Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Differentiation ; 138: 100782, 2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-38810379

RESUMEN

The mandible is composed of several musculoskeletal tissues including bone, cartilage, and tendon that require precise patterning to ensure structural and functional integrity. Interestingly, most of these tissues are derived from one multipotent cell population called cranial neural crest cells (CNCCs). How CNCCs are properly instructed to differentiate into various tissue types remains nebulous. To better understand the mechanisms necessary for the patterning of mandibular musculoskeletal tissues we utilized the avian mutant talpid2 (ta2) which presents with several malformations of the facial skeleton including dysplastic tendons, mispatterned musculature, and bilateral ectopic cartilaginous processes extending off Meckel's cartilage. We found an ectopic epithelial BMP signaling domain in the ta2 mandibular prominence (MNP) that correlated with the subsequent expansion of SOX9+ cartilage precursors. These findings were validated with conditional murine models suggesting an evolutionarily conserved mechanism for CNCC-derived musculoskeletal patterning. Collectively, these data support a model in which cilia are required to define epithelial signal centers essential for proper musculoskeletal patterning of CNCC-derived mesenchyme.

2.
Am J Med Genet A ; : e63615, 2024 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-38563316

RESUMEN

The Society for Craniofacial Genetics and Developmental Biology (SCGDB) held its 46th Annual Meeting at Cincinnati Children's Hospital Medical Center in Cincinnati, Ohio on October 10th-12th, 2023. On the first day of the meeting, Drs. Sally Moody and Justin Cotney were each honored with the SCGDB Distinguished Scientist Awards for their exceptional contributions to the field of craniofacial biology. The following two days of the meeting featured five sessions that highlighted new discoveries in signaling and genomic mechanisms regulating craniofacial development, human genetics, translational and regenerative approaches, and clinical management of craniofacial differences. Interactive workshops on spatial transcriptomics and scientific communication, as well as a poster session facilitated meaningful interactions among the 122 attendees representing diverse career stages and research backgrounds in developmental biology and genetics, strengthened the SCGDB community.

3.
bioRxiv ; 2024 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-38645005

RESUMEN

Cleft lip and cleft palate are among the most common congenital anomalies and are the result of incomplete fusion of embryonic craniofacial processes or palatal shelves, respectively. We know that genetics play a large role in these anomalies but the list of known causal genes is far from complete. As part of a larger sequencing effort of patients with micrognathia and cleft palate we identified a candidate variant in transforming growth factor beta receptor 2 (TGFBR2) which is rare, changing a highly conserved amino acid, and predicted to be pathogenic by a number of metrics. The family history and population genetics would suggest this specific variant would be incompletely penetrant, but this gene has been convincingly implicated in craniofacial development. In order to test the hypothesis this might be a causal variant, we used genome editing to create the orthologous variant in a new mouse model. Surprisingly, Tgfbr2V387M mice did not exhibit craniofacial anomalies or have reduced survival suggesting this is, in fact, not a causal variant for cleft palate/ micrognathia. The discrepancy between in silico predictions and mouse phenotypes highlights the complexity of translating human genetic findings to mouse models. We expect these findings will aid in interpretation of future variants seen in TGFBR2 from ongoing sequencing of patients with congenital craniofacial anomalies.

4.
Nat Commun ; 15(1): 136, 2024 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-38167838

RESUMEN

Craniofacial abnormalities account for approximately one third of birth defects. The regulatory programs that build the face require precisely controlled spatiotemporal gene expression, achieved through tissue-specific enhancers. Clusters of coactivated enhancers and their target genes, known as superenhancers, are important in determining cell identity but have been largely unexplored in development. In this study we identified superenhancer regions unique to human embryonic craniofacial tissue. To demonstrate the importance of such regions in craniofacial development and disease, we focused on an ~600 kb noncoding region located between NPVF and NFE2L3. We identified long range interactions with this region in both human and mouse embryonic craniofacial tissue with the anterior portion of the HOXA gene cluster. Mice lacking this superenhancer exhibit perinatal lethality, and present with highly penetrant skull defects and orofacial clefts phenocopying Hoxa2-/- mice. Moreover, we identified two cases of de novo copy number changes of the superenhancer in humans both with severe craniofacial abnormalities. This evidence suggests we have identified a critical noncoding locus control region that specifically regulates anterior HOXA genes and copy number changes are pathogenic in human patients.


Asunto(s)
Labio Leporino , Fisura del Paladar , Embarazo , Femenino , Humanos , Ratones , Animales , Labio Leporino/genética , Regulación del Desarrollo de la Expresión Génica , Fisura del Paladar/genética , Genes Homeobox , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética
5.
bioRxiv ; 2023 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-37904917

RESUMEN

Bacterial artificial chromosome transgenic models, including most Cre-recombinases, enable potent interrogation of gene function in vivo but require rigorous validation as limitations emerge. Due to its high relevance to metabolic studies, we performed comprehensive analysis of the Ucp1-CreEvdr line which is widely used for brown fat research. Hemizygotes exhibited major brown and white fat transcriptomic dysregulation, indicating potential altered tissue function. Ucp1-CreEvdr homozygotes also show high mortality, growth defects, and craniofacial abnormalities. Mapping the transgene insertion site revealed insertion in chromosome 1 accompanied by large genomic alterations disrupting several genes expressed in a range of tissues. Notably, Ucp1-CreEvdr transgene retains an extra Ucp1 gene copy that may be highly expressed under high thermogenic burden. Our multi-faceted analysis highlights a complex phenotype arising from the presence of the Ucp1-CreEvdr transgene independently of the intended genetic manipulations. Overall, comprehensive validation of transgenic mice is imperative to maximize discovery while mitigating unexpected, off-target effects.

6.
Development ; 150(8)2023 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-36971348

RESUMEN

Primary cilia are nearly ubiquitous organelles that transduce molecular and mechanical signals. Although the basic structure of the cilium and the cadre of genes that contribute to ciliary formation and function (the ciliome) are believed to be evolutionarily conserved, the presentation of ciliopathies with narrow, tissue-specific phenotypes and distinct molecular readouts suggests that an unappreciated heterogeneity exists within this organelle. Here, we provide a searchable transcriptomic resource for a curated primary ciliome, detailing various subgroups of differentially expressed genes within the ciliome that display tissue and temporal specificity. Genes within the differentially expressed ciliome exhibited a lower level of functional constraint across species, suggesting organism and cell-specific function adaptation. The biological relevance of ciliary heterogeneity was functionally validated by using Cas9 gene-editing to disrupt ciliary genes that displayed dynamic gene expression profiles during osteogenic differentiation of multipotent neural crest cells. Collectively, this novel primary cilia-focused resource will allow researchers to explore longstanding questions related to how tissue and cell-type specific functions and ciliary heterogeneity may contribute to the range of phenotypes associated with ciliopathies.


Asunto(s)
Ciliopatías , Osteogénesis , Humanos , Cilios/genética , Cilios/metabolismo , Ciliopatías/genética , Desarrollo Embrionario/genética , Diferenciación Celular/genética
7.
Dis Model Mech ; 15(8)2022 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-35818799

RESUMEN

Ciliopathies represent a disease class characterized by a broad range of phenotypes including polycystic kidneys and skeletal anomalies. Ciliopathic skeletal phenotypes are among the most common and most difficult to treat due to a poor understanding of the pathological mechanisms leading to disease. Using an avian model (talpid2) for a human ciliopathy with both kidney and skeletal anomalies (orofaciodigital syndrome 14), we identified disruptions in the FGF23-PTH axis that resulted in reduced calcium uptake in the developing mandible and subsequent micrognathia. Although pharmacological intervention with the U.S. Food and Drug Administration (FDA)-approved pan-FGFR inhibitor AZD4547 alone rescued expression of the FGF target SPRY2, it did not significantly rescue micrognathia. In contrast, treatment with a cocktail of AZD4547 and teriparatide acetate, a PTH agonist and FDA-approved treatment for osteoporosis, resulted in molecular, cellular and phenotypic rescue of ciliopathic micrognathia in talpid2 mutants. Together, these data provide novel insight into pathological molecular mechanisms associated with ciliopathic skeletal phenotypes and a potential therapeutic strategy for a pleiotropic disease class with limited to no treatment options.


Asunto(s)
Ciliopatías , Micrognatismo , Cilios/metabolismo , Ciliopatías/tratamiento farmacológico , Ciliopatías/genética , Ciliopatías/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/metabolismo , Micrognatismo/metabolismo , Micrognatismo/patología , Fenotipo , Proteínas/metabolismo
8.
Am J Med Genet A ; 188(7): 2258-2266, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35352468

RESUMEN

The Society for Craniofacial Genetics and Developmental Biology (SCGDB) held its 44th Annual Meeting in a virtual format on October 18-19, 2021. The SCGDB meeting included presentation of the SCGDB Distinguished Scientists in Craniofacial Research Awards to Drs. Paul Trainor and Jeff Bush and four scientific sessions on the genomics of craniofacial development, craniofacial morphogenesis and regeneration, translational craniofacial biology and signaling during craniofacial development. The meeting also included workshops on professional development for faculty and trainees, National Institutes of Health (NIH)/National Institute of Craniofacial and Dental Research funding and usage of Genomics Software, as well as two poster sessions. An exhibitor booth run by FaceBase was also present to facilitate the upload and download of datasets relevant to the craniofacial community. Over 200 attendees from 12 countries and 23 states, representing over 80 different scientific institutions, participated. This diverse group of scientists included cell biologists, developmental biologists, and clinical geneticists. Although the continuing COVID-19 pandemic forced a virtual meeting format for a second year in a row, the meeting platform provided ample opportunities for participant interactions and discussions, thus strengthening the community.


Asunto(s)
COVID-19 , Pandemias , Biología Evolutiva , Genómica , Humanos , Programas Informáticos , Estados Unidos
9.
Nat Commun ; 12(1): 6469, 2021 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-34753942

RESUMEN

Subunit switches in the BAF chromatin remodeler are essential during development. ARID1B and its paralog ARID1A encode for mutually exclusive BAF subunits. De novo ARID1B haploinsufficient mutations cause neurodevelopmental disorders, including Coffin-Siris syndrome, which is characterized by neurological and craniofacial features. Here, we leveraged ARID1B+/- Coffin-Siris patient-derived iPSCs and modeled cranial neural crest cell (CNCC) formation. We discovered that ARID1B is active only during the first stage of this process, coinciding with neuroectoderm specification, where it is part of a lineage-specific BAF configuration (ARID1B-BAF). ARID1B-BAF regulates exit from pluripotency and lineage commitment by attenuating thousands of enhancers and genes of the NANOG and SOX2 networks. In iPSCs, these enhancers are maintained active by ARID1A-containing BAF. At the onset of differentiation, cells transition from ARID1A- to ARID1B-BAF, eliciting attenuation of the NANOG/SOX2 networks and triggering pluripotency exit. Coffin-Siris patient cells fail to perform the ARID1A/ARID1B switch, and maintain ARID1A-BAF at the pluripotency enhancers throughout all stages of CNCC formation. This leads to persistent NANOG/SOX2 activity which impairs CNCC formation. Despite showing the typical neural crest signature (TFAP2A/SOX9-positive), ARID1B-haploinsufficient CNCCs are also aberrantly NANOG-positive. These findings suggest a connection between ARID1B mutations, neuroectoderm specification and a pathogenic mechanism for Coffin-Siris syndrome.


Asunto(s)
Cromatina/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteína Homeótica Nanog/metabolismo , Cresta Neural/metabolismo , Factores de Transcripción/metabolismo , Western Blotting , Proteínas de Unión al ADN/genética , Citometría de Flujo , Células HEK293 , Humanos , Mutación/genética , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Factores de Transcripción/genética
10.
Front Cell Dev Biol ; 9: 647391, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34211969

RESUMEN

The primary cilium is a ubiquitous, microtubule-based cellular organelle. Primary cilia dysfunction results in a group of disorders termed ciliopathies. C2 domain containing 3 centriole elongation regulator (C2cd3), encodes a centriolar protein essential for ciliogenesis. Mutations in human C2CD3 are associated with the human ciliopathy Oral-Facial-Digital syndrome type 14 (OFD14). In order to better understand the etiology of ciliopathies including OFD14, we generated numerous murine models targeting C2cd3. Initial analysis revealed several tissue-specific isoforms of C2cd3, and while the loss of C2cd3 has previously been reported to result in exencephaly, tight mesencephalic flexure, pericardial edema, abnormal heart looping and a twisted body axis, further analysis revealed that genetic background may also contribute to phenotypic variation. Additional analyses of a conditional allelic series targeting C-terminal PKC-C2 domains or the N-terminal C2CD3N-C2 domain of C2cd3 revealed a variable degree of phenotypic severity, suggesting that while the N-terminal C2CD3N-C2 domain was critical for early embryonic development as a whole, there was also a craniofacial specific role for the C2CD3N-C2 domains. Together, through generation of novel models and evaluation of C2cd3 expression, these data provide valuable insight into mechanisms of pathology for craniofacial ciliopathies that can be further explored in the future.

11.
J Dev Biol ; 9(2)2021 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-33805906

RESUMEN

Primary cilia are ubiquitous microtubule-based organelles that serve as signaling hubs for numerous developmental pathways, most notably the Hedgehog (Hh) pathway. Defects in the structure or function of primary cilia result in a class of diseases called ciliopathies. It is well known that primary cilia participate in transducing a Hh signal, and as such ciliopathies frequently present with phenotypes indicative of aberrant Hh function. Interestingly, the exact mechanisms of cilia-dependent Hh signaling transduction are unclear as some ciliopathic animal models simultaneously present with gain-of-Hh phenotypes in one organ system and loss-of-Hh phenotypes in another. To better understand how Hh signaling is perturbed across different tissues in ciliopathic conditions, we examined four distinct Hh-dependent signaling centers in the naturally occurring avian ciliopathic mutant talpid2 (ta2). In addition to the well-known and previously reported limb and craniofacial malformations, we observed dorsal-ventral patterning defects in the neural tube, and a shortened gastrointestinal tract. Molecular analyses for elements of the Hh pathway revealed that the loss of cilia impact transduction of an Hh signal in a tissue-specific manner at variable levels of the pathway. These studies will provide increased knowledge into how impaired ciliogenesis differentially regulates Hh signaling across tissues and will provide potential avenues for future targeted therapeutic treatments.

12.
Dev Dyn ; 250(9): 1358-1367, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33605505

RESUMEN

BACKGROUND: The naturally occurring chicken mutant talpid2 (ta2 ), best known for its limb and craniofacial defects, has long served as a valuable tool for developmental biologists studying growth and patterning of craniofacial structures and the limb. The mutant provides a unique tool to examine the molecular and cellular processes regulating limb development. RESULTS: This mutant also provides unique insights into the evolution of developmental genetic programs. Previous work defined the appearance of atavistic dentition in ta2 embryos. Herein we describe the appearance of ancestral characters of the hindlimb in embryonic ta2 chicken embryos. CONCLUSION: As the ta2 phenotype arises as a result of mutation in C2CD3 and disrupted cilia function, this mutant provides genetic and developmental insight into the causes of asymmetry in the limb and also a model for the evolution of the avian hindlimb.


Asunto(s)
Cilios , Extremidades , Animales , Embrión de Pollo , Pollos/genética , Miembro Posterior , Organogénesis
13.
Development ; 148(4)2021 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-33589509

RESUMEN

Ciliopathies represent a growing class of diseases caused by defects in microtubule-based organelles called primary cilia. Approximately 30% of ciliopathies are characterized by craniofacial phenotypes such as craniosynostosis, cleft lip/palate and micrognathia. Patients with ciliopathic micrognathia experience a particular set of difficulties, including impaired feeding and breathing, and have extremely limited treatment options. To understand the cellular and molecular basis for ciliopathic micrognathia, we used the talpid2 (ta2 ), a bona fide avian model for the human ciliopathy oral-facial-digital syndrome subtype 14. Histological analyses revealed that the onset of ciliopathic micrognathia in ta2 embryos occurred at the earliest stages of mandibular development. Neural crest-derived skeletal progenitor cells were particularly sensitive to a ciliopathic insult, undergoing unchecked passage through the cell cycle and subsequent increased proliferation. Furthermore, whereas neural crest-derived skeletal differentiation was initiated, osteoblast maturation failed to progress to completion. Additional molecular analyses revealed that an imbalance in the ratio of bone deposition and resorption also contributed to ciliopathic micrognathia in ta2 embryos. Thus, our results suggest that ciliopathic micrognathia is a consequence of multiple aberrant cellular processes necessary for skeletal development, and provide potential avenues for future therapeutic treatments.


Asunto(s)
Remodelación Ósea , Ciliopatías/etiología , Micrognatismo/etiología , Organogénesis , Fenotipo , Animales , Remodelación Ósea/genética , Resorción Ósea , Ciclo Celular/genética , Ciliopatías/diagnóstico , Anomalías Craneofaciales/genética , Susceptibilidad a Enfermedades , Embrión no Mamífero , Regulación del Desarrollo de la Expresión Génica , Estudios de Asociación Genética , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Micrognatismo/diagnóstico , Organogénesis/genética , Osteoblastos/metabolismo , Proteína con Dedos de Zinc GLI1/genética , Proteína con Dedos de Zinc GLI1/metabolismo
14.
Elife ; 92020 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-33006313

RESUMEN

Despite a common understanding that Gli TFs are utilized to convey a Hh morphogen gradient, genetic analyses suggest craniofacial development does not completely fit this paradigm. Using the mouse model (Mus musculus), we demonstrated that rather than being driven by a Hh threshold, robust Gli3 transcriptional activity during skeletal and glossal development required interaction with the basic helix-loop-helix TF Hand2. Not only did genetic and expression data support a co-factorial relationship, but genomic analysis revealed that Gli3 and Hand2 were enriched at regulatory elements for genes essential for mandibular patterning and development. Interestingly, motif analysis at sites co-occupied by Gli3 and Hand2 uncovered mandibular-specific, low-affinity, 'divergent' Gli-binding motifs (dGBMs). Functional validation revealed these dGBMs conveyed synergistic activation of Gli targets essential for mandibular patterning and development. In summary, this work elucidates a novel, sequence-dependent mechanism for Gli transcriptional activity within the craniofacial complex that is independent of a graded Hh signal.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Desarrollo Maxilofacial , Ratones/genética , Proteínas del Tejido Nervioso/genética , Cráneo/crecimiento & desarrollo , Proteína Gli3 con Dedos de Zinc/genética , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Femenino , Masculino , Desarrollo Maxilofacial/genética , Ratones/metabolismo , Modelos Animales , Proteínas del Tejido Nervioso/metabolismo , Cráneo/metabolismo , Proteína Gli3 con Dedos de Zinc/metabolismo
15.
Dev Biol ; 447(1): 28-41, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29548942

RESUMEN

Molecular signals are the guiding force of development, imparting direction upon cells to divide, migrate, differentiate, etc. The mechanisms by which a cell can receive and transduce these signals into measurable actions remains a 'black box' in developmental biology. Primary cilia are ubiquitous, microtubule-based organelles that dynamically extend from a cell to receive and process molecular and mechanical signaling cues. In the last decade, this organelle has become increasingly intriguing to the research community due to its ability to act as a cellular antenna, receive and transduce molecular stimuli, and initiate a cellular response. In this review, we discuss the structure of primary cilia, emphasizing how the ciliary components contribute to the transduction of signaling pathways. Furthermore, we address how the cilium integrates these signals and conveys them into cellular processes such as proliferation, migration and tissue patterning. Gaining a deeper understanding of the mechanisms used by primary cilia to receive and integrate molecular signals is essential, as it opens the door for the identification of therapeutic targets within the cilium that could alleviate pathological conditions brought on by aberrant molecular signaling.


Asunto(s)
Cilios , Transducción de Señal , Animales , Cilios/metabolismo , Cilios/patología , Humanos
17.
Development ; 145(15)2018 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-29986869

RESUMEN

In mammals, the epithelial tissues of major salivary glands generate saliva and drain it into the oral cavity. For submandibular salivary glands (SMGs), the epithelial tissues arise during embryogenesis from naïve oral ectoderm adjacent to the base of the tongue, which begins to thicken, express SOX9 and invaginate into underlying mesenchyme. The developmental mechanisms initiating salivary gland development remain unexplored. In this study, we show that retinoic acid (RA) signaling activity at the site of gland initiation is colocalized with expression of retinol metabolic genes Rdh10 and Aldh1a2 in the underlying SMG mesenchyme. Utilizing a novel ex vivo assay for SMG initiation developed for this study, we show that RDH10 and RA are required for salivary gland initiation. Moreover, we show that the requirement for RA in gland initiation involves canonical signaling through retinoic acid receptors (RAR). Finally, we show that RA signaling essential for gland initiation is transduced specifically through RARα, with no contribution from other RAR isoforms. This is the first study to identify a molecular signal regulating mammalian salivary gland initiation.


Asunto(s)
Oxidorreductasas de Alcohol/fisiología , Receptores de Ácido Retinoico/metabolismo , Glándulas Salivales/embriología , Glándula Submandibular/embriología , Tretinoina/metabolismo , Vitamina A/metabolismo , Oxidorreductasas de Alcohol/genética , Animales , Embrión de Mamíferos , Desarrollo Embrionario/efectos de los fármacos , Desarrollo Embrionario/genética , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Embarazo , Receptores de Ácido Retinoico/genética , Glándulas Salivales/efectos de los fármacos , Glándulas Salivales/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Glándula Submandibular/efectos de los fármacos , Glándula Submandibular/metabolismo , Tretinoina/farmacología
18.
Dev Dyn ; 247(6): 818-831, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29532549

RESUMEN

BACKGROUND: Submandibular glands (SMGs) are specialized epithelial structures which generate saliva necessary for mastication and digestion. Loss of SMGs can lead to inflammation, oral lesions, fungal infections, problems with chewing/swallowing, and tooth decay. Understanding the development of the SMG is important for developing therapeutic options for patients with impaired SMG function. Recent studies have suggested Sonic hedgehog (Shh) signaling in the epithelium plays an integral role in SMG development; however, the mechanism by which Shh influences gland development remains nebulous. RESULTS: Using the Kif3af/f ;Wnt1-Cre ciliopathic mouse model to prevent Shh signal transduction by means of the loss of primary cilia in neural crest cells, we report that mesenchymal Shh activity is necessary for gland development. Furthermore, using a variety of murine transgenic lines with aberrant mesenchymal Shh signal transduction, we determine that loss of Shh activity, by means of loss of the Gli activator, rather than gain of Gli repressor, is sufficient to cause the SMG aplasia. Finally, we determine that loss of the SMG correlates with reduced Neuregulin1 (Nrg1) expression and lack of innervation of the SMG epithelium. CONCLUSIONS: Together, these data suggest a novel mechanistic role for mesenchymal Shh signaling during SMG development. Developmental Dynamics 247:818-831, 2018. © 2018 Wiley Periodicals, Inc.


Asunto(s)
Cilios/metabolismo , Peces/embriología , Peces/metabolismo , Proteínas Hedgehog/metabolismo , Glándula Submandibular/embriología , Glándula Submandibular/metabolismo , Animales , Regulación del Desarrollo de la Expresión Génica/genética , Regulación del Desarrollo de la Expresión Génica/fisiología , Organogénesis/genética , Organogénesis/fisiología , Transducción de Señal/fisiología
19.
Dev Biol ; 431(2): 168-178, 2017 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-28941984

RESUMEN

Development of the brain directly influences the development of the face via both physical growth and Sonic hedgehog (SHH) activity; however, little is known about how neural crest cells (NCCs), the mesenchymal population that comprise the developing facial prominences, influence the development of the brain. We utilized the conditional ciliary mutant Wnt1-Cre;Kif3afl/fl to demonstrate that loss of primary cilia on NCCs resulted in a widened ventral forebrain. We found that neuroectodermal Shh expression, dorsal/ventral patterning, and amount of proliferation in the ventral neuroectoderm was not changed in Wnt1-Cre;Kif3afl/fl mutants; however, tissue polarity and directional cell division were disrupted. Furthermore, NCCs of Wnt1-Cre;Kif3afl/fl mutants failed to respond to a SHH signal emanating from the ventral forebrain. We were able to recapitulate the ventral forebrain phenotype by removing Smoothened from NCCs (Wnt1-Cre;Smofl/fl) indicating that changes in the ventral forebrain were mediated through a Hedgehog-dependent mechanism. Together, these data suggest a novel, cilia-dependent mechanism for NCCs during forebrain development.


Asunto(s)
División Celular , Cilios/metabolismo , Proteínas Hedgehog/metabolismo , Morfogénesis , Cresta Neural/citología , Prosencéfalo/citología , Prosencéfalo/embriología , Animales , Tipificación del Cuerpo/genética , Polaridad Celular , Cara/embriología , Regulación del Desarrollo de la Expresión Génica , Integrasas/metabolismo , Cinesinas/metabolismo , Ratones , Modelos Biológicos , Morfogénesis/genética , Mutación/genética , Cresta Neural/metabolismo , Placa Neural/citología , Placa Neural/embriología , Placa Neural/metabolismo , Fenotipo , Prosencéfalo/metabolismo , Recombinación Genética/genética , Factores de Transcripción SOXE/metabolismo , Telencéfalo/embriología , Telencéfalo/metabolismo , Proteínas Wnt/metabolismo
20.
PLoS One ; 12(3): e0174206, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28346501

RESUMEN

Primary cilia are nearly ubiquitous, cellular projections that function to transduce molecular signals during development. Loss of functional primary cilia has a particularly profound effect on the developing craniofacial complex, causing several anomalies including craniosynostosis, micrognathia, midfacial dysplasia, cleft lip/palate and oral/dental defects. Development of the craniofacial complex is an intricate process that requires interactions between several different tissues including neural crest cells, neuroectoderm and surface ectoderm. To understand the tissue-specific requirements for primary cilia during craniofacial development we conditionally deleted three separate intraflagellar transport genes, Kif3a, Ift88 and Ttc21b with three distinct drivers, Wnt1-Cre, Crect and AP2-Cre which drive recombination in neural crest, surface ectoderm alone, and neural crest, surface ectoderm and neuroectoderm, respectively. We found that tissue-specific conditional loss of ciliary genes with different functions produces profoundly different facial phenotypes. Furthermore, analysis of basic cellular behaviors in these mutants suggests that loss of primary cilia in a distinct tissue has unique effects on development of adjacent tissues. Together, these data suggest specific spatiotemporal roles for intraflagellar transport genes and the primary cilium during craniofacial development.


Asunto(s)
Anomalías Craneofaciales/genética , Cara/embriología , Regulación del Desarrollo de la Expresión Génica , Cráneo/embriología , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Cilios/genética , Cara/anomalías , Femenino , Eliminación de Gen , Cinesinas/genética , Masculino , Ratones , Cresta Neural/embriología , Cresta Neural/metabolismo , Placa Neural/embriología , Placa Neural/metabolismo , Cráneo/anomalías , Cráneo/metabolismo , Proteínas Supresoras de Tumor/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA