Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 58
Filtrer
1.
Genesis ; 59(12): e23453, 2021 12.
Article de Anglais | MEDLINE | ID: mdl-34664392

RÉSUMÉ

The vertebrate Six (Sine oculis homeobox) family of homeodomain transcription factors plays critical roles in the development of several organs. Six1 plays a central role in cranial placode development, including the precursor tissues of the inner ear, as well as other cranial sensory organs and the kidney. In humans, mutations in SIX1 underlie some cases of Branchio-oto-renal (BOR) syndrome, which is characterized by moderate-to-severe hearing loss. We utilized CRISPR/Cas9 technology to establish a six1 mutant line in Xenopus tropicalis that is available to the research community. We demonstrate that at larval stages, the six1-null animals show severe disruptions in gene expression of putative Six1 target genes in the otic vesicle, cranial ganglia, branchial arch, and neural tube. At tadpole stages, six1-null animals display dysmorphic Meckel's, ceratohyal, and otic capsule cartilage morphology. This mutant line will be of value for the study of the development of several organs as well as congenital syndromes that involve these tissues.


Sujet(s)
Syndrome branchio-oto-rénal/génétique , Malformations/génétique , Perte d'audition/génétique , Protéines à homéodomaine/génétique , Protéines de Xénope/génétique , Animaux , Région branchiale/croissance et développement , Région branchiale/anatomopathologie , Syndrome branchio-oto-rénal/physiopathologie , Systèmes CRISPR-Cas/génétique , Malformations/anatomopathologie , Développement embryonnaire/génétique , Ganglions parasympathiques/croissance et développement , Ganglions parasympathiques/anatomopathologie , Expression des gènes , Régulation de l'expression des gènes au cours du développement/génétique , Perte d'audition/physiopathologie , Humains , Tube neural/croissance et développement , Tube neural/anatomopathologie , Crâne/croissance et développement , Crâne/anatomopathologie , Facteurs de transcription/génétique , Xenopus/génétique , Xenopus/croissance et développement
2.
Development ; 148(22)2021 11 15.
Article de Anglais | MEDLINE | ID: mdl-35020873

RÉSUMÉ

The dynamics of multipotent neural crest cell differentiation and invasion as cells travel throughout the vertebrate embryo remain unclear. Here, we preserve spatial information to derive the transcriptional states of migrating neural crest cells and the cellular landscape of the first four chick cranial to cardiac branchial arches (BA1-4) using label-free, unsorted single-cell RNA sequencing. The faithful capture of branchial arch-specific genes led to identification of novel markers of migrating neural crest cells and 266 invasion genes common to all BA1-4 streams. Perturbation analysis of a small subset of invasion genes and time-lapse imaging identified their functional role to regulate neural crest cell behaviors. Comparison of the neural crest invasion signature to other cell invasion phenomena revealed a shared set of 45 genes, a subset of which showed direct relevance to human neuroblastoma cell lines analyzed after exposure to the in vivo chick embryonic neural crest microenvironment. Our data define an important spatio-temporal reference resource to address patterning of the vertebrate head and neck, and previously unidentified cell invasion genes with the potential for broad impact.


Sujet(s)
Région branchiale/croissance et développement , Tête/croissance et développement , Cou/croissance et développement , Crête neurale/croissance et développement , Animaux , Plan d'organisation du corps/génétique , Région branchiale/embryologie , Différenciation cellulaire/génétique , Mouvement cellulaire/génétique , Microenvironnement cellulaire/génétique , Embryon de poulet , Embryon de mammifère , Embryon non mammalien , Développement embryonnaire/génétique , Tête/embryologie , Humains , Mésoderme/croissance et développement , Cellules souches multipotentes/cytologie , Cou/embryologie , Crête neurale/métabolisme , Neuroblastome/génétique , Neuroblastome/anatomopathologie , Organogenèse/génétique , Microenvironnement tumoral/génétique , Vertébrés/génétique , Vertébrés/croissance et développement
3.
Development ; 147(24)2020 12 23.
Article de Anglais | MEDLINE | ID: mdl-33158927

RÉSUMÉ

Pharyngeal arches (PAs) are segmented by endodermal outpocketings called pharyngeal pouches (PPs). Anterior and posterior PAs appear to be generated by different mechanisms, but it is unclear how the anterior and posterior PAs combine. Here, we addressed this issue with precise live imaging of PP development and cell tracing of pharyngeal endoderm in zebrafish embryos. We found that two endodermal bulges are initially generated in the future second PP (PP2) region, which separates anterior and posterior PAs. Subsequently, epithelial remodeling causes contact between these two bulges, resulting in the formation of mature PP2 with a bilayered morphology. The rostral and caudal bulges develop into the operculum and gill, respectively. Development of the caudal PP2 and more posterior PPs is affected by impaired retinoic acid signaling or pax1a/b dysfunction, suggesting that the rostral front of posterior PA development corresponds to the caudal PP2. Our study clarifies an aspect of PA development that is essential for generation of a seamless array of PAs in zebrafish.


Sujet(s)
Région branchiale/croissance et développement , Développement embryonnaire/génétique , Endoderme/croissance et développement , Facteurs de transcription PAX/génétique , Protéines de poisson-zèbre/génétique , Animaux , Plan d'organisation du corps/génétique , Embryon non mammalien , Endoderme/métabolisme , Épithélium/croissance et développement , Épithélium/métabolisme , Régulation de l'expression des gènes au cours du développement/génétique , Branchies/croissance et développement , Mésoderme/croissance et développement , Crête neurale/croissance et développement , Pharynx/croissance et développement , Transduction du signal/génétique , Danio zébré/génétique , Danio zébré/croissance et développement
4.
Zygote ; 28(3): 208-216, 2020 Jun.
Article de Anglais | MEDLINE | ID: mdl-32077403

RÉSUMÉ

In the present study, the morphological development of the Brycon amazonicus digestive tract is described to provide basic knowledge for nutritional studies and, therefore, increase the survival of this species during larviculture. Samples were collected from hatching up to 25 days of age, measured, processed and observed under a stereomicroscope and light microscopy. Newly hatched larvae presented their digestive tract as a straight tube, dorsal to the yolk sac, lined with a single layer of undifferentiated cells. At 24 h post-hatching (hPH), the buccopharyngeal cavity was open, but the posterior region of the digestive tube remained closed. At 25 hPH, the digestive tube was completely open and could be divided into buccopharyngeal cavity, oesophagus and intestine. At 35 hPH, the intestine presented a dilatation in the proximal region, which had the function of storing food. Differentiation of the stomach started at 83 hPH, and mucous cells were observed in the epithelium. These cells are important in the production of mucus, whose function is to protect the organ against acidity, although the gastric glands began developing only from 171 hPH, when three stomach regions were observed: cardiac, fundic and pyloric. The gastric glands were observed in the cardiac region, indicating that this organ already had digestive functionality. From 243 hPH, the absorption and assimilation of nutrients were already possible but, only from 412 hPH, the digestive tract was completely developed and functional.


Sujet(s)
Characiformes/croissance et développement , Tube digestif/croissance et développement , Animaux , Région branchiale/cytologie , Région branchiale/embryologie , Région branchiale/croissance et développement , Characiformes/anatomie et histologie , Characiformes/embryologie , Embryon non mammalien/cytologie , Embryon non mammalien/embryologie , Développement embryonnaire/physiologie , Muqueuse gastrique/cytologie , Muqueuse gastrique/embryologie , Muqueuse gastrique/croissance et développement , Tube digestif/cytologie , Tube digestif/embryologie , Larve/cytologie , Larve/croissance et développement , Muqueuse de la bouche/cytologie , Muqueuse de la bouche/embryologie , Muqueuse de la bouche/croissance et développement , Facteurs temps
5.
Nucleic Acids Res ; 48(5): e27, 2020 03 18.
Article de Anglais | MEDLINE | ID: mdl-31974574

RÉSUMÉ

Transcription factors (TFs) can bind DNA in a cooperative manner, enabling a mutual increase in occupancy. Through this type of interaction, alternative binding sites can be preferentially bound in different tissues to regulate tissue-specific expression programmes. Recently, deep learning models have become state-of-the-art in various pattern analysis tasks, including applications in the field of genomics. We therefore investigate the application of convolutional neural network (CNN) models to the discovery of sequence features determining cooperative and differential TF binding across tissues. We analyse ChIP-seq data from MEIS, TFs which are broadly expressed across mouse branchial arches, and HOXA2, which is expressed in the second and more posterior branchial arches. By developing models predictive of MEIS differential binding in all three tissues, we are able to accurately predict HOXA2 co-binding sites. We evaluate transfer-like and multitask approaches to regularizing the high-dimensional classification task with a larger regression dataset, allowing for the creation of deeper and more accurate models. We test the performance of perturbation and gradient-based attribution methods in identifying the HOXA2 sites from differential MEIS data. Our results show that deep regularized models significantly outperform shallow CNNs as well as k-mer methods in the discovery of tissue-specific sites bound in vivo.


Sujet(s)
Région branchiale/métabolisme , Apprentissage profond , Protéines à homéodomaine/génétique , Protéine du site-1 d'intégration des virus myéloïdes écotropiques/génétique , ARN/génétique , Animaux , Sites de fixation , Région branchiale/croissance et développement , Immunoprécipitation de la chromatine , Biologie informatique/méthodes , Biologie informatique/statistiques et données numériques , Embryon de mammifère , Régulation de l'expression des gènes au cours du développement , Séquençage nucléotidique à haut débit , Protéines à homéodomaine/métabolisme , Souris , Modèles génétiques , Protéine du site-1 d'intégration des virus myéloïdes écotropiques/métabolisme , Spécificité d'organe , Loi de Poisson , Liaison aux protéines , Isoformes de protéines/génétique , Isoformes de protéines/métabolisme , ARN/métabolisme
6.
J Biol Chem ; 295(51): 17632-17645, 2020 12 18.
Article de Anglais | MEDLINE | ID: mdl-33454003

RÉSUMÉ

Thoracic great vessels such as the aorta and subclavian arteries are formed through dynamic remodeling of embryonic pharyngeal arch arteries (PAAs). Previous work has shown that loss of a basic helix-loop-helix transcription factor Hey1 in mice causes abnormal fourth PAA development and lethal great vessel anomalies resembling congenital malformations in humans. However, how Hey1 mediates vascular formation remains unclear. In this study, we revealed that Hey1 in vascular endothelial cells, but not in smooth muscle cells, played essential roles for PAA development and great vessel morphogenesis in mouse embryos. Tek-Cre-mediated Hey1 deletion in endothelial cells affected endothelial tube formation and smooth muscle differentiation in embryonic fourth PAAs and resulted in interruption of the aortic arch and other great vessel malformations. Cell specificity and signal responsiveness of Hey1 expression were controlled through multiple cis-regulatory regions. We found two distal genomic regions that had enhancer activity in endothelial cells and in the pharyngeal epithelium and somites, respectively. The novel endothelial enhancer was conserved across species and was specific to large-caliber arteries. Its transcriptional activity was regulated by Notch signaling in vitro and in vivo, but not by ALK1 signaling and other transcription factors implicated in endothelial cell specificity. The distal endothelial enhancer was not essential for basal Hey1 expression in mouse embryos but may likely serve for Notch-dependent transcriptional control in endothelial cells together with the proximal regulatory region. These findings help in understanding the significance and regulation of endothelial Hey1 as a mediator of multiple signaling pathways in embryonic vascular formation.


Sujet(s)
Protéines du cycle cellulaire/métabolisme , Endothélium/métabolisme , Récepteurs Notch/métabolisme , Animaux , Artères/croissance et développement , Artères/métabolisme , Région branchiale/vascularisation , Région branchiale/croissance et développement , Protéines du cycle cellulaire/déficit , Protéines du cycle cellulaire/génétique , Différenciation cellulaire , Embryon de mammifère/métabolisme , Endothélium/cytologie , Femelle , Humains , Souris , Souris knockout , Morphogenèse , Myocytes du muscle lisse/cytologie , Myocytes du muscle lisse/métabolisme , /métabolisme , Séquences d'acides nucléiques régulatrices , Transduction du signal , Activation de la transcription
7.
Dev Dyn ; 248(9): 795-812, 2019 09.
Article de Anglais | MEDLINE | ID: mdl-31219639

RÉSUMÉ

BACKGROUND: Congenital cardiovascular malformations are the most common birth defects affecting children. Several of these defects occur in structures developing from neural crest cells. One of the key signaling pathways regulating cardiac neural crest cell (CNCC) development involves the endothelin-A receptor (Ednra). However, the exact function of Ednra signaling in CNCC is unknown. RESULTS: The fate mapping of CNCC in Ednra embryos indicated that the migration of these cells is aberrant in the cardiac outflow tract (OFT), but not in the pharyngeal arches. This premature arrest of CNCC migration occurs independently of CNCC proliferation and apoptosis changes and major gene expression changes. Analysis of the Rho family of small GTPases in the mutant embryos revealed that Cdc42 failed to localize normally in the CNCC migrating in the OFT. The inhibition of Cdc42 activity in cultured embryos recapitulated the migratory phenotype observed in Ednra mice. Further analyses revealed that Cdc42 is part of the signaling pathway activated by endothelin specifically in OFT CNCC to control their migration. CONCLUSIONS: These results indicated that the activation of Cdc42 by endothelin signaling is important for CNCC migration in the OFT but this pathway is not involved in mandibular or pharyngeal arch artery patterning.


Sujet(s)
Mouvement cellulaire , Endothélines/physiologie , Myocarde/cytologie , Crête neurale/cytologie , Protéine G cdc42/métabolisme , Animaux , Région branchiale/embryologie , Région branchiale/croissance et développement , Embryon de mammifère , Souris , Organogenèse , Récepteur de type A de l'endothéline/métabolisme , Récepteur de type A de l'endothéline/physiologie , Transduction du signal/physiologie
8.
J Fish Biol ; 94(2): 223-230, 2019 Feb.
Article de Anglais | MEDLINE | ID: mdl-30565702

RÉSUMÉ

This study provides a comprehensive description of chondrocranial development before, during and after larval metamorphosis in the tongue sole Cynoglossus semilaevis, a commercially valuable flatfish in China. Samples were collected at regular intervals ranging from 1 to 23 days post hatching (dph). Based on observations of cleared and double-stained specimens and images from sections stained with safranin O-fast green, major morphological events during early development were described. No cartilaginous structure was visible at hatching. From 2 dph onwards, cartilaginous structures such as the trabecular bar and some elements of the mandibular, hyoid and branchial arches appeared. At this time also, cartilaginous structures of the neurocranium started to form. Hypertrophic chondrocytes could be observed in many splanchnocranium elements at 5 dph. The start of ossification was indicated by alizarin red stain visible at 14 dph. At 17 dph, most of the cartilaginous skeleton was ossified. Soon after, the right eye started to migrate and pass through a slit beneath the dorsal-fin base and above the skull. Metamorphosis was complete at 20 dph, at which time the dorsal-fin base cartilage extended onto the anterior region of the head. Meanwhile, extremities of the hyoid and branchial arch elements remained cartilaginous. At 23 dph, endochondral ossification of the splanchnocranium was nearly complete. Unlike previous observations of other Pleuronectiformes, our study indicates that endochondral ossification of C. semilaevis skull cartilage occurs before metamorphosis.


Sujet(s)
Cartilage/croissance et développement , Pleuronectidae/croissance et développement , Métamorphose biologique , Crâne/croissance et développement , Animaux , Région branchiale/croissance et développement , Chine , Poissons plats , Pleuronectidae/anatomie et histologie , Mandibule/croissance et développement , Ostéogenèse
9.
Morphologie ; 102(339): 243-249, 2018 Dec.
Article de Anglais | MEDLINE | ID: mdl-30268353

RÉSUMÉ

The origin of the mammalian middle ear ossicles from mandibular and hyoid pharyngeal arches remains controversial and discussed. Two adverse theories are proposed. The first claims that malleus and incus derive from the Meckel's cartilage of the mandibular arch, and stapes from Reichert's cartilage of the hyoid arch. The second postulates that handle of malleus and long process of the incus are derived from the second arch as well as the stapes. Contradictory analyses support alternatively each theory without any experimental evidence. In order to bring new data, we analyzed by immunohistochemistry the expression of Hox-A2 protein in ossicular anlagen in E11 to 13 mouse embryos. HOXA2 gene is known to be expressed in second arch cells and to be absent from mandibular arch derivatives. Surprisingly, Hox-A2 protein was present in all ossicular primordia, as well in Reichert's cartilage. Meckel's cartilage was free of staining. Unlabeled cells were also present in ossicular blastemata. These results suggest that ossicular condensations could arise from mixed cell populations originated in both mandibular and hyoid pharyngeal arches. However, we cannot exclude that diffuse Hox-A2 immunoreactivity could correspond to a secondary expression in craniofacial mesenchyme independently from the branchial origin of cells.


Sujet(s)
Région branchiale/croissance et développement , Osselets de l'audition/croissance et développement , Protéines à homéodomaine/métabolisme , Animaux , Région branchiale/métabolisme , Osselets de l'audition/métabolisme , Embryon de mammifère , Femelle , Souris , Grossesse
10.
Article de Anglais | MEDLINE | ID: mdl-28887162

RÉSUMÉ

Gulf toadfish (Opsanus beta) can switch from continuously excreting ammonia as their primary nitrogenous waste to excreting predominantly urea in distinct pulses. Previous studies have shown that the neurotransmitter serotonin (5-HT) is involved in controlling this process, but it is unknown if 5-HT availability is under central nervous control or if the 5-HT signal originates from a peripheral source. Following up on a previous study, cranial nerves IX (glossopharyngeal) and X (vagus) were sectioned to further characterize their role in controlling pulsatile urea excretion and 5-HT release within the gill. In contrast to an earlier study, nerve sectioning did not result in a change in urea pulse frequency. Total urea excretion, average pulse size, total nitrogen excretion, and percent ureotely were reduced the first day post-surgery in nerve-sectioned fish but recovered by 72h post-surgery. Nerve sectioning also had no effect on toadfish urea transporter (tUT), 5-HT transporter (SERT), or 5-HT2A receptor mRNA expression or 5-HT and 5-hydroxyindoleacetic acid (5-HIAA) abundance in the gill, all of which were found consistently across the three gill arches except 5-HIAA, which was undetectable in the first gill arch. Our findings indicate that the central nervous system does not directly control pulsatile urea excretion or local changes in gill 5-HT and 5-HIAA abundance.


Sujet(s)
Batrachoïdiformes/physiologie , Région branchiale/métabolisme , Branchies/métabolisme , Sérotonine/métabolisme , Urée/métabolisme , Animaux , Océan Atlantique , Batrachoïdiformes/sang , Batrachoïdiformes/croissance et développement , Région branchiale/croissance et développement , Région branchiale/innervation , Surpeuplement , Dénervation/médecine vétérinaire , Protéines de poisson/génétique , Protéines de poisson/métabolisme , Floride , Régulation de l'expression des gènes au cours du développement , Branchies/croissance et développement , Branchies/innervation , Nerf glossopharyngien/chirurgie , Hydrocortisone/sang , Acide 5-hydroxy-indole-3-acétique/métabolisme , Protéines de transport membranaire/génétique , Protéines de transport membranaire/métabolisme , Récepteur de la sérotonine de type 5-HT2A/génétique , Récepteur de la sérotonine de type 5-HT2A/métabolisme , Sérotonine/sang , Transporteurs de la sérotonine/génétique , Transporteurs de la sérotonine/métabolisme , Stress physiologique , Urée/sang , Nerf vague/chirurgie ,
11.
Nat Commun ; 8: 14640, 2017 03 03.
Article de Anglais | MEDLINE | ID: mdl-28256502

RÉSUMÉ

Vasculogenic defects of great vessels (GVs) are a major cause of congenital cardiovascular diseases. However, genetic regulators of endothelial precursors in GV vasculogenesis remain largely unknown. Here we show that Stat4, a transcription factor known for its regulatory role of pro-inflammatory signalling, promotes GV vasculogenesis in zebrafish. We find stat4 transcripts highly enriched in nkx2.5+ endothelial precursors in the pharynx and demonstrate that genetic ablation of stat4 causes stenosis of pharyngeal arch arteries (PAAs) by suppressing PAAs 3-6 angioblast development. We further show that stat4 is a downstream target of nkx2.5 and that it autonomously promotes proliferation of endothelial precursors of the mesoderm. Mechanistically, stat4 regulates the emerging PAA angioblasts by inhibiting the expression of hdac3 and counteracting the effect of stat1a. Altogether, our study establishes a role for Stat4 in zebrafish great vessel development, and suggests that Stat4 may serve as a therapeutic target for GV defects.


Sujet(s)
Artères/croissance et développement , Maladies cardiovasculaires/génétique , Régulation de l'expression des gènes au cours du développement , Morphogenèse/génétique , Facteur de transcription STAT-4/physiologie , Protéines de poisson-zèbre/métabolisme , Animaux , Animal génétiquement modifié , Artères/malformations , Région branchiale/vascularisation , Région branchiale/croissance et développement , Différenciation cellulaire/génétique , Prolifération cellulaire/génétique , Embryon non mammalien , Cellules endothéliales/physiologie , Techniques de knock-down de gènes , Histone deacetylases/génétique , Histone deacetylases/métabolisme , Protéine homéotique Nkx-2.5/génétique , Protéine homéotique Nkx-2.5/métabolisme , Mésoderme/cytologie , Mésoderme/croissance et développement , Modèles animaux , Morpholinos/génétique , Facteur de transcription STAT-4/génétique , Facteur de transcription STAT-4/métabolisme , Danio zébré , Protéines de poisson-zèbre/génétique
12.
J Morphol ; 278(3): 418-442, 2017 03.
Article de Anglais | MEDLINE | ID: mdl-28176372

RÉSUMÉ

The head is considered the major novelty of the vertebrates and directly linked to their evolutionary success. Its form and development as well as its function, for example in feeding, is of major interest for evolutionary biologists. In this study, we describe the skeletal development of the cranium and pectoral girdle in Siberian (Acipenser baerii) and Russian sturgeon (A. gueldenstaedtii), two species that are commonly farmed in aquaculture and increasingly important in developmental studies. This study comprises the development of the neuro-, viscero- and dermatocranium and the dermal and chondral components of the pectoral girdle, from first condensation of chondrocytes in prehatchlings to the early juvenile stage and reveals a clear pattern in formation. The otic capsules, the parachordal cartilages, and the trabeculae cranii are the first centers of chondrification, at 8.4mm TL. These are followed by the mandibular, then the hyoid, and later the branchial arches. Teeth form early on the dentary, dermopalatine, and palatopterygoid, and then appear later in the buccal cavity as dorsal and ventral toothplates. With ongoing chondrification in the neurocranium a capsule around the brain and a strong rostrum are formed. Dermal ossifications start to form before closure of the dorsal neurocranial fenestrae. Perichondral ossification of cartilage bones occurs much later in ontogeny. Our results contribute data bearing on the homology of elements such as the lateral rostral canal bone that we regard homologous to the antorbital of other actinopterygians based on its sequence of formation, position and form. We further raise doubts on the homology of the posterior ceratobranchial among Actinopteri based on the formation of the hyoid arch elements. We also investigate the basibranchials and the closely associated unidentified gill-arch elements and show that they are not homologous. J. Morphol. 278:418-442, 2017. © 2017 Wiley Periodicals, Inc.


Sujet(s)
Os du membre supérieur/croissance et développement , Poissons/croissance et développement , Crâne/croissance et développement , Animaux , Évolution biologique , Os du membre supérieur/embryologie , Région branchiale/embryologie , Région branchiale/croissance et développement , Cartilage/croissance et développement , Poissons/embryologie , Poissons/génétique , Os hyoïde/embryologie , Os hyoïde/croissance et développement , Ostéogenèse , Crâne/embryologie
13.
Dev Biol ; 426(2): 245-254, 2017 06 15.
Article de Anglais | MEDLINE | ID: mdl-27364468

RÉSUMÉ

The dorsal ventral axis of vertebrates requires high BMP activity for ventral development and inhibition of BMP activity for dorsal development. Presumptive dorsal regions of the embryo are protected from the ventralizing activity of BMPs by the secretion of BMP antagonists from the mesoderm. Noggin, one such antagonist, binds BMP ligands and prevents them from binding their receptors, however, a unique role for Noggin in amphibian development has remained unclear. Previously, we used zinc-finger nucleases to mutagenize the noggin locus in Xenopus tropicalis. Here, we report on the phenotype of noggin mutant frogs as a result of breeding null mutations to homozygosity. Early homozygous noggin mutant embryos are indistinguishable from wildtype siblings, with normal neural induction and neural tube closure. However, in late tadpole stages mutants present severe ventral craniofacial defects, notably a fusion of Meckel's cartilage to the palatoquadrate cartilage. Consistent with a noggin loss-of-function, mutants show expansions of BMP target gene expression and the mutant phenotype can be rescued with transient BMP inhibition. These results demonstrate that in amphibians, Noggin is dispensable for early embryonic patterning but is critical for cranial skeletogenesis.


Sujet(s)
Région branchiale/croissance et développement , Protéines de transport/physiologie , Protéines de Xénope/physiologie , Xenopus/croissance et développement , Allèles , Animaux , Plan d'organisation du corps , Protéines morphogénétiques osseuses/pharmacologie , Protéines morphogénétiques osseuses/physiologie , Protéines de transport/génétique , Cartilage/malformations , Différenciation cellulaire , Embryon non mammalien/métabolisme , Embryon non mammalien/ultrastructure , Follistatine/déficit , Follistatine/génétique , Techniques de knock-out de gènes , Glycoprotéines/déficit , Glycoprotéines/génétique , Homozygote , Protéines et peptides de signalisation intercellulaire/déficit , Protéines et peptides de signalisation intercellulaire/génétique , Larve , Mandibule/malformations , Morpholinos/pharmacologie , Crâne/malformations , Xenopus/embryologie , Protéines de Xénope/déficit , Protéines de Xénope/génétique
14.
Hum Mol Genet ; 24(R1): R50-9, 2015 Oct 15.
Article de Anglais | MEDLINE | ID: mdl-26085576

RÉSUMÉ

Development of the human skull and face is a highly orchestrated and complex three-dimensional morphogenetic process, involving hundreds of genes controlling the coordinated patterning, proliferation and differentiation of tissues having multiple embryological origins. Craniofacial malformations that occur because of abnormal development (including cleft lip and/or palate, craniosynostosis and facial dysostoses), comprise over one-third of all congenital birth defects. High-throughput sequencing has recently led to the identification of many new causative disease genes and functional studies have clarified their mechanisms of action. We present recent findings in craniofacial genetics and discuss how this information together with developmental studies in animal models is helping to increase understanding of normal craniofacial development.


Sujet(s)
Malformations crâniofaciales/génétique , Malformations crâniofaciales/métabolisme , Morphogenèse , Palais/croissance et développement , Animaux , Région branchiale/malformations , Région branchiale/croissance et développement , Région branchiale/métabolisme , Malformations crâniofaciales/embryologie , Modèles animaux de maladie humaine , Génomique , Humains , Palais/malformations , Palais/métabolisme , Crâne/malformations , Crâne/croissance et développement , Crâne/métabolisme
15.
Endocrinology ; 156(7): 2384-94, 2015 Jul.
Article de Anglais | MEDLINE | ID: mdl-25872007

RÉSUMÉ

In vertebrates, parathyroid hormone (PTH) is important for skeletogenesis and Ca(2+) homeostasis. However, little is known about the molecular mechanisms by which PTH regulates skeleton formation and Ca(2+) balance during early development. Using larval zebrafish as an in vivo model system, we determined that PTH1 regulates the differentiation of epithelial cells and the development of craniofacial cartilage. We demonstrated that translational gene knockdown of PTH1 decreased Ca(2+) uptake at 4 days after fertilization. We also observed that PTH1-deficient fish exhibited reduced numbers of epithelial Ca(2+) channel (ecac)-expressing cells, Na(+)/K(+)-ATPase-rich cells, and H(+)-ATPase-rich cells. Additionally, the density of epidermal stem cells was decreased substantially in the fish experiencing PTH1 knockdown. Knockdown of PTH1 caused a shortening of the jaw and impeded the development of branchial arches. Results from in situ hybridization suggested that the expression of collagen 2a1a (marker for proliferating chondrocytes) was substantially reduced in the cartilage that forms the jaw and branchial aches. Disorganization of chondrocytes in craniofacial cartilage also was observed in PTH1-deficient fish. The results of real-time PCR demonstrated that PTH1 morphants failed to express the transcription factor glial cell missing 2 (gcm2). Coinjection of PTH1 morpholino with gcm2 capped RNA rescued the phenotypes observed in the PTH1 morphants, suggesting that the defects in PTH1-deficient fish were caused, at least in part, by the suppression of gcm2. Taken together, the results of the present study reveal critical roles for PTH1 in promoting the differentiation of epidermal stem cells into mature ionocytes and cartilage formation during development.


Sujet(s)
Cartilage/croissance et développement , Protéines de liaison à l'ADN/génétique , Régulation de l'expression des gènes au cours du développement/génétique , Branchies/croissance et développement , Hormone parathyroïdienne/métabolisme , ARN messager/métabolisme , Facteurs de transcription/génétique , Protéines de poisson-zèbre/métabolisme , Animaux , Région branchiale/croissance et développement , Région branchiale/métabolisme , Cartilage/métabolisme , Différenciation cellulaire , Chondrocytes/cytologie , Chondrocytes/métabolisme , Cellules épithéliales/cytologie , Cellules épithéliales/métabolisme , Techniques de knock-down de gènes , Branchies/métabolisme , Larve , Proton-Translocating ATPases/métabolisme , Réaction de polymérisation en chaine en temps réel , Sodium-Potassium-Exchanging ATPase/métabolisme , Canaux cationiques TRPV/métabolisme , Danio zébré , Protéines de poisson-zèbre/génétique
16.
Article de Anglais | MEDLINE | ID: mdl-25193178

RÉSUMÉ

This study investigated the effects of two rearing salinities, and acute salinity transfer, on the energetic costs of osmoregulation and the expression of metabolic and osmoregulatory genes in the gill of Mozambique tilapia. Using automated, intermittent-flow respirometry, measured standard metabolic rates (SMRs) of tilapia reared in seawater (SW, 130 mg O2 kg⁻¹ h⁻¹) were greater than those reared in fresh water (FW, 103 mg O2 kg⁻¹ h⁻¹), when normalized to a common mass of 0.05 kg and at 25±1°C. Transfer from FW to 75% SW increased SMR within 18h, to levels similar to SW-reared fish, while transfer from SW to FW decreased SMR to levels similar to FW-reared fish. Branchial gene expression of Na⁺-K⁺-2Cl⁻ cotransporter (NKCC), an indicator of SW-type mitochondria-rich (MR) cells, was positively correlated with SMR, while Na⁺-Cl⁻ cotransporter (NCC), an indicator of FW-type MR cells, was negatively correlated. Principal Components Analysis also revealed that branchial expression of cytochrome c oxidase subunit IV (COX-IV), glycogen phosphorylase (GP), and a putative mitochondrial biogenesis regulator in fish, peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α), were correlated with a higher SMR, plasma osmolality, and environmental salinity, while expression of glycogen synthase (GS), PGC-1ß, and nuclear respiratory factor 1 (NRF-1) had negative correlations. These results suggest that the energetic costs of osmoregulation are higher in SW than in FW, which may be related to the salinity-dependent differences in osmoregulatory mechanisms found in the gills of Mozambique tilapia.


Sujet(s)
Région branchiale/physiologie , Métabolisme énergétique , Régulation de l'expression des gènes au cours du développement , Osmorégulation , Stress physiologique , Tilapia/physiologie , Animaux , Aquaculture , Région branchiale/enzymologie , Région branchiale/croissance et développement , Complexe IV de la chaîne respiratoire/génétique , Complexe IV de la chaîne respiratoire/métabolisme , Protéines de poisson/génétique , Protéines de poisson/métabolisme , Eau douce , Branchies/enzymologie , Branchies/croissance et développement , Branchies/physiologie , Glycogen phosphorylase/génétique , Glycogen phosphorylase/métabolisme , Mâle , Analyse en composantes principales , Salinité , Eau de mer , Membre-1 de la famille-12 des transporteurs de solutés/génétique , Membre-1 de la famille-12 des transporteurs de solutés/métabolisme , Membre-3 de la famille-12 des transporteurs de solutés/génétique , Membre-3 de la famille-12 des transporteurs de solutés/métabolisme , Tilapia/croissance et développement , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme
17.
Biochem Biophys Res Commun ; 452(3): 655-60, 2014 Sep 26.
Article de Anglais | MEDLINE | ID: mdl-25193697

RÉSUMÉ

Cell-adhesion molecule-related/Downregulated by Oncogenes (CDO or CDON) was identified as a receptor for the classic morphogen Sonic Hedgehog (SHH). It has been shown that, in cell culture, CDO also behaves as a SHH dependence receptor: CDO actively triggers apoptosis in absence of SHH via a proteolytic cleavage in CDO intracellular domain. We present evidence that CDO is also pro-apoptotic in the developing neural tube where SHH is known to act as a survival factor. SHH, produced by the ventral foregut endoderm, was shown to promote survival of facial neural crest cells (NCCs) that colonize the first branchial arch (BA1). We show here that the survival activity of SHH on neural crest cells is due to SHH-mediated inhibition of CDO pro-apoptotic activity. Silencing of CDO rescued NCCs from apoptosis observed upon SHH inhibition in the ventral foregut endoderm. Thus, the pair SHH/dependence receptor CDO may play an important role in neural crest cell survival during the formation of the first branchial arch.


Sujet(s)
Molécules d'adhérence cellulaire/génétique , Protéines Hedgehog/génétique , Crête neurale/métabolisme , Animaux , Apoptose , Région branchiale/cytologie , Région branchiale/croissance et développement , Région branchiale/métabolisme , Molécules d'adhérence cellulaire/métabolisme , Survie cellulaire , Embryon de poulet , Endoderme/cytologie , Endoderme/croissance et développement , Endoderme/métabolisme , Régulation de l'expression des gènes au cours du développement , Cellules HEK293 , Protéines Hedgehog/métabolisme , Humains , Mésoderme/cytologie , Mésoderme/croissance et développement , Mésoderme/métabolisme , Souris , Cellules NIH 3T3 , Crête neurale/cytologie , Tube neural/cytologie , Tube neural/croissance et développement , Tube neural/métabolisme , Transduction du signal
18.
Article de Anglais | MEDLINE | ID: mdl-25152533

RÉSUMÉ

Red drum, Sciaenops ocellatus, is an estuarine-dependent fish species commonly found in the Gulf of Mexico and along the coast of the southeastern United States. This economically important species has demonstrated freshwater tolerance; however, the physiological mechanisms and costs related to freshwater exposure remain poorly understood. The current study therefore investigated the physiological response of red drum using an acute freshwater transfer protocol. Plasma osmolality, Cl⁻, Mg²âº and Ca²âº were all significantly reduced by 24h post-transfer; Cl⁻ and Mg²âº recovered to control levels by 7days post-transfer. No effect of transfer was observed on muscle water content; however, muscle Cl⁻ was significantly reduced. Interestingly, plasma and muscle Na⁺ content was unaffected by freshwater transfer. Intestinal fluid was absent by 24h post-transfer indicating cessation of drinking. Branchial gene expression analysis showed that both CFTR and NKCC1 exhibited significant down-regulation at 8 and 24h post-transfer, respectively, although transfer had no impact on NHE2, NHE3 or Na⁺, K⁺ ATPase (NKA) activity. These general findings are supported by immunohistochemical analysis, which revealed no apparent NKCC containing cells in the gills at 7days post transfer while NKA cells localization was unaffected. The results of the current study suggest that red drum can effectively regulate Na⁺ balance upon freshwater exposure using already present Na⁺ uptake pathways while also down-regulating ion excretion mechanisms.


Sujet(s)
Serran/physiologie , Région branchiale/physiologie , Protéine CFTR/métabolisme , Régulation de l'expression des gènes au cours du développement , Osmorégulation , Membre-2 de la famille-12 des transporteurs de solutés/métabolisme , Stress physiologique , Animaux , Aquaculture , Serran/sang , Serran/croissance et développement , Région branchiale/cytologie , Région branchiale/croissance et développement , Protéine CFTR/génétique , Régulation négative , Protéines de poisson/génétique , Protéines de poisson/métabolisme , Eau douce , Cinétique , Muscles squelettiques/croissance et développement , Muscles squelettiques/physiologie , Isoformes de protéines/génétique , Isoformes de protéines/métabolisme , ARN messager/métabolisme , Salinité , Antiport des ions sodium-hydrogène/génétique , Antiport des ions sodium-hydrogène/métabolisme , Sodium-Potassium-Exchanging ATPase/génétique , Sodium-Potassium-Exchanging ATPase/métabolisme , Membre-2 de la famille-12 des transporteurs de solutés/génétique , Texas
19.
PLoS Genet ; 10(7): e1004479, 2014 Jul.
Article de Anglais | MEDLINE | ID: mdl-25058015

RÉSUMÉ

Craniofacial development requires signals from epithelia to pattern skeletogenic neural crest (NC) cells, such as the subdivision of each pharyngeal arch into distinct dorsal (D) and ventral (V) elements. Wnt signaling has been implicated in many aspects of NC and craniofacial development, but its roles in D-V arch patterning remain unclear. To address this we blocked Wnt signaling in zebrafish embryos in a temporally-controlled manner, using transgenics to overexpress a dominant negative Tcf3, (dntcf3), (Tg(hsp70I:tcf3-GFP), or the canonical Wnt inhibitor dickkopf1 (dkk1), (Tg(hsp70i:dkk1-GFP) after NC migration. In dntcf3 transgenics, NC cells in the ventral arches of heat-shocked embryos show reduced proliferation, expression of ventral patterning genes (hand2, dlx3b, dlx5a, msxe), and ventral cartilage differentiation (e.g. lower jaws). These D-V patterning defects resemble the phenotypes of zebrafish embryos lacking Bmp or Edn1 signaling, and overexpression of dntcf3 dramatically reduces expression of a subset of Bmp receptors in the arches. Addition of ectopic BMP (or EDN1) protein partially rescues ventral development and expression of dlx3b, dlx5a, and msxe in Wnt signaling-deficient embryos, but surprisingly does not rescue hand2 expression. Thus Wnt signaling provides ventralizing patterning cues to arch NC cells, in part through regulation of Bmp and Edn1 signaling, but independently regulates hand2. Similarly, heat-shocked dkk1+ embryos exhibit ventral arch reductions, but also have mandibular clefts at the ventral midline not seen in dntcf3+ embryos. Dkk1 is expressed in pharyngeal endoderm, and cell transplantation experiments reveal that dntcf3 must be overexpressed in pharyngeal endoderm to disrupt D-V arch patterning, suggesting that distinct endodermal roles for Wnts and Wnt antagonists pattern the developing skeleton.


Sujet(s)
Endothéline-1/biosynthèse , Crête neurale/croissance et développement , Voie de signalisation Wnt/génétique , Protéines de poisson-zèbre/biosynthèse , Danio zébré/croissance et développement , Animaux , Animal génétiquement modifié , Plan d'organisation du corps/génétique , Protéines morphogénétiques osseuses/génétique , Région branchiale/croissance et développement , Région branchiale/métabolisme , Endothéline-1/génétique , Régulation de l'expression des gènes au cours du développement , Biosynthèse des protéines , Danio zébré/génétique , Protéines de poisson-zèbre/génétique
20.
Int J Dev Biol ; 58(10-12): 917-27, 2014.
Article de Anglais | MEDLINE | ID: mdl-26154332

RÉSUMÉ

The duality of amphibians is epitomized by their pharyngeal arch skeletons, the larval and adult morphologies of which enable very different feeding and breathing behaviors in aquatic and terrestrial life. To accomplish this duality, amphibian pharyngeal arch skeletons undergo two periods of patterning: embryogenesis and metamorphosis, and two periods of growth: larval and postmetamorphic. Their extreme ontogenetic variation, however, is coupled with relatively limited phylogenetic variation. I propose that amphibians face an evolutionary tradeoff between their ontogenetic and phylogenetic diversification that stems from the need to grow and transform the pharyngeal arch skeleton in cartilage rather than bone. Cartilage differs fundamentally from bone in its histology, function, development and growth. Cartilage is also the first skeletal tissue to form embryonically and provides more cellular pathways for shape change than bone. This article combines morphological, histological and experimental perspectives to explore how pharyngeal arch cartilage shape is controlled in amphibian embryogenesis, growth and metamorphosis, and how amphibian skeletal ontogenies are impacted by using cartilage to evolve a complex life cycle and in evolving away from a complex life cycle.


Sujet(s)
Plan d'organisation du corps/physiologie , Os et tissu osseux/embryologie , Région branchiale/embryologie , Cartilage/embryologie , Urodela/embryologie , Animaux , Évolution biologique , Développement osseux , Région branchiale/croissance et développement , Cartilage/croissance et développement , Larve/métabolisme , Métamorphose biologique/physiologie , Urodela/anatomie et histologie , Urodela/croissance et développement
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...