Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Front Cell Dev Biol ; 11: 1274788, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37854072

RESUMEN

Introduction: The Six1 transcription factor plays important roles in the development of cranial sensory organs, and point mutations underlie craniofacial birth defects. Because Six1's transcriptional activity can be modulated by interacting proteins, we previously screened for candidate interactors and identified zinc-finger MYM-containing protein 4 (Zmym4) by its inclusion of a few domains with a bona fide cofactor, Sine oculis binding protein (Sobp). Although Zmym4 has been implicated in regulating early brain development and certain cancers, its role in craniofacial development has not previously been described. Methods: We used co-immunoprecipitation and luciferase-reporter assays in cultured cells to test interactions between Zmym4 and Six1. We used knock-down and overexpression of Zmym4 in embryos to test for its effects on early ectodermal gene expression, neural crest migration and craniofacial cartilage formation. Results: We found no evidence that Zmym4 physically or transcriptionally interacts with Six1 in cultured cells. Nonetheless, knockdown of endogenous Zmym4 in embryos resulted in altered early cranial gene expression, including those expressed in the neural border, neural plate, neural crest and preplacodal ectoderm. Experimentally increasing Zmym4 levels had minor effects on neural border or neural plate genes, but altered the expression of neural crest and preplacodal genes. At larval stages, genes expressed in the otic vesicle and branchial arches showed reduced expression in Zmym4 morphants. Although we did not detect defects in neural crest migration into the branchial arches, loss of Zmym4 resulted in aberrant morphology of several craniofacial cartilages. Discussion: Although Zmym4 does not appear to function as a Six1 transcriptional cofactor, it plays an important role in regulating the expression of embryonic cranial genes in tissues critical for normal craniofacial development.

2.
Dev Dyn ; 252(10): 1303-1315, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37183792

RESUMEN

BACKGROUND: Genetic variants of the transcription factor SIX1 and its co-factor EYA1 underlie 50% of Branchio-oto-renal syndrome (BOR) cases. BOR is characterized by craniofacial defects, including malformed middle ear ossicles leading to conductive hearing loss. In this work, we expand our knowledge of the Six1 gene regulatory network by using a Six1-null mouse line to assess gene expression profiles of E10.5 mandibular arches, which give rise to the neural crest (NC)-derived middle ear ossicles and lower jaw, via bulk RNA sequencing. RESULTS: Our transcriptomic analysis led to the identification of 808 differentially expressed genes that are related to translation, NC cell differentiation, osteogenesis, and chondrogenesis including components of the WNT signaling pathway. As WNT signaling is a known contributor to bone development, we demonstrated that SIX1 is required for expression of the WNT antagonist Frzb in the mandibular arch, and determined that SIX1 expression results in repression of WNT signaling. CONCLUSION: Our results clarify the mechanisms by which SIX1 regulates the development of NC-derived craniofacial elements that are altered in SIX1-associated disorders. In addition, this work identifies novel genes that could be causative to this birth defect and establishes a link between SIX1 and WNT signaling during patterning of NC cells.

3.
J Dev Biol ; 10(3)2022 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-35893122

RESUMEN

Each year approximately 35% of babies are born with craniofacial abnormalities of the skull, jaws, ears, and/or teeth, which in turn can lead to problems in feeding, hearing, and sight [...].

4.
Dev Biol ; 489: 62-75, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35697116

RESUMEN

Mcrs1 is a multifunctional protein that is critical for many cellular processes in a wide range of cell types. Previously, we showed that Mcrs1 binds to the Six1 transcription factor and reduces the ability of the Six1-Eya1 complex to upregulate transcription, and that Mcrs1 loss-of-function leads to the expansion of several neural plate genes, reduction of neural border and pre-placodal ectoderm (PPR) genes, and pleiotropic effects on various neural crest (NC) genes. Because the affected embryonic structures give rise to several of the cranial tissues affected in Branchio-otic/Branchio-oto-renal (BOR) syndrome, herein we tested whether these gene expression changes subsequently alter the development of the proximate precursors of BOR affected structures - the otic vesicles (OV) and branchial arches (BA). We found that Mcrs1 is required for the expression of several OV genes involved in inner ear formation, patterning and otic capsule cartilage formation. Mcrs1 knockdown also reduced the expression domains of many genes expressed in the larval BA, derived from either NC or PPR, except for emx2, which was expanded. Reduced Mcrs1 also diminished the length of the expression domain of tbx1 in BA1 and BA2 and interfered with cranial NC migration from the dorsal neural tube; this subsequently resulted in defects in the morphology of lower jaw cartilages derived from BA1 and BA2, including the infrarostral, Meckel's, and ceratohyal as well as the otic capsule. These results demonstrate that Mcrs1 plays an important role in processes that lead to the formation of craniofacial cartilages and its loss results in phenotypes consistent with reduced Six1 activity associated with BOR.


Asunto(s)
Región Branquial , Síndrome Branquio Oto Renal , Región Branquial/metabolismo , Síndrome Branquio Oto Renal/genética , Síndrome Branquio Oto Renal/metabolismo , Cartílago/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Cresta Neural , Placa Neural/metabolismo , Proteínas de Unión al ARN/metabolismo
5.
Front Cell Dev Biol ; 10: 786052, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35198557

RESUMEN

The embryonic ectoderm is composed of four domains: neural plate, neural crest, pre-placodal region (PPR) and epidermis. Their formation is initiated during early gastrulation by dorsal-ventral and anterior-posterior gradients of signaling factors that first divide the embryonic ectoderm into neural and non-neural domains. Next, the neural crest and PPR domains arise, either via differential competence of the neural and non-neural ectoderm (binary competence model) or via interactions between the neural and non-neural ectoderm tissues to produce an intermediate neural border zone (NB) (border state model) that subsequently separates into neural crest and PPR. Many previous gain- and loss-of-function experiments demonstrate that numerous TFs are expressed in initially overlapping zones that gradually resolve into patterns that by late neurula stages are characteristic of each of the four domains. Several of these studies suggested that this is accomplished by a combination of repressive TF interactions and competence to respond to local signals. In this study, we ectopically expressed TFs that at neural plate stages are characteristic of one domain in a different domain to test whether they act cell autonomously as repressors. We found that almost all tested TFs caused reduced expression of the other TFs. At gastrulation these effects were strictly within the lineage-labeled cells, indicating that the effects were cell autonomous, i.e., due to TF interactions within individual cells. Analysis of previously published single cell RNAseq datasets showed that at the end of gastrulation, and continuing to neural tube closure stages, many ectodermal cells express TFs characteristic of more than one neural plate stage domain, indicating that different TFs have the opportunity to interact within the same cell. At neurula stages repression was observed both in the lineage-labeled cells and in adjacent cells not bearing detectable lineage label, suggesting that cell-to-cell signaling has begun to contribute to the separation of the domains. Together, these observations directly demonstrate previous suggestions in the literature that the segregation of embryonic ectodermal domains initially involves cell autonomous, repressive TF interactions within an individual cell followed by the subsequent advent of non-cell autonomous signaling to neighbors.

6.
Genesis ; 59(12): e23453, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34664392

RESUMEN

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.


Asunto(s)
Síndrome Branquio Oto Renal/genética , Anomalías Congénitas/genética , Pérdida Auditiva/genética , Proteínas de Homeodominio/genética , Proteínas de Xenopus/genética , Animales , Región Branquial/crecimiento & desarrollo , Región Branquial/patología , Síndrome Branquio Oto Renal/fisiopatología , Sistemas CRISPR-Cas/genética , Anomalías Congénitas/patología , Desarrollo Embrionario/genética , Ganglios Parasimpáticos/crecimiento & desarrollo , Ganglios Parasimpáticos/patología , Expresión Génica , Regulación del Desarrollo de la Expresión Génica/genética , Pérdida Auditiva/fisiopatología , Humanos , Tubo Neural/crecimiento & desarrollo , Tubo Neural/patología , Cráneo/crecimiento & desarrollo , Cráneo/patología , Factores de Transcripción/genética , Xenopus/genética , Xenopus/crecimiento & desarrollo
7.
Development ; 148(17)2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34414417

RESUMEN

Branchio-oto-renal syndrome (BOR) is a disorder characterized by hearing loss, and craniofacial and/or renal defects. Variants in the transcription factor Six1 and its co-factor Eya1, both of which are required for otic development, are linked to BOR. We previously identified Sobp as a potential Six1 co-factor, and SOBP variants in mouse and humans cause otic phenotypes; therefore, we asked whether Sobp interacts with Six1 and thereby may contribute to BOR. Co-immunoprecipitation and immunofluorescence experiments demonstrate that Sobp binds to and colocalizes with Six1 in the cell nucleus. Luciferase assays show that Sobp interferes with the transcriptional activation of Six1+Eya1 target genes. Experiments in Xenopus embryos that either knock down or increase expression of Sobp show that it is required for formation of ectodermal domains at neural plate stages. In addition, altering Sobp levels disrupts otic vesicle development and causes craniofacial cartilage defects. Expression of Xenopus Sobp containing the human variant disrupts the pre-placodal ectoderm similar to full-length Sobp, but other changes are distinct. These results indicate that Sobp modifies Six1 function and is required for vertebrate craniofacial development, and identify Sobp as a potential candidate gene for BOR.


Asunto(s)
Desarrollo Óseo , Proteínas de Homeodominio/metabolismo , Metaloproteínas/metabolismo , Proteínas Nucleares/metabolismo , Proteínas de Xenopus/metabolismo , Animales , Síndrome Branquio Oto Renal/embriología , Síndrome Branquio Oto Renal/genética , Núcleo Celular/metabolismo , Oído Interno/embriología , Oído Interno/metabolismo , Ectodermo/embriología , Ectodermo/metabolismo , Expresión Génica , Proteínas de Homeodominio/genética , Larva/crecimiento & desarrollo , Metaloproteínas/genética , Cresta Neural/embriología , Cresta Neural/metabolismo , Proteínas Nucleares/genética , Unión Proteica , Proteínas Tirosina Fosfatasas/metabolismo , Activación Transcripcional , Proteínas de Xenopus/genética , Xenopus laevis
8.
J Dev Biol ; 9(3)2021 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-34208995

RESUMEN

Several single-nucleotide mutations in SIX1 underlie branchio-otic/branchio-oto-renal (BOR) syndrome, but the clinical literature has not been able to correlate different variants with specific phenotypes. We previously assessed whether variants in either the cofactor binding domain (V17E, R110W) or the DNA binding domain (W122R, Y129C) might differentially affect early embryonic gene expression, and found that each variant had a different combination of effects on neural crest and placode gene expression. Since the otic vesicle gives rise to the inner ear, which is consistently affected in BOR, herein we focused on whether the variants differentially affected the otic expression of genes previously found to be likely Six1 targets. We found that V17E, which does not bind Eya cofactors, was as effective as wild-type Six1 in reducing most otic target genes, whereas R110W, W122R and Y129C, which bind Eya, were significantly less effective. Notably, V17E reduced the otic expression of prdm1, whereas R110W, W122R and Y129C expanded it. Since each mutant has defective transcriptional activity but differs in their ability to interact with Eya cofactors, we propose that altered cofactor interactions at the mutated sites differentially interfere with their ability to drive otic gene expression, and these differences may contribute to patient phenotype variability.

9.
Eur J Dent Educ ; 25(2): 332-341, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33012127

RESUMEN

INTRODUCTION: Technological resources are considered important for the practice and training in endodontics. It is not yet clear the extent of the insertion of these resources in predoctoral dental programmes and the effect of such resources to the general dentist training. AIM: To evaluate the faculty perception regarding the insertion of technological resources, particularly rotary instruments, and the impact of such resources to the student's learning process graded at the end of predoctoral dental programmes in Brazil by the National Students Performance Exam (ENADE). METHODS: The endodontic department faculties in all 205 programmes that participated in the 2016 ENADE received a questionnaire by email. The institution, faculty credentials, and insertion of technologies in each programme were described, and an association between these data and the ENADE score was tested. RESULTS: 149 educators (72.7%) answered the form. From this total, 73.2% of them were from private institutions and 26.8% from public ones. Educators mix manual and rotary instrumentation to treat selected patients in 47.7% of the programmes. Most educators (89.9%) consider the utilisation of technology in endodontics as positive. Whilst there was not a significant association between the use of rotary instrumentation and better performance in ENADE, there was a significant association between the performance and the supply of special instruments by the institutions. CONCLUSION: These results show that even though most faculties consider the insertion of technologies as positive, the factor that caused a significant impact to the training of the general dentist is the general infrastructure of the institutions.


Asunto(s)
Endodoncia , Preparación del Conducto Radicular , Brasil , Odontólogos , Educación en Odontología , Humanos , Tecnología
11.
Am J Med Genet A ; 182(5): 1104-1116, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32133772

RESUMEN

Craniofacial morphogenesis is regulated in part by signaling from the Endothelin receptor type A (EDNRA). Pathogenic variants in EDNRA signaling pathway components EDNRA, GNAI3, PCLB4, and EDN1 cause Mandibulofacial Dysostosis with Alopecia (MFDA), Auriculocondylar syndrome (ARCND) 1, 2, and 3, respectively. However, cardiovascular development is normal in MFDA and ARCND individuals, unlike Ednra knockout mice. One explanation may be that partial EDNRA signaling remains in MFDA and ARCND, as mice with reduced, but not absent, EDNRA signaling also lack a cardiovascular phenotype. Here we report an individual with craniofacial and cardiovascular malformations mimicking the Ednra -/- mouse phenotype, including a distinctive micrognathia with microstomia and a hypoplastic aortic arch. Exome sequencing found a novel homozygous missense variant in EDNRA (c.1142A>C; p.Q381P). Bioluminescence resonance energy transfer assays revealed that this amino acid substitution in helix 8 of EDNRA prevents recruitment of G proteins to the receptor, abrogating subsequent receptor activation by its ligand, Endothelin-1. This homozygous variant is thus the first reported loss-of-function EDNRA allele, resulting in a syndrome we have named Oro-Oto-Cardiac Syndrome. Further, our results illustrate that EDNRA signaling is required for both normal human craniofacial and cardiovascular development, and that limited EDNRA signaling is likely retained in ARCND and MFDA individuals. This work illustrates a straightforward approach to identifying the functional consequence of novel genetic variants in signaling molecules associated with malformation syndromes.


Asunto(s)
Anomalías Craneofaciales/genética , Enfermedades del Oído/genética , Oído/anomalías , Predisposición Genética a la Enfermedad , Disostosis Mandibulofacial/genética , Receptor de Endotelina A/genética , Animales , Anomalías Craneofaciales/fisiopatología , Oído/fisiopatología , Enfermedades del Oído/fisiopatología , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/genética , Regulación del Desarrollo de la Expresión Génica/genética , Humanos , Mutación con Pérdida de Función/genética , Disostosis Mandibulofacial/fisiopatología , Ratones , Ratones Noqueados , Morfogénesis/genética , Cresta Neural/crecimiento & desarrollo , Cresta Neural/patología , Fenotipo , Transducción de Señal/genética
12.
Hum Mol Genet ; 29(6): 1002-1017, 2020 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-32047912

RESUMEN

LgDel mice, which model the heterozygous deletion of genes at human chromosome 22q11.2 associated with DiGeorge/22q11.2 deletion syndrome (22q11DS), have cranial nerve and craniofacial dysfunction as well as disrupted suckling, feeding and swallowing, similar to key 22q11DS phenotypes. Divergent trigeminal nerve (CN V) differentiation and altered trigeminal ganglion (CNgV) cellular composition prefigure these disruptions in LgDel embryos. We therefore asked whether a distinct transcriptional state in a specific population of early differentiating LgDel cranial sensory neurons, those in CNgV, a major source of innervation for appropriate oropharyngeal function, underlies this departure from typical development. LgDel versus wild-type (WT) CNgV transcriptomes differ significantly at E10.5 just after the ganglion has coalesced. Some changes parallel altered proportions of cranial placode versus cranial neural crest-derived CNgV cells. Others are consistent with a shift in anterior-posterior patterning associated with divergent LgDel cranial nerve differentiation. The most robust quantitative distinction, however, is statistically verifiable increased variability of expression levels for most of the over 17 000 genes expressed in common in LgDel versus WT CNgV. Thus, quantitative expression changes of functionally relevant genes and increased stochastic variation across the entire CNgV transcriptome at the onset of CN V differentiation prefigure subsequent disruption of cranial nerve differentiation and oropharyngeal function in LgDel mice.


Asunto(s)
Síndrome de DiGeorge/patología , Modelos Animales de Enfermedad , Embrión de Mamíferos/patología , Regulación de la Expresión Génica , Células Receptoras Sensoriales/patología , Transcriptoma , Nervio Trigémino/patología , Animales , Síndrome de DiGeorge/genética , Embrión de Mamíferos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Receptoras Sensoriales/metabolismo , Nervio Trigémino/metabolismo
13.
Dis Model Mech ; 13(3)2020 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-31980437

RESUMEN

Single-nucleotide mutations in human SIX1 result in amino acid substitutions in either the protein-protein interaction domain or the homeodomain, and cause ∼4% of branchio-otic (BOS) and branchio-oto-renal (BOR) cases. The phenotypic variation between patients with the same mutation, even within affected members of the same family, make it difficult to functionally distinguish between the different SIX1 mutations. We made four of the BOS/BOR substitutions in the Xenopus Six1 protein (V17E, R110W, W122R, Y129C), which is 100% identical to human in both the protein-protein interaction domain and the homeodomain, and expressed them in embryos to determine whether they cause differential changes in early craniofacial gene expression, otic gene expression or otic morphology. We confirmed that, similar to the human mutants, all four mutant Xenopus Six1 proteins access the nucleus but are transcriptionally deficient. Analysis of craniofacial gene expression showed that each mutant causes specific, often different and highly variable disruptions in the size of the domains of neural border zone, neural crest and pre-placodal ectoderm genes. Each mutant also had differential effects on genes that pattern the otic vesicle. Assessment of the tadpole inner ear demonstrated that while the auditory and vestibular structures formed, the volume of the otic cartilaginous capsule, otoliths, lumen and a subset of the hair cell-containing sensory patches were reduced. This detailed description of the effects of BOS/BOR-associated SIX1 mutations in the embryo indicates that each causes subtle changes in gene expression in the embryonic ectoderm and otocyst, leading to inner ear morphological anomalies.


Asunto(s)
Síndrome Branquio Oto Renal/genética , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Mutación/genética , Cráneo/embriología , Secuencia de Aminoácidos , Animales , Oído , Células HEK293 , Proteínas de Homeodominio/química , Proteínas de Homeodominio/metabolismo , Humanos , Cresta Neural/metabolismo , Membrana Otolítica/metabolismo , Proteínas Tirosina Fosfatasas/metabolismo , Transcripción Genética , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo , Xenopus laevis/embriología , Xenopus laevis/genética
14.
Dev Dyn ; 247(3): 542-554, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28631378

RESUMEN

BACKGROUND: Although normally linked to bone and cartilage development, the Runt-related transcription factor, RUNX2, was reported in the mouse heart during development of the valves. We examined RUNX2 expression and function in the developing avian heart as it related to the epithelial-mesenchymal transition (EMT) in the atrioventricular canal. EMT can be separated into an activation stage involving hypertrophy and cell separation and an invasion stage where cells invade the extracellular matrix. The localization and activity of RUNX2 was explored in relation to these steps in the heart. As RUNX2 was also reported in cancer tissues, we examined its expression in the progression of esophageal cancer in staged tissues. RESULTS: A specific isoform, RUNX2-I, is present and required for EMT by endothelia of the atrioventricular canal. Knockdown of RUNX2-I inhibits the cell-cell separation that is characteristic of initial activation of EMT. Loss of RUNX2-I altered expression of EMT markers to a greater extent during activation than during subsequent cell invasion. Transforming growth factor beta 2 (TGFß2) mediates activation during cardiac endothelial EMT. Consistent with a role in activation, RUNX2-I is regulated by TGFß2 and its activity is independent of similarly expressed Snai2 in regulation of EMT. Examination of RUNX2 expression in esophageal cancer showed its upregulation concomitant with the development of dysplasia and continued expression in adenocarcinoma. CONCLUSIONS: These data introduce the RUNX2-I isoform as a critical early transcription factor mediating EMT in the developing heart after induction by TGFß2. Its expression in tumor tissue suggests a similar role for RUNX2 in the EMT of metastasis. Developmental Dynamics 247:542-554, 2018. © 2017 Wiley Periodicals, Inc.


Asunto(s)
Subunidad alfa 1 del Factor de Unión al Sitio Principal/fisiología , Transición Epitelial-Mesenquimal , Animales , Embrión de Pollo , Pollos , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Neoplasias/metabolismo , Isoformas de Proteínas , Activación Transcripcional , Factor de Crecimiento Transformador beta
15.
Development ; 144(11): 2021-2031, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28455376

RESUMEN

Jaw morphogenesis is a complex event mediated by inductive signals that establish and maintain the distinct developmental domains required for formation of hinged jaws, the defining feature of gnathostomes. The mandibular portion of pharyngeal arch 1 is patterned dorsally by Jagged-Notch signaling and ventrally by endothelin receptor A (EDNRA) signaling. Loss of EDNRA signaling disrupts normal ventral gene expression, the result of which is homeotic transformation of the mandible into a maxilla-like structure. However, loss of Jagged-Notch signaling does not result in significant changes in maxillary development. Here we show in mouse that the transcription factor SIX1 regulates dorsal arch development not only by inducing dorsal Jag1 expression but also by inhibiting endothelin 1 (Edn1) expression in the pharyngeal endoderm of the dorsal arch, thus preventing dorsal EDNRA signaling. In the absence of SIX1, but not JAG1, aberrant EDNRA signaling in the dorsal domain results in partial duplication of the mandible. Together, our results illustrate that SIX1 is the central mediator of dorsal mandibular arch identity, thus ensuring separation of bone development between the upper and lower jaws.


Asunto(s)
Endotelina-1/metabolismo , Proteínas de Homeodominio/metabolismo , Maxilar/embriología , Maxilar/metabolismo , Transducción de Señal , Animales , Tipificación del Cuerpo/genética , Región Branquial/metabolismo , Anomalías Craneofaciales/embriología , Anomalías Craneofaciales/genética , Anomalías Craneofaciales/patología , Embrión de Mamíferos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Integrasas/metabolismo , Ratones , Modelos Biológicos , Cresta Neural/metabolismo , Receptor de Endotelina A/metabolismo , Receptores Notch/metabolismo , Proteínas Serrate-Jagged/metabolismo , Factor de Transcripción Sp7 , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Regulación hacia Arriba/genética , Cigoma/embriología , Cigoma/metabolismo
16.
Genesis ; 55(3)2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28109039

RESUMEN

In gnathostomes, dorsoventral (D-V) patterning of neural crest cells (NCC) within the pharyngeal arches is crucial for the development of hinged jaws. One of the key signals that mediate this process is Endothelin-1 (EDN1). Loss of EDN1 binding to the Endothelin-A receptor (EDNRA) results in loss of EDNRA signaling and subsequent facial birth defects in humans, mice and zebrafish. A rate-limiting step in this crucial signaling pathway is the conversion of immature EDN1 into a mature active form by Endothelin converting enzyme-1 (ECE1). However, surprisingly little is known about how Ece1 transcription is induced or regulated. We show here that Nkx2.5 is required for proper craniofacial development in zebrafish and acts in part by upregulating ece1 expression. Disruption of nkx2.5 in zebrafish embryos results in defects in both ventral and dorsal pharyngeal arch-derived elements, with changes in ventral arch gene expression consistent with a disruption in Ednra signaling. ece1 mRNA rescues the nkx2.5 morphant phenotype, indicating that Nkx2.5 functions through modulating Ece1 expression or function. These studies illustrate a new function for Nkx2.5 in embryonic development and provide new avenues with which to pursue potential mechanisms underlying human facial disorders.


Asunto(s)
Enzimas Convertidoras de Endotelina/genética , Regulación del Desarrollo de la Expresión Génica , Proteína Homeótica Nkx-2.5/genética , Cresta Neural/metabolismo , Proteínas de Pez Cebra/genética , Animales , Enzimas Convertidoras de Endotelina/metabolismo , Proteína Homeótica Nkx-2.5/metabolismo , Ratones , Cresta Neural/embriología , Faringe/embriología , Faringe/metabolismo , Regulación hacia Arriba , Pez Cebra , Proteínas de Pez Cebra/metabolismo
17.
Curr Top Dev Biol ; 115: 335-75, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26589932

RESUMEN

Defects in craniofacial development represent a majority of observed human birth defects, occurring at a rate as high as 1:800 live births. These defects often occur due to changes in neural crest cell (NCC) patterning and development and can affect non-NCC-derived structures due to interactions between NCCs and the surrounding cell types. Proper craniofacial development requires an intricate array of gene expression networks that are tightly controlled spatiotemporally by a number of regulatory mechanisms. One of these mechanisms involves the action of microRNAs (miRNAs), a class of noncoding RNAs that repress gene expression by binding to miRNA recognition sequences typically located in the 3' UTR of target mRNAs. Recent evidence illustrates that miRNAs are crucial for vertebrate facial morphogenesis, with changes in miRNA expression leading to facial birth defects, including some in complex human syndromes such as 22q11 (DiGeorge Syndrome). In this review, we highlight the current understanding of miRNA biogenesis, the roles of miRNAs in overall craniofacial development, the impact that loss of miRNAs has on normal development and the requirement for miRNAs in the development of specific craniofacial structures, including teeth. From these studies, it is clear that miRNAs are essential for normal facial development and morphogenesis, and a potential key in establishing new paradigms for repair and regeneration of facial defects.


Asunto(s)
MicroARNs/genética , Morfogénesis/genética , Cráneo/embriología , Cráneo/metabolismo , Regiones no Traducidas 3'/genética , Animales , Huesos Faciales/embriología , Huesos Faciales/metabolismo , Regulación del Desarrollo de la Expresión Génica , Humanos , MicroARNs/metabolismo , Modelos Genéticos , ARN Mensajero/genética , ARN Mensajero/metabolismo
18.
Am J Hum Genet ; 96(4): 519-31, 2015 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-25772936

RESUMEN

The endothelin receptor type A (EDNRA) signaling pathway is essential for the establishment of mandibular identity during development of the first pharyngeal arch. We report four unrelated individuals with the syndrome mandibulofacial dysostosis with alopecia (MFDA) who have de novo missense variants in EDNRA. Three of the four individuals have the same substitution, p.Tyr129Phe. Tyr129 is known to determine the selective affinity of EDNRA for endothelin 1 (EDN1), its major physiological ligand, and the p.Tyr129Phe variant increases the affinity of the receptor for EDN3, its non-preferred ligand, by two orders of magnitude. The fourth individual has a somatic mosaic substitution, p.Glu303Lys, and was previously described as having Johnson-McMillin syndrome. The zygomatic arch of individuals with MFDA resembles that of mice in which EDNRA is ectopically activated in the maxillary prominence, resulting in a maxillary to mandibular transformation, suggesting that the p.Tyr129Phe variant causes an EDNRA gain of function in the developing upper jaw. Our in vitro and in vivo assays suggested complex, context-dependent effects of the EDNRA variants on downstream signaling. Our findings highlight the importance of finely tuned regulation of EDNRA signaling during human craniofacial development and suggest that modification of endothelin receptor-ligand specificity was a key step in the evolution of vertebrate jaws.


Asunto(s)
Alopecia/genética , Disostosis Mandibulofacial/genética , Receptor de Endotelina A/genética , Alopecia/patología , Animales , Secuencia de Bases , Endotelina-1/metabolismo , Exoma/genética , Humanos , Hibridación in Situ , Disostosis Mandibulofacial/patología , Datos de Secuencia Molecular , Morfolinos/genética , Mutación Missense/genética , Linaje , ARN Mensajero/administración & dosificación , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor de Endotelina A/metabolismo , Análisis de Secuencia de ADN , Síndrome , Tomografía Computarizada por Rayos X , Pez Cebra , Cigoma/patología
19.
Dev Biol ; 400(2): 191-201, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25725491

RESUMEN

Endothelin-1 (EDN1) influences both craniofacial and cardiovascular development and a number of adult physiological conditions by binding to one or both of the known endothelin receptors, thus initiating multiple signaling cascades. Animal models containing both conventional and conditional loss of the Edn1 gene have been used to dissect EDN1 function in both embryos and adults. However, while transgenic Edn1 over-expression or targeted genomic insertion of Edn1 has been performed to understand how elevated levels of Edn1 result in or exacerbate disease states, an animal model in which Edn1 over-expression can be achieved in a spatiotemporal-specific manner has not been reported. Here we describe the creation of Edn1 conditional over-expression transgenic mouse lines in which the chicken ß-actin promoter and an Edn1 cDNA are separated by a strong stop sequence flanked by loxP sites. In the presence of Cre, the stop cassette is removed, leading to Edn1 expression. Using the Wnt1-Cre strain, in which Cre expression is targeted to the Wnt1-expressing domain of the central nervous system (CNS) from which neural crest cells (NCCs) arise, we show that stable chicken ß-actin-Edn1 (CBA-Edn1) transgenic lines with varying EDN1 protein levels develop defects in NCC-derived tissues of the face, though the severity differs between lines. We also show that Edn1 expression can be achieved in other embryonic tissues utilizing other Cre strains, with this expression also resulting in developmental defects. CBA-Edn1 transgenic mice will be useful in investigating diverse aspects of EDN1-mediated-development and disease, including understanding how NCCs achieve and maintain a positional and functional identity and how aberrant EDN1 levels can lead to multiple physiological changes and diseases.


Asunto(s)
Modelos Animales de Enfermedad , Endotelina-1/genética , Técnicas Genéticas , Integrasas , Animales , Pollos , Embrión de Mamíferos/metabolismo , Ratones , Ratones Transgénicos , Cresta Neural/citología , Proteínas Recombinantes/genética
20.
Am J Med Genet C Semin Med Genet ; 163C(4): 306-17, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24123988

RESUMEN

Among human birth defect syndromes, malformations affecting the face are perhaps the most striking due to cultural and psychological expectations of facial shape. One such syndrome is auriculocondylar syndrome (ACS), in which patients present with defects in ear and mandible development. Affected structures arise from cranial neural crest cells, a population of cells in the embryo that reside in the pharyngeal arches and give rise to most of the bone, cartilage and connective tissue of the face. Recent studies have found that most cases of ACS arise from defects in signaling molecules associated with the endothelin signaling pathway. Disruption of this signaling pathway in both mouse and zebrafish results in loss of identity of neural crest cells of the mandibular portion of the first pharyngeal arch and the subsequent repatterning of these cells, leading to homeosis of lower jaw structures into more maxillary-like structures. These findings illustrate the importance of endothelin signaling in normal human craniofacial development and illustrate how clinical and basic science approaches can coalesce to improve our understanding of the genetic basis of human birth defect syndromes. Further, understanding the genetic basis for ACS that lies outside of known endothelin signaling components may help elucidate unknown aspects critical to the establishment of neural crest cell patterning during facial morphogenesis.


Asunto(s)
Tipificación del Cuerpo/genética , Región Branquial/fisiopatología , Enfermedades del Oído/fisiopatología , Oído/anomalías , Transducción de Señal , Animales , Región Branquial/crecimiento & desarrollo , Modelos Animales de Enfermedad , Oído/fisiopatología , Enfermedades del Oído/genética , Cara/patología , Regulación del Desarrollo de la Expresión Génica , Humanos , Mandíbula/crecimiento & desarrollo , Mandíbula/patología , Desarrollo Maxilofacial/genética , Ratones , Cresta Neural/crecimiento & desarrollo , Cresta Neural/patología , Pez Cebra
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...