Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
1.
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
2.
Differentiation ; 138: 100782, 2024.
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.


Asunto(s)
Mandíbula , Cresta Neural , Animales , Cresta Neural/citología , Cresta Neural/metabolismo , Ratones , Mandíbula/crecimiento & desarrollo , Mandíbula/metabolismo , Tipificación del Cuerpo/genética , Cartílago/metabolismo , Cartílago/crecimiento & desarrollo , Cartílago/citología , Cilios/metabolismo , Cilios/genética , Mesodermo/citología , Mesodermo/metabolismo , Mesodermo/crecimiento & desarrollo , Regulación del Desarrollo de la Expresión Génica , Proteínas Aviares/genética , Proteínas Aviares/metabolismo , Transducción de Señal , Diferenciación Celular , Embrión de Pollo , Pollos/genética , Factor de Transcripción SOX9/metabolismo , Factor de Transcripción SOX9/genética
3.
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
4.
Am J Med Genet A ; 194(8): e63615, 2024 Aug.
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.


Asunto(s)
Anomalías Craneofaciales , Biología Evolutiva , Humanos , Anomalías Craneofaciales/genética , Anomalías Craneofaciales/patología , Distinciones y Premios
5.
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
6.
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
7.
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
8.
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
9.
PLoS Genet ; 12(11): e1006351, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27802276

RESUMEN

Ciliopathies represent a broad class of disorders that affect multiple organ systems. The craniofacial complex is among those most severely affected when primary cilia are not functional. We previously reported that loss of primary cilia on cranial neural crest cells, via a conditional knockout of the intraflagellar transport protein KIF3a, resulted in midfacial widening due to a gain of Hedgehog (HH) activity. Here, we examine the molecular mechanism of how a loss of primary cilia can produce facial phenotypes associated with a gain of HH function. We show that loss of intraflagellar transport proteins (KIF3a or IFT88) caused aberrant GLI processing such that the amount of GLI3FL and GLI2FL was increased, thus skewing the ratio of GLIFL to GLIR in favor of the FL isoform. Genetic addition of GLI3R partially rescued the ciliopathic midfacial widening. Interestingly, despite several previous studies suggesting midfacial development relies heavily on GLI3R activity, the conditional loss of GLI3 alone did not reproduce the ciliopathic phenotype. Only the combined loss of both GLI2 and GLI3 was able to phenocopy the ciliopathic midfacial appearance. Our findings suggest that ciliopathic facial phenotypes are generated via loss of both GLI3R and GLI2R and that this pathology occurs via a de-repression mechanism. Furthermore, these studies suggest a novel role for GLI2R in craniofacial development.


Asunto(s)
Cilios/genética , Ciliopatías/genética , Cara/embriología , Factores de Transcripción de Tipo Kruppel/genética , Proteínas del Tejido Nervioso/genética , Animales , Cilios/patología , Ciliopatías/patología , Cara/patología , Regulación del Desarrollo de la Expresión Génica , Cinesinas/genética , Ratones , Ratones Transgénicos , Fenotipo , Isoformas de Proteínas/genética , Modificación Traduccional de las Proteínas/genética , Transducción de Señal/genética , Proteína Gli2 con Dedos de Zinc , Proteína Gli3 con Dedos de Zinc
10.
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
11.
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
12.
Dev Biol ; 424(2): 124-137, 2017 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-28286175

RESUMEN

Ciliopathies are a class of diseases caused by the loss of a ubiquitous, microtubule-based organelle called a primary cilium. Ciliopathies commonly result in defective development of the craniofacial complex, causing midfacial defects, craniosynostosis, micrognathia and aglossia. Herein, we explored how the conditional loss of primary cilia on neural crest cells (Kif3af/f;Wnt1-Cre) generated aglossia. On a cellular level, our data revealed that aglossia in Kif3af/f;Wnt1-Cre embryos was due to a loss of mesoderm-derived muscle precursors migrating into and surviving in the tongue anlage. To determine the molecular basis for this phenotype, we performed RNA-seq, in situ hybridization, qPCR and Western blot analyses. We found that transduction of the Sonic hedgehog (Shh) pathway, rather than other pathways previously implicated in tongue development, was aberrant in Kif3af/f;Wnt1-Cre embryos. Despite increased production of full-length GLI2 and GLI3 isoforms, previously identified GLI targets important for mandibular and glossal development (Foxf1, Foxf2, Foxd1 and Foxd2) were transcriptionally downregulated in Kif3af/f;Wnt1-Cre embryos. Genetic removal of GLI activator (GLIA) isoforms in neural crest cells recapitulated the aglossia phenotype and downregulated Fox gene expression. Genetic addition of GLIA isoforms in neural crest cells partially rescued the aglossia phenotype and Fox gene expression in Kif3af/f;Wnt1-Cre embryos. Together, our data suggested that glossal development requires primary cilia-dependent GLIA activity in neural crest cells. Furthermore, these data, in conjunction with our previous work, suggested prominence specific roles for GLI isoforms; with development of the frontonasal prominence relying heavily on the repressor isoform and the development of the mandibular prominence/tongue relying heavily on the activator isoform.


Asunto(s)
Cilios/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Cresta Neural/metabolismo , Lengua/embriología , Lengua/metabolismo , Animales , Apoptosis , Movimiento Celular , Eliminación de Gen , Proteínas Hedgehog/metabolismo , Integrasas/metabolismo , Cinesinas , Mandíbula/embriología , Mandíbula/metabolismo , Mesodermo/patología , Ratones , Modelos Biológicos , Músculos/patología , Mutación/genética , Cresta Neural/patología , Organogénesis , Fenotipo , Transducción de Señal , Células Madre/patología , Proteína Wnt1/metabolismo , Proteína Gli2 con Dedos de Zinc , Proteína Gli3 con Dedos de Zinc
13.
Dev Biol ; 415(2): 326-337, 2016 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-26597494

RESUMEN

The chicken has been a particularly useful model for the study of craniofacial development and disease for over a century due to their relatively large size, accessibility, and amenability for classical bead implantation and transplant experiments. Several naturally occurring mutant lines with craniofacial anomalies also exist and have been heavily utilized by developmental biologist for several decades. Two of the most well known lines, talpid(2) (ta(2)) and talpid(3) (ta(3)), represent the first spontaneous mutants to have the causative genes identified. Despite having distinct genetic causes, both mutants have recently been identified as ciliopathic. Excitingly, both of these mutants have been classified as models for human craniofacial ciliopathies: Oral-facial-digital syndrome (ta(2)) and Joubert syndrome (ta(3)). Herein, we review and compare these two models of craniofacial disease and highlight what they have revealed about the molecular and cellular etiology of ciliopathies. Furthermore, we outline how applying classical avian experiments and new technological advances (transgenics and genome editing) with naturally occurring avian mutants can add a tremendous amount to what we currently know about craniofacial ciliopathies.


Asunto(s)
Pollos/genética , Ciliopatías/genética , Anomalías Craneofaciales/genética , Modelos Animales de Enfermedad , Desarrollo Maxilofacial/genética , Anomalías Múltiples/genética , Anomalías Múltiples/metabolismo , Animales , Animales Modificados Genéticamente , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/fisiología , Cerebelo/anomalías , Cerebelo/metabolismo , Embrión de Pollo , Ciliopatías/embriología , Ciliopatías/veterinaria , Anomalías Craneofaciales/embriología , Anomalías Craneofaciales/veterinaria , Anomalías del Ojo/genética , Anomalías del Ojo/metabolismo , Genes Letales , Estudios de Asociación Genética , Humanos , Enfermedades Renales Quísticas/genética , Enfermedades Renales Quísticas/metabolismo , Ratones , Mutación , Síndromes Orofaciodigitales/embriología , Síndromes Orofaciodigitales/genética , Polidactilia/genética , Polidactilia/veterinaria , Enfermedades de las Aves de Corral/embriología , Enfermedades de las Aves de Corral/genética , Retina/anomalías , Retina/metabolismo
14.
Hum Mol Genet ; 24(12): 3399-409, 2015 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-25759469

RESUMEN

Autosomal dominant omodysplasia is a rare skeletal dysplasia characterized by short humeri, radial head dislocation, short first metacarpals, facial dysmorphism and genitourinary anomalies. We performed next-generation whole-exome sequencing and comparative analysis of a proband with omodysplasia, her unaffected parents and her affected daughter. We identified a de novo mutation in FRIZZLED2 (FZD2) in the proband and her daughter that was not found in unaffected family members. The FZD2 mutation (c.1644G>A) changes a tryptophan residue at amino acid 548 to a premature stop (p.Trp548*). This altered protein is still produced in vitro, but we show reduced ability of this mutant form of FZD2 to interact with its downstream target DISHEVELLED. Furthermore, expressing the mutant form of FZD2 in vitro is not able to facilitate the cellular response to canonical Wnt signaling like wild-type FZD2. We therefore conclude that the FRIZZLED2 mutation is a de novo, novel cause for autosomal dominant omodysplasia.


Asunto(s)
Receptores Frizzled/genética , Receptores Frizzled/metabolismo , Húmero/anomalías , Huesos del Metacarpo/anomalías , Mutación , Osteocondrodisplasias/genética , Osteocondrodisplasias/metabolismo , Vía de Señalización Wnt , Adulto , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Huesos/diagnóstico por imagen , Huesos/patología , Análisis Mutacional de ADN , Exoma , Facies , Femenino , Receptores Frizzled/química , Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Húmero/metabolismo , Lactante , Huesos del Metacarpo/metabolismo , Osteocondrodisplasias/diagnóstico , Linaje , Fenotipo , Unión Proteica , Transporte de Proteínas , Radiografía
15.
Development ; 141(15): 3003-12, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25053433

RESUMEN

talpid(2) is an avian autosomal recessive mutant with a myriad of congenital malformations, including polydactyly and facial clefting. Although phenotypically similar to talpid(3), talpid(2) has a distinct facial phenotype and an unknown cellular, molecular and genetic basis. We set out to determine the etiology of the craniofacial phenotype of this mutant. We confirmed that primary cilia were disrupted in talpid(2) mutants. Molecularly, we found disruptions in Hedgehog signaling. Post-translational processing of GLI2 and GLI3 was aberrant in the developing facial prominences. Although both GLI2 and GLI3 processing were disrupted in talpid(2) mutants, only GLI3 activator levels were significantly altered in the nucleus. Through additional fine mapping and whole-genome sequencing, we determined that the talpid(2) phenotype was linked to a 1.4 Mb region on GGA1q that contained the gene encoding the ciliary protein C2CD3. We cloned the avian ortholog of C2CD3 and found its expression was ubiquitous, but most robust in the developing limbs and facial prominences. Furthermore, we found that C2CD3 is localized proximal to the ciliary axoneme and is important for docking the mother centriole to the ciliary vesicle and cell membrane. Finally, we identified a 19 bp deletion in talpid(2) C2CD3 that produces a premature stop codon, and thus a truncated protein, as the likely causal allele for the phenotype. Together, these data provide insight into the cellular, molecular and genetic etiology of the talpid(2) phenotype. Our data suggest that, although the talpid(2) and talpid(3) mutations affect a common ciliogenesis pathway, they are caused by mutations in different ciliary proteins that result in differences in craniofacial phenotype.


Asunto(s)
Anomalías Craneofaciales/genética , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Factores de Transcripción de Tipo Kruppel/genética , Mutación , Alelos , Animales , Membrana Celular/metabolismo , Núcleo Celular , Centriolos/metabolismo , Embrión de Pollo , Mapeo Cromosómico , Cilios/metabolismo , Codón de Terminación , Fibroblastos/metabolismo , Proteínas Hedgehog/fisiología , Heterocigoto , Fenotipo , Polimorfismo Genético , Procesamiento Proteico-Postraduccional , Análisis de Secuencia de ADN , Transducción de Señal , Proteína Gli2 con Dedos de Zinc
16.
Nature ; 470(7333): 279-83, 2011 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-21160473

RESUMEN

Cell-fate transitions involve the integration of genomic information encoded by regulatory elements, such as enhancers, with the cellular environment. However, identification of genomic sequences that control human embryonic development represents a formidable challenge. Here we show that in human embryonic stem cells (hESCs), unique chromatin signatures identify two distinct classes of genomic elements, both of which are marked by the presence of chromatin regulators p300 and BRG1, monomethylation of histone H3 at lysine 4 (H3K4me1), and low nucleosomal density. In addition, elements of the first class are distinguished by the acetylation of histone H3 at lysine 27 (H3K27ac), overlap with previously characterized hESC enhancers, and are located proximally to genes expressed in hESCs and the epiblast. In contrast, elements of the second class, which we term 'poised enhancers', are distinguished by the absence of H3K27ac, enrichment of histone H3 lysine 27 trimethylation (H3K27me3), and are linked to genes inactive in hESCs and instead are involved in orchestrating early steps in embryogenesis, such as gastrulation, mesoderm formation and neurulation. Consistent with the poised identity, during differentiation of hESCs to neuroepithelium, a neuroectoderm-specific subset of poised enhancers acquires a chromatin signature associated with active enhancers. When assayed in zebrafish embryos, poised enhancers are able to direct cell-type and stage-specific expression characteristic of their proximal developmental gene, even in the absence of sequence conservation in the fish genome. Our data demonstrate that early developmental enhancers are epigenetically pre-marked in hESCs and indicate an unappreciated role of H3K27me3 at distal regulatory elements. Moreover, the wealth of new regulatory sequences identified here provides an invaluable resource for studies and isolation of transient, rare cell populations representing early stages of human embryogenesis.


Asunto(s)
Cromatina/genética , Desarrollo Embrionario/genética , Células Madre Embrionarias/metabolismo , Elementos de Facilitación Genéticos/genética , Regulación del Desarrollo de la Expresión Génica/genética , Acetilación , Animales , Diferenciación Celular , Línea Celular , Cromatina/metabolismo , Inmunoprecipitación de Cromatina , ADN Helicasas/metabolismo , Células Madre Embrionarias/citología , Epigénesis Genética/genética , Gastrulación/genética , Estratos Germinativos/embriología , Estratos Germinativos/metabolismo , Histonas/química , Histonas/metabolismo , Humanos , Lisina/metabolismo , Mesodermo/citología , Mesodermo/embriología , Metilación , Placa Neural/citología , Neurulación/genética , Proteínas Nucleares/metabolismo , ARN/análisis , ARN/genética , Factores de Transcripción/metabolismo , Pez Cebra/embriología , Pez Cebra/genética , Factores de Transcripción p300-CBP/metabolismo
18.
Proc Natl Acad Sci U S A ; 110(46): 18555-60, 2013 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-24167250

RESUMEN

The tongue is a critical element of the feeding system in tetrapod animals for their successful adaptation to terrestrial life. Whereas the oral part of the mammalian tongue contains soft tissues only, the avian tongue has an internal skeleton extending to the anterior tip. The mechanisms underlying the evolutionary divergence in tongue skeleton formation are completely unknown. We show here that the odd-skipped related-1 (Osr1) transcription factor is expressed throughout the neural crest-derived tongue mesenchyme in mouse, but not in chick, embryos during early tongue morphogenesis. Neural crest-specific inactivation of Osr1 resulted in formation of an ectopic cartilage in the mouse tongue, reminiscent in shape and developmental ontogeny of the anterior tongue cartilage in chick. SRY-box containing gene-9 (Sox9), the master regulator of chondrogenesis, is widely expressed in the nascent tongue mesenchyme at the onset of tongue morphogenesis but its expression is dramatically down-regulated concomitant with activation of Osr1 expression in the developing mouse tongue. In Osr1 mutant mouse embryos, expression of Sox9 persisted in the developing tongue mesenchyme where chondrogenesis is subsequently activated to form the ectopic cartilage. Furthermore, we show that Osr1 binds to the Sox9 gene promoter and that overexpression of Osr1 suppressed expression of endogenous Sox9 mRNAs and Sox9 promoter-driven reporter. These data indicate that Osr1 normally prevents chondrogenesis in the mammalian tongue through repression of Sox9 expression and suggest that changes in regulation of Osr1 expression in the neural crest-derived tongue mesenchyme underlie the evolutionary divergence of birds from other vertebrates in tongue morphogenesis.


Asunto(s)
Evolución Biológica , Condrogénesis/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Cresta Neural/fisiología , Lengua/embriología , Factores de Transcripción/metabolismo , Animales , Embrión de Pollo , Galactósidos , Hibridación in Situ , Indoles , Luciferasas , Ratones , Ratones Transgénicos , Cresta Neural/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción SOX9/metabolismo , Análisis de Secuencia de ARN , Especificidad de la Especie , Lengua/metabolismo , beta-Galactosidasa/metabolismo
19.
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.

20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA