23 weeks of gestation in Thai fetuses. METHODS: An observational study was conducted. The inferior facial angle, anteroposterior mandibular diameter, mandible width, maxilla width, and mandible length were measured in 123 normal fetuses, using 2D ultrasonography, by three operators with different levels of experience. Each participant was examined by two operators. Each operator performed three independent measurements for each parameter and was blinded to the results of the other. Reliability of measurement was evaluated using intraclass correlation coefficient. Bland-Altman analysis was used to evaluate agreement. RESULTS: The ability to obtain a mandible length measurement was highest (100%) among the five parameters. Intraobserver variabilities of anteroposterior mandibular diameter, mandible width, maxilla width, and mandible length measurements were excellent for all operators (ICC 0.958-0.986), while those of inferior facial angle measurements were moderate to excellent (ICC 0.560-0.923), depending on the operators' experience. Interobserver variabilities varied between pairs of operators; only two parameters, anteroposterior mandibular diameter and mandible length, showed excellent interobserver variabilities (ICC >0.9), with good agreement. CONCLUSIONS: The mandible length measurement was the best parameter in terms of feasibility and reliability.


Subject(s)
Face/diagnostic imaging , Face/embryology , Fetus/diagnostic imaging , Ultrasonography, Prenatal/methods , Female , Humans , Observer Variation , Pregnancy , Reproducibility of Results , Ultrasonography, Prenatal/standards
10.
Ultrasound Obstet Gynecol ; 58(1): 56-66, 2021 07.
Article in English | MEDLINE | ID: mdl-32438493

ABSTRACT

OBJECTIVES: To develop and validate a nomogram based on fetal nuchal translucency thickness (NT) and ultrasonographic facial markers for screening for trisomy 21 in the first trimester of pregnancy. METHODS: This was a retrospective case-control study using stored two-dimensional midsagittal fetal profile images captured at 11 + 0 to 13 + 6 weeks' gestation in singleton pregnancies. We included images from 302 trisomy-21 pregnancies and 322 euploid pregnancies. Cases were divided into a training set (200 euploid + 200 with trisomy 21) and a validation set (122 euploid + 102 with trisomy 21) at a ratio of approximately 2:1. For each, the maternal age, gestational age, fetal NT and karyotype were noted, and 12 ultrasonographic fetal facial markers were measured. The least absolute shrinkage and selection operator (LASSO) method and multivariable analysis were used to select automatically the discriminative markers. Logistic regression was used to develop a LASSO model, based on the selected markers, to screen for trisomy 21 in the first trimester of pregnancy. Furthermore, 60 of the 624 images were selected randomly as a retest set to evaluate the model's robustness. The predictive performance of screening for trisomy 21 of a model based on fetal NT and maternal age and of the LASSO model was assessed using the area under the receiver-operating-characteristics curve (AUC). A nomogram was developed as an individualized tool to predict patient-specific probability for trisomy 21, which is a more visual presentation of the LASSO model. The performance of the nomogram was assessed using the C-index and calibration curve. RESULTS: Into the LASSO model were incorporated eight markers, including fetal NT, prenasal-thickness-to-nasal-bone-length ratio, facial profile line, frontomaxillary facial angle, frontonasal facial angle, mandibulomaxillary facial angle, maxilla-nasion-mandible angle and d2 (distance between the anterior edge of the prefrontal skin and the mandibulomaxillary line) (all P < 0.05). The AUCs of the LASSO model for screening for trisomy 21 were 0.983 (95% CI, 0.971-0.994) in the training set and 0.979 (95% CI, 0.966-0.993) in the validation set, and these were higher than the AUCs of all eight individual ultrasonographic markers included in the model. The AUC of the LASSO model in the retest set was 0.997 (95% CI, 0.990-1.000), indicating good robustness of the LASSO model. The AUC of the LASSO model was significantly higher than that of the model based on fetal NT and maternal age in both training and validation sets (P < 0.001 for both). The nomogram of the LASSO model showed good discrimination of trisomy 21, with C-indices of 0.983 in the training set and 0.981 in the validation set. CONCLUSIONS: We present an individualized nomogram which incorporates fetal NT and a series of ultrasonographic facial profile markers selected by the LASSO method and multivariable analysis. This nomogram can potentially be utilized as a convenient and effective tool in screening for trisomy 21 in the first trimester of pregnancy. © 2020 The Authors. Ultrasound in Obstetrics & Gynecology published by John Wiley & Sons Ltd on behalf of International Society of Ultrasound in Obstetrics and Gynecology.


Subject(s)
Down Syndrome/diagnosis , Face/embryology , Nomograms , Pregnancy Trimester, First , Prenatal Diagnosis/methods , Adult , Area Under Curve , Biomarkers/analysis , Case-Control Studies , Down Syndrome/embryology , Face/diagnostic imaging , Female , Humans , Logistic Models , Maternal Age , Nuchal Translucency Measurement , Predictive Value of Tests , Pregnancy , Retrospective Studies
11.
Development ; 148(2)2021 01 19.
Article in English | MEDLINE | ID: mdl-33318149

ABSTRACT

Mutations in the RNA helicase DDX3 have emerged as a frequent cause of intellectual disability in humans. Because many individuals carrying DDX3 mutations have additional defects in craniofacial structures and other tissues containing neural crest (NC)-derived cells, we hypothesized that DDX3 is also important for NC development. Using Xenopus tropicalis as a model, we show that DDX3 is required for normal NC induction and craniofacial morphogenesis by regulating AKT kinase activity. Depletion of DDX3 decreases AKT activity and AKT-dependent inhibitory phosphorylation of GSK3ß, leading to reduced levels of ß-catenin and Snai1: two GSK3ß substrates that are crucial for NC induction. DDX3 function in regulating these downstream signaling events during NC induction is likely mediated by RAC1, a small GTPase whose translation depends on the RNA helicase activity of DDX3. These results suggest an evolutionarily conserved role of DDX3 in NC development by promoting AKT activity, and provide a potential mechanism for the NC-related birth defects displayed by individuals harboring mutations in DDX3 and its downstream effectors in this signaling cascade.


Subject(s)
DEAD-box RNA Helicases/metabolism , Neural Crest/embryology , Neural Crest/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Xenopus Proteins/metabolism , Xenopus/embryology , Xenopus/metabolism , Animals , Cartilage/embryology , Cartilage/metabolism , Embryo, Nonmammalian/metabolism , Face/embryology , Gene Expression Regulation, Developmental , Glycogen Synthase Kinase 3 beta/metabolism , Morphogenesis/genetics , Phosphorylation , Protein Stability , Skull/embryology , Skull/metabolism , Snail Family Transcription Factors/metabolism , Wnt Signaling Pathway , Xenopus/genetics , beta Catenin/metabolism , rac1 GTP-Binding Protein/metabolism
12.
Nat Genet ; 53(1): 45-53, 2021 01.
Article in English | MEDLINE | ID: mdl-33288918

ABSTRACT

The human face is complex and multipartite, and characterization of its genetic architecture remains challenging. Using a multivariate genome-wide association study meta-analysis of 8,246 European individuals, we identified 203 genome-wide-significant signals (120 also study-wide significant) associated with normal-range facial variation. Follow-up analyses indicate that the regions surrounding these signals are enriched for enhancer activity in cranial neural crest cells and craniofacial tissues, several regions harbor multiple signals with associations to different facial phenotypes, and there is evidence for potential coordinated actions of variants. In summary, our analyses provide insights into the understanding of how complex morphological traits are shaped by both individual and coordinated genetic actions.


Subject(s)
Face/anatomy & histology , Genome-Wide Association Study , Acetylation , Enhancer Elements, Genetic/genetics , Epistasis, Genetic , Extremities/embryology , Face/embryology , Genetic Loci , Histones/metabolism , Humans , Lysine/metabolism , Meta-Analysis as Topic , Multivariate Analysis , Neural Crest/cytology , Phenotype , Polymorphism, Single Nucleotide/genetics , Skull/embryology , United Kingdom , United States
13.
Genesis ; 58(12): e23397, 2020 12.
Article in English | MEDLINE | ID: mdl-33197123

ABSTRACT

Inborn errors of cholesterol metabolism occur as a result of mutations in the cholesterol synthesis pathway (CSP). Although mutations in the CSP cause a multiple congenital anomaly syndrome, craniofacial abnormalities are a hallmark phenotype associated with these disorders. Previous studies have established that mutation of the zebrafish hmgcs1 gene (Vu57 allele), which encodes the first enzyme in the CSP, causes defects in craniofacial development and abnormal neural crest cell (NCC) differentiation. However, the molecular mechanisms by which the products of the CSP disrupt NCC differentiation are not completely known. Cholesterol is known to regulate the activity of WNT signaling, an established regulator of NCC differentiation. We hypothesized that defects in cholesterol synthesis are associated with reduced WNT signaling, consequently resulting in abnormal craniofacial development. To test our hypothesis we performed a combination of pharmaceutical inhibition, gene expression assays, and targeted rescue experiments to understand the function of the CSP and WNT signaling during craniofacial development. We demonstrate reduced expression of four canonical WNT downstream target genes in homozygous carriers of the Vu57 allele and reduced axin2 expression, a known WNT target gene, in larvae treated with Ro-48-8071, an inhibitor of cholesterol synthesis. Moreover, activation of WNT signaling via treatment with WNT agonist I completely restored the craniofacial defects present in a subset of animals carrying the Vu57 allele. Collectively, these data suggest interplay between the CSP and WNT signaling during craniofacial development.


Subject(s)
Axin Protein , Cholesterol/metabolism , Craniofacial Abnormalities/genetics , Gene Expression Regulation, Developmental/drug effects , Neural Crest/metabolism , Wnt Signaling Pathway/drug effects , Zebrafish/genetics , Alleles , Animals , Axin Protein/genetics , Axin Protein/metabolism , Cell Differentiation/drug effects , Collagen Type II/genetics , Collagen Type II/metabolism , Cyclin D1/genetics , Cyclin D1/metabolism , Down-Regulation , Embryo, Nonmammalian/metabolism , Endothelin-1/genetics , Endothelin-1/metabolism , Face/embryology , Female , Genotype , Male , Mutation , Neural Crest/embryology , Phenotype , SOXE Transcription Factors/genetics , SOXE Transcription Factors/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Zebrafish/embryology , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
14.
Exp Cell Res ; 396(2): 112300, 2020 11 15.
Article in English | MEDLINE | ID: mdl-32986984

ABSTRACT

Metazoan development relies on intricate cell differentiation, communication, and migration pathways, which ensure proper formation of specialized cell types, tissues, and organs. These pathways are crucially controlled by ubiquitylation, a reversible post-translational modification that regulates the stability, activity, localization, or interaction landscape of substrate proteins. Specificity of ubiquitylation is ensured by E3 ligases, which bind substrates and co-operate with E1 and E2 enzymes to mediate ubiquitin transfer. Cullin3-RING ligases (CRL3s) are a large class of multi-subunit E3s that have emerged as important regulators of cell differentiation and development. In particular, recent evidence from human disease genetics, animal models, and mechanistic studies have established their involvement in the control of craniofacial and brain development. Here, we summarize regulatory principles of CRL3 assembly, substrate recruitment, and ubiquitylation that allow this class of E3s to fulfill their manifold functions in development. We further review our current mechanistic understanding of how specific CRL3 complexes orchestrate neuroectodermal differentiation and highlight diseases associated with their dysregulation. Based on evidence from human disease genetics, we propose that other unknown CRL3 complexes must help coordinate craniofacial and brain development and discuss how combining emerging strategies from the field of disease gene discovery with biochemical and human pluripotent stem cell approaches will likely facilitate their identification.


Subject(s)
Brain/embryology , Cullin Proteins/metabolism , Disease/genetics , Face/embryology , Skull/embryology , Ubiquitin-Protein Ligases/metabolism , Ubiquitin/metabolism , Animals , Humans , Ubiquitin-Protein Ligases/chemistry
15.
Curr Biol ; 30(21): 4263-4269.e2, 2020 11 02.
Article in English | MEDLINE | ID: mdl-32857974

ABSTRACT

The first dinosaur embryos found inside megaloolithid eggs from Auca Mahuevo, Patagonia, were assigned to sauropod dinosaurs that lived approximately 80 million years ago. Discovered some 25 years ago, these considerably flattened specimens still remain the only unquestionable embryonic remains of a sauropod dinosaur providing an initial glimpse into titanosaurian in ovo ontogeny. Here we describe an almost intact embryonic skull, which indicates the early development of stereoscopic vision, and an unusual monocerotic face for a sauropod. The new fossil also reveals a neurovascular sensory system in the premaxilla and a partly calcified braincase, which potentially refines estimates of its prenatal stage. The embryo was found in an egg with thicker eggshell and a partly different geochemical signature than those from the egg-bearing layers described in Auca Mahuevo. The cranial bones are comparably ossified as in previously described specimens but differ in facial anatomy and size. The new specimen reveals significant heterochrony in cranial ossifications when compared with non-sauropod sauropodomorph embryos, and demonstrates that the specialized craniofacial morphology preceded the postnatal transformation of the skull anatomy in adults of related titanosaurians.


Subject(s)
Dinosaurs/embryology , Embryo, Nonmammalian/anatomy & histology , Face/embryology , Skull/embryology , Animals , Argentina , Biological Evolution , Dinosaurs/growth & development , Embryonic Development/physiology , Fossils/anatomy & histology , Maxillofacial Development/physiology , Osteogenesis/physiology , Skull/growth & development
16.
PLoS One ; 15(7): e0233582, 2020.
Article in English | MEDLINE | ID: mdl-32735620

ABSTRACT

The craniofacial developmental disorder Burn-McKeown Syndrome (BMKS) is caused by biallelic variants in the pre-messenger RNA splicing factor gene TXNL4A/DIB1. The majority of affected individuals with BMKS have a 34 base pair deletion in the promoter region of one allele of TXNL4A combined with a loss-of-function variant on the other allele, resulting in reduced TXNL4A expression. However, it is unclear how reduced expression of this ubiquitously expressed spliceosome protein results in craniofacial defects during development. Here we reprogrammed peripheral mononuclear blood cells from a BMKS patient and her unaffected mother into induced pluripotent stem cells (iPSCs) and differentiated the iPSCs into induced neural crest cells (iNCCs), the key cell type required for correct craniofacial development. BMKS patient-derived iPSCs proliferated more slowly than both mother- and unrelated control-derived iPSCs, and RNA-Seq analysis revealed significant differences in gene expression and alternative splicing. Patient iPSCs displayed defective differentiation into iNCCs compared to maternal and unrelated control iPSCs, in particular a delay in undergoing an epithelial-to-mesenchymal transition (EMT). RNA-Seq analysis of differentiated iNCCs revealed widespread gene expression changes and mis-splicing in genes relevant to craniofacial and embryonic development that highlight a dampened response to WNT signalling, the key pathway activated during iNCC differentiation. Furthermore, we identified the mis-splicing of TCF7L2 exon 4, a key gene in the WNT pathway, as a potential cause of the downregulated WNT response in patient cells. Additionally, mis-spliced genes shared common sequence properties such as length, branch point to 3' splice site (BPS-3'SS) distance and splice site strengths, suggesting that splicing of particular subsets of genes is particularly sensitive to changes in TXNL4A expression. Together, these data provide the first insight into how reduced TXNL4A expression in BMKS patients might compromise splicing and NCC function, resulting in defective craniofacial development in the embryo.


Subject(s)
Alternative Splicing , Choanal Atresia/pathology , Deafness/congenital , Gene Expression Regulation, Developmental , Heart Defects, Congenital/pathology , Induced Pluripotent Stem Cells/cytology , Models, Biological , Ribonucleoprotein, U5 Small Nuclear/deficiency , Spliceosomes/physiology , Apoptosis , Cell Differentiation , Cellular Reprogramming Techniques , Choanal Atresia/genetics , Clone Cells , Deafness/genetics , Deafness/pathology , Epithelial-Mesenchymal Transition , Exons/genetics , Face/embryology , Facies , Female , Head/embryology , Heart Defects, Congenital/genetics , Humans , Neural Crest/cytology , Promoter Regions, Genetic/genetics , RNA Splice Sites , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ribonucleoprotein, U5 Small Nuclear/genetics , Sequence Deletion , Transcription Factor 7-Like 2 Protein/genetics , Wnt Signaling Pathway
17.
Int J Dev Biol ; 64(4-5-6): 383-391, 2020.
Article in English | MEDLINE | ID: mdl-32658997

ABSTRACT

Clinical dysmorphology is a medical specialty which requires training to systematically observe aberrations in facial development and to understand patterns in the recognition of underlying genetic syndromes. An understanding of normal facial embryology and structure, genetic mechanisms that contribute to facial development and the influence of age, sex, epigenetic, environmental and teratogen effects that contribute to facial dysmorphology are essential. The role of software programmes and databases in achieving diagnoses in subtler phenotypes is growing. A description of specific dysmorphisms of various parts of the human face and key genetic and mechanistic pathways are discussed in this review. Recognizing facial patterns and genetic syndromes efficiently aids in planning appropriate tests, securing an accurate diagnosis, counselling and predicting outcomes and offering interventions and therapies where available.


Subject(s)
Congenital Abnormalities/genetics , Embryonic Development/genetics , Face/embryology , Gene Expression Regulation, Developmental , Craniosynostoses/embryology , Craniosynostoses/genetics , Craniosynostoses/metabolism , Female , Humans , Male , Mesoderm/embryology , Mesoderm/metabolism , Neural Crest/embryology , Neural Crest/metabolism
18.
Adv Exp Med Biol ; 1236: 137-155, 2020.
Article in English | MEDLINE | ID: mdl-32304072

ABSTRACT

The formation of the head and face is a complex process which involves many different signaling cues regulating the migration, differentiation, and proliferation of the neural crest. This highly complex process is very error-prone, resulting in craniofacial defects in nearly 10,000 births in the United States annually. Due to the highly conserved mechanisms of craniofacial development, animal models are widely used to understand the pathogenesis of various human diseases and assist in the diagnosis and generation of preventative therapies and treatments. Here, we provide a brief background of craniofacial development and discuss several rare diseases affecting craniofacial bone development. We focus on rare congenital diseases of the cranial bone, facial jaw bones, and two classes of diseases, ciliopathies and RASopathies. Studying the animal models of these rare diseases sheds light not only on the etiology and pathology of each disease, but also provides meaningful insights towards the mechanisms which regulate normal development of the head and face.


Subject(s)
Craniofacial Abnormalities , Disease Models, Animal , Head/embryology , Animals , Craniofacial Abnormalities/diagnosis , Craniofacial Abnormalities/prevention & control , Craniofacial Abnormalities/therapy , Face/embryology , Humans , Neural Crest/embryology , Skull/embryology
19.
Dis Model Mech ; 13(5)2020 05 26.
Article in English | MEDLINE | ID: mdl-32152089

ABSTRACT

A critical barrier in the treatment of endosomal and lysosomal diseases is the lack of understanding of the in vivo functions of the putative causative genes. We addressed this by investigating a key pair of endocytic adaptor proteins, PH domain-containing endocytic trafficking adaptor 1 and 2 (PHETA1/2; also known as FAM109A/B, Ses1/2, IPIP27A/B), which interact with the protein product of OCRL, the causative gene for Lowe syndrome. Here, we conducted the first study of PHETA1/2 in vivo, utilizing the zebrafish system. We found that impairment of both zebrafish orthologs, pheta1 and pheta2, disrupted endocytosis and ciliogenesis in renal tissues. In addition, pheta1/2 mutant animals exhibited reduced jaw size and delayed chondrocyte differentiation, indicating a role in craniofacial development. Deficiency of pheta1/2 resulted in dysregulation of cathepsin K, which led to an increased abundance of type II collagen in craniofacial cartilages, a marker of immature cartilage extracellular matrix. Cathepsin K inhibition rescued the craniofacial phenotypes in the pheta1/2 double mutants. The abnormal renal and craniofacial phenotypes in the pheta1/2 mutant animals were consistent with the clinical presentation of a patient with a de novo arginine (R) to cysteine (C) variant (R6C) of PHETA1. Expressing the patient-specific variant in zebrafish exacerbated craniofacial deficits, suggesting that the R6C allele acts in a dominant-negative manner. Together, these results provide insights into the in vivo roles of PHETA1/2 and suggest that the R6C variant is contributory to the pathogenesis of disease in the patient.This article has an associated First Person interview with the first author of the paper.


Subject(s)
Adaptor Proteins, Signal Transducing/deficiency , Endocytosis , Face/embryology , Kidney/embryology , Skull/embryology , Zebrafish Proteins/deficiency , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Sequence , Animals , CRISPR-Cas Systems/genetics , Cathepsin K/metabolism , Cell Differentiation , Chondrocytes/pathology , Cilia/pathology , Collagen Type II/metabolism , Genes, Dominant , HeLa Cells , Humans , Morphogenesis , Motor Activity , Mutation/genetics , Pronephros/pathology , Undiagnosed Diseases/diagnostic imaging , Undiagnosed Diseases/genetics , Vesicular Transport Proteins/chemistry , Vesicular Transport Proteins/genetics , Zebrafish , Zebrafish Proteins/chemistry , Zebrafish Proteins/metabolism
20.
Birth Defects Res ; 112(5): 404-417, 2020 03.
Article in English | MEDLINE | ID: mdl-31944002

ABSTRACT

BACKGROUND: Previous studies suggested associations between maternal smoking, a source of exposure to polycyclic aromatic hydrocarbons (PAHs) and other chemicals, and central nervous system and face birth defects; however, no previous studies have evaluated maternal occupational PAH exposure itself. METHODS: Jobs held in the periconceptional period were retrospectively assigned for occupational PAH exposures. Associations between maternal occupational PAH exposure and selected rare defects of the face (cataracts, microphthalmia, glaucoma, microtia, and choanal atresia) and central nervous system (holoprosencephaly, hydrocephaly, cerebellar hypoplasia, and Dandy-Walker malformation) were evaluated using data from the National Birth Defects Prevention Study, a population-based case-control study in the United States. Crude and adjusted odds ratios (ORs) with 95% confidence intervals were calculated to estimate associations between each evaluated defect and PAH exposure using multivariable logistic regression. RESULTS: Food and beverage serving, as well as cooks and food preparation occupations, were among the most frequent jobs held by exposed mothers. Cataracts, microtia, microphthalmia, and holoprosencephaly were significantly associated with PAH exposure with evidence of dose-response (P-values for trend ≤.05). Hydrocephaly was associated with any PAH exposure, but not significant for trend. Sensitivity analyses that reduced possible sources of exposure misclassification tended to strengthen associations. CONCLUSIONS: This is the first population-based case-control study to evaluate associations between maternal occupational PAH exposures and these rare birth defects of the central nervous system and face.


Subject(s)
Congenital Abnormalities/etiology , Polycyclic Aromatic Hydrocarbons/adverse effects , Abnormalities, Drug-Induced/etiology , Adult , Case-Control Studies , Central Nervous System/embryology , Face/abnormalities , Face/embryology , Female , Humans , Logistic Models , Maternal Exposure , Middle Aged , Mothers , Occupational Exposure , Odds Ratio , Retrospective Studies , Risk Factors , United States , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...