Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
PLoS Genet ; 20(7): e1011364, 2024 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-39052671

RESUMEN

How the dorsal-ventral axis of the vertebrate jaw, particularly the position of tooth initiation site, is established remains a critical and unresolved question. Tooth development starts with the formation of the dental lamina, a localized thickened strip within the maxillary and mandibular epithelium. To identify transcriptional regulatory networks (TRN) controlling the specification of dental lamina from the naïve mandibular epithelium, we utilized Laser Microdissection coupled low-input RNA-seq (LMD-RNA-seq) to profile gene expression of different domains of the mandibular epithelium along the dorsal-ventral axis. We comprehensively identified transcription factors (TFs) and signaling pathways that are differentially expressed along mandibular epithelial domains (including the dental lamina). Specifically, we found that the TFs Sox2 and Tfap2 (Tfap2a/Tfap2b) formed complimentary expression domains along the dorsal-ventral axis of the mandibular epithelium. Interestingly, both classic and novel dental lamina specific TFs-such as Pitx2, Ascl5 and Zfp536-were found to localize near the Sox2:Tfap2a/Tfap2b interface. To explore the functional significance of these domain specific TFs, we next examined loss-of-function mouse models of these domain specific TFs, including the dental lamina specific TF, Pitx2, and the ventral surface ectoderm specific TFs Tfap2a and Tfap2b. We found that disruption of domain specific TFs leads to an upregulation and expansion of the alternative domain's TRN. The importance of this cross-repression is evident by the ectopic expansion of Pitx2 and Sox2 positive dental lamina structure in Tfap2a/Tfap2b ectodermal double knockouts and the emergence of an ectopic tooth in the ventral surface ectoderm. Finally, we uncovered an unappreciated interface of mesenchymal SHH and WNT signaling pathways, at the site of tooth initiation, that were established by the epithelial domain specific TFs including Pitx2 and Tfap2a/Tfap2b. These results uncover a previously unknown molecular mechanism involving cross-repression of domain specific TFs including Pitx2 and Tfap2a/Tfap2b in patterning the dorsal-ventral axis of the mouse mandible, specifically the regulation of tooth initiation site.

2.
J Biol Chem ; 299(11): 105324, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37806494

RESUMEN

Wolf-Hirschhorn syndrome (WHS) is a developmental disorder attributed to a partial deletion on the short arm of chromosome 4. WHS patients suffer from oral manifestations including cleft lip and palate, hypodontia, and taurodontism. WHS candidate 1 (WHSC1) gene is a H3K36-specific methyltransferase that is deleted in every reported case of WHS. Mutation in this gene also results in tooth anomalies in patients. However, the correlation between genetic abnormalities and the tooth anomalies has remained controversial. In our study, we aimed to clarify the role of WHSC1 in tooth development. We profiled the Whsc1 expression pattern during mouse incisor and molar development by immunofluorescence staining and found Whsc1 expression is reduced as tooth development proceeds. Using real-time quantitative reverse transcription PCR, Western blot, chromatin immunoprecipitation, and luciferase assays, we determined that Whsc1 and Pitx2, the initial transcription factor involved in tooth development, positively and reciprocally regulate each other through their gene promoters. miRNAs are known to regulate gene expression posttranscriptionally during development. We previously reported miR-23a/b and miR-24-1/2 were highly expressed in the mature tooth germ. Interestingly, we demonstrate here that these two miRs directly target Whsc1 and repress its expression. Additionally, this miR cluster is also negatively regulated by Pitx2. We show the expression of these two miRs and Whsc1 are inversely correlated during mouse mandibular development. Taken together, our results provide new insights into the potential role of Whsc1 in regulating tooth development and a possible molecular mechanism underlying the dental defects in WHS.


Asunto(s)
Labio Leporino , Fisura del Paladar , MicroARNs , Síndrome de Wolf-Hirschhorn , Animales , Ratones , MicroARNs/genética , Factores de Transcripción , Síndrome de Wolf-Hirschhorn/genética , Síndrome de Wolf-Hirschhorn/metabolismo , Proteína del Homeodomínio PITX2
3.
J Biol Chem ; 298(9): 102295, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35872015

RESUMEN

The chromatin-associated high mobility group protein N2 (HMGN2) cofactor regulates transcription factor activity through both chromatin and protein interactions. Hmgn2 expression is known to be developmentally regulated, but the post-transcriptional mechanisms that regulate Hmgn2 expression and its precise roles in tooth development remain unclear. Here, we demonstrate that HMGN2 inhibits the activity of multiple transcription factors as a general mechanism to regulate early development. Bimolecular fluorescence complementation, pull-down, and coimmunoprecipitation assays show that HMGN2 interacts with the transcription factor Lef-1 through its HMG-box domain as well as with other early development transcription factors, Dlx2, FoxJ1, and Pitx2. Furthermore, EMSAs demonstrate that HMGN2 binding to Lef-1 inhibits its DNA-binding activity. We found that Pitx2 and Hmgn2 associate with H4K5ac and H3K4me2 chromatin marks in the proximal Dlx2 promoter, demonstrating Hmgn2 association with open chromatin. In addition, we demonstrate that microRNAs (miRs) mir-23a and miR-23b directly target Hmgn2, promoting transcriptional activation at several gene promoters, including the amelogenin promoter. In vivo, we found that decreased Hmgn2 expression correlates with increased miR-23 expression in craniofacial tissues as the murine embryo develops. Finally, we show that ablation of Hmgn2 in mice results in increased amelogenin expression because of increased Pitx2, Dlx2, Lef-1, and FoxJ1 transcriptional activity. Taken together, our results demonstrate both post-transcriptional regulation of Hmgn2 by miR-23a/b and post-translational regulation of gene expression by Hmgn2-transcription factor interactions. We conclude that HMGN2 regulates tooth development through its interaction with multiple transcription factors.


Asunto(s)
Amelogénesis , Regulación de la Expresión Génica , Proteína HMGN2 , Proteínas de Homeodominio , Factor de Unión 1 al Potenciador Linfoide , Factores de Transcripción , Transcripción Genética , Amelogénesis/genética , Amelogenina/genética , Animales , Cromatina/metabolismo , Proteína HMGN2/genética , Proteína HMGN2/metabolismo , Proteínas de Homeodominio/metabolismo , Factor de Unión 1 al Potenciador Linfoide/metabolismo , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Factores de Transcripción/metabolismo , Proteína del Homeodomínio PITX2
4.
Development ; 147(11)2020 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-32439755

RESUMEN

Epithelial signaling centers control epithelial invagination and organ development, but how these centers are specified remains unclear. We report that Pitx2 (the first transcriptional marker for tooth development) controls the embryonic formation and patterning of epithelial signaling centers during incisor development. We demonstrate using Krt14Cre /Pitx2flox/flox (Pitx2cKO ) and Rosa26CreERT/Pitx2flox/flox mice that loss of Pitx2 delays epithelial invagination, and decreases progenitor cell proliferation and dental epithelium cell differentiation. Developmentally, Pitx2 regulates formation of the Sox2+ labial cervical loop (LaCL) stem cell niche in concert with two signaling centers: the initiation knot and enamel knot. The loss of Pitx2 disrupted the patterning of these two signaling centers, resulting in tooth arrest at E14.5. Mechanistically, Pitx2 transcriptional activity and DNA binding is inhibited by Sox2, and this interaction controls gene expression in specific Sox2 and Pitx2 co-expression progenitor cell domains. We demonstrate new transcriptional mechanisms regulating signaling centers by Pitx2, Sox2, Lef1 and Irx1.


Asunto(s)
Células Epiteliales/metabolismo , Proteínas de Homeodominio/metabolismo , Factor de Unión 1 al Potenciador Linfoide/metabolismo , Factores de Transcripción SOXB1/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Diferenciación Celular , Proliferación Celular , Esmalte Dental/metabolismo , Embrión de Mamíferos/metabolismo , Células Epiteliales/citología , Regulación del Desarrollo de la Expresión Génica , Proteínas Hedgehog/metabolismo , Proteínas de Homeodominio/genética , Factor de Unión 1 al Potenciador Linfoide/genética , Ratones , Ratones Noqueados , Odontogénesis , Factores de Transcripción SOXB1/genética , Nicho de Células Madre , Células Madre/citología , Células Madre/metabolismo , Diente/citología , Diente/crecimiento & desarrollo , Diente/metabolismo , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Proteínas Señalizadoras YAP , Proteína del Homeodomínio PITX2
5.
Biochem Soc Trans ; 51(2): 841-854, 2023 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-37073783

RESUMEN

microRNAs (miRs) have been reported over the decades as important regulators in bone development and bone regeneration. They play important roles in maintaining the stem cell signature as well as regulating stem cell fate decisions. Thus, delivering miRs and miR inhibitors to the defect site is a potential treatment towards craniofacial bone defects. However, there are challenges in translation of basic research to clinics, including the efficiency, specificity, and efficacy of miR manipulation methods and the safety of miR delivery systems. In this review, we will compare miR oligonucleotides, mimics and antagomirs as therapeutic reagents to treat disease and regenerate tissues. Newer technology will be discussed as well as the efficiency and efficacy of using these technologies to express or inhibit miRs in treating and repairing oral tissues. Delivery of these molecules using extracellular vesicles and nanoparticles can achieve different results and depending on their composition will elicit specific effects. We will highlight the specificity, toxicity, stability, and effectiveness of several miR systems in regenerative medicine.


Asunto(s)
MicroARNs , MicroARNs/genética , MicroARNs/uso terapéutico , Medicina Regenerativa , Diferenciación Celular , Células Madre , Regeneración Ósea/genética
6.
J Oral Pathol Med ; 52(3): 263-270, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36715450

RESUMEN

OBJECTIVE: Ameloblastomas are a group of relatively common odontogenic tumors that frequently originate from the dental epithelium. These tumors are aggressive in nature and present as slow-growing painless cortical expansion of the jaw. Histologically, the follicular and plexiform subtypes constitute two-thirds of solid/multicystic ameloblastomas. The objective of this study was to understand the genetic architecture of follicular and plexiform ameloblastomas using deep whole-exome sequencing. METHODS: Archived formalin-fixed paraffin-embedded tissue blocks of follicular (n = 4) and plexiform (n = 6) ameloblastomas were retrieved and genomic DNAs were isolated from the tumor tissue dissected from the formalin-fixed paraffin-embedded block. The exomes were enriched using the Integrated DNA Technologies Exome Research Panel (IDT, Coralville, IA) and paired-end sequencing was completed on an Illumina NovaSeq 6000 with an average output of 20 GB of data resulting in a mean coverage of 400×. Variant analysis was completed using custom-developed software: Rapid Understanding of Nucleotide variant Effect Software and variant integration and knowledge interpretation in genomes. RESULTS: Our analyses focused on examining somatic variants (gnomAD minor allele frequency ≤1%) in genes found on an Food and Drug Administration -approved clinical cancer sequencing panel (FoundationOne®CDx). In follicular tumors, variants (>20% of the reads) were identified in BRAF, KMT2D, and ABL1 genes. In plexiform tumors, variants (>20% of the reads) were identified in ALK, BRAF, KRAS, KMT2D, SMO, KMT2A, and BRCA2 genes. Enrichment analysis showed a significant role of DNA repair genes in the development of these tumors. CONCLUSION: The variants identified in follicular and plexiform ameloblastomas were enriched in DNA-repair genes. The observed genetic heterogeneity in these ameloblastomas may contribute to the aggressive nature and recurrence risk of these tumors.


Asunto(s)
Ameloblastoma , Tumores Odontogénicos , Humanos , Ameloblastoma/genética , Ameloblastoma/patología , Proteínas Proto-Oncogénicas B-raf/genética , Heterogeneidad Genética , Tumores Odontogénicos/genética , Formaldehído
7.
Orthod Craniofac Res ; 26 Suppl 1: 39-47, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37073503

RESUMEN

OBJECTIVE: The objective of the study was to investigate differential gene expression between murine right and left maxilla-mandibular (MxMn) complexes. SETTING AND SAMPLE POPULATION: Wild-type (WT) C57BL/6 embryonic (E) day 14.5 (n = 3) and 18.5 (n = 3) murine embryos. METHODS: The E14.5 and 18.5 embryos were harvested and hemi-sectioned the MxMn complexes into right and left halves in the mid-sagittal plane. We isolated total RNA using Trizol reagent and further purified using the RNA-easy kit (QIAGEN). We confirmed equal expression of house-keeping genes in right and left halves using RT-PCR and then performed paired-end whole mRNA sequencing in LC Sciences (Houston, TX) followed by differential transcript analyses (>1 or <-1 log fold change; p < .05; q < .05; and FPKM >0.5 in 2/3 samples). The Mouse Genome Informatics and Online Mendelian Inheritance in Man databases as well as gnomAD constraint scores were used to prioritize differentially expressed transcripts. RESULTS: There were 19 upregulated and 19 downregulated transcripts at E14.5 and 8 upregulated and 17 downregulated transcripts at E18.5 time-points. These differentially expressed transcripts were statistically significant and shown to be associated with craniofacial phenotypes in mouse models. These transcripts also have significant gnomAD constraint scores and are enriched in biological processes critical for embryogenesis. CONCLUSIONS: We identified significant differential expression of transcripts between E14.5 and 18.5 murine right and left MxMn complexes. These findings when extrapolated to humans, they may provide a biological basis for facial asymmetry. Further experiments are required to validate these findings in murine models with craniofacial asymmetry.


Asunto(s)
Maxilar , Transcriptoma , Humanos , Animales , Ratones , Transcriptoma/genética , Ratones Endogámicos C57BL , Perfilación de la Expresión Génica , ARN
8.
Biochem Soc Trans ; 50(6): 1897-1909, 2022 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-36484693

RESUMEN

microRNAs (miRs) are small RNA molecules that regulate many cellular and developmental processes. They control gene expression pathways during specific developmental time points and are required for tissue homeostasis and stem cell maintenance. miRs as therapeutic reagents in tissue regeneration and repair hold great promise and new technologies are currently being designed to facilitate their expression or inhibition. Due to the large amount of miR research in cells and cancer many cellular processes and gene networks have been delineated however, their in vivo response can be different in complex tissues and organs. Specifically, this report will discuss animal developmental models to understand the role of miRs as well as xenograft, disease, and injury models. We will discuss the role of miRs in clinical studies including their diagnostic function, as well as their potential ability to correct craniofacial diseases.


Asunto(s)
MicroARNs , Animales , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Células Madre/metabolismo , Homeostasis
9.
Stem Cells ; 39(6): 761-775, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33529466

RESUMEN

The murine lower incisor ectodermal organ contains a single epithelial stem cell (SC) niche that provides epithelial progenitor cells to the continuously growing rodent incisor. The dental stem cell niche gives rise to several cell types and we demonstrate that the miR-200 family regulates these cell fates. The miR-200 family is highly enriched in the differentiated dental epithelium and absent in the stem cell niche. In this study, we inhibited the miR-200 family in developing murine embryos using new technology, resulting in an expanded epithelial stem cell niche and lack of cell differentiation. Inhibition of individual miRs within the miR-200 cluster resulted in differential developmental and cell morphology defects. miR-200 inhibition increased the expression of dental epithelial stem cell markers, expanded the stem cell niche and decreased progenitor cell differentiation. RNA-seq. identified miR-200 regulatory pathways involved in cell differentiation and compartmentalization of the stem cell niche. The miR-200 family regulates signaling pathways required for cell differentiation and cell cycle progression. The inhibition of miR-200 decreased the size of the lower incisor due to increased autophagy and cell death. New miR-200 targets demonstrate gene networks and pathways controlling cell differentiation and maintenance of the stem cell niche. This is the first report demonstrating how the miR-200 family is required for in vivo progenitor cell proliferation and differentiation.


Asunto(s)
Diferenciación Celular/genética , Proliferación Celular/genética , MicroARNs/genética , Nicho de Células Madre/genética , Animales , Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Células Epiteliales/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Ratones , MicroARNs/metabolismo , Nicho de Células Madre/fisiología , Células Madre/metabolismo
10.
Nature ; 534(7605): 119-23, 2016 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-27251288

RESUMEN

Myocardial infarction results in compromised myocardial function and heart failure owing to insufficient cardiomyocyte self-renewal. Unlike many vertebrates, mammalian hearts have only a transient neonatal renewal capacity. Reactivating primitive reparative ability in the mature mammalian heart requires knowledge of the mechanisms that promote early heart repair. By testing an established Hippo-deficient heart regeneration mouse model for factors that promote renewal, here we show that the expression of Pitx2 is induced in injured, Hippo-deficient ventricles. Pitx2-deficient neonatal mouse hearts failed to repair after apex resection, whereas adult mouse cardiomyocytes with Pitx2 gain-of-function efficiently regenerated after myocardial infarction. Genomic analyses indicated that Pitx2 activated genes encoding electron transport chain components and reactive oxygen species scavengers. A subset of Pitx2 target genes was cooperatively regulated with the Hippo pathway effector Yap. Furthermore, Nrf2, a regulator of the antioxidant response, directly regulated the expression and subcellular localization of Pitx2. Pitx2 mutant myocardium had increased levels of reactive oxygen species, while antioxidant supplementation suppressed the Pitx2 loss-of-function phenotype. These findings reveal a genetic pathway activated by tissue damage that is essential for cardiac repair.


Asunto(s)
Antioxidantes/metabolismo , Lesiones Cardíacas/metabolismo , Proteínas de Homeodominio/metabolismo , Infarto del Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Regeneración/fisiología , Factores de Transcripción/metabolismo , Cicatrización de Heridas/fisiología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Animales Recién Nacidos , Antioxidantes/farmacología , Proteínas de Ciclo Celular , Modelos Animales de Enfermedad , Transporte de Electrón/efectos de los fármacos , Transporte de Electrón/genética , Femenino , Depuradores de Radicales Libres/metabolismo , Lesiones Cardíacas/genética , Lesiones Cardíacas/patología , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/metabolismo , Vía de Señalización Hippo , Proteínas de Homeodominio/genética , Masculino , Ratones , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Miocardio/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Factor 2 Relacionado con NF-E2/metabolismo , Fosfoproteínas/metabolismo , Proteínas Serina-Treonina Quinasas/deficiencia , Especies Reactivas de Oxígeno/metabolismo , Regeneración/efectos de los fármacos , Regeneración/genética , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Cicatrización de Heridas/efectos de los fármacos , Cicatrización de Heridas/genética , Proteínas Señalizadoras YAP , Proteína del Homeodomínio PITX2
11.
Int J Mol Sci ; 23(10)2022 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-35628416

RESUMEN

This study was carried out to quantitate the expression levels of microRNA-17, -19a, -34a, -155, and -210 (miRs) expressed in nine clear cell renal cell carcinoma (ccRCC) and one chromophobe renal cell carcinoma cell line with and without sarcomatoid differentiation, and in six primary kidney tumors with matching normal kidney tissues. The data in the five non-sarcomatoid ccRCC cell lines-RC2, CAKI-1, 786-0, RCC4, and RCC4/VHL-and in the four ccRCC with sarcomatoid differentiation-RCJ41T1, RCJ41T2, RCJ41M, and UOK-127-indicated that miR-17 and -19a were expressed at lower levels relative to miR-34a, -155, and -210. Compared with RPTEC normal epithelial cells, miR-34a, miR-155, and miR-210 were expressed at higher levels, independent of the sarcomatoid differentiation status and hypoxia-inducible factors 1α and 2α (HIFs) isoform expression. In the one chromophobe renal cell carcinoma cell line, namely, UOK-276 with sarcomatoid differentiation, and expressing tumor suppressor gene TP53, miR-34a, which is a tumor suppressor gene, was expressed at higher levels than miR-210, -155, -17, and -19a. The pilot results generated in six tumor biopsies with matching normal kidney tissues indicated that while the expression of miR-17 and -19a were similar to the normal tissue expression profile, miR-210, -155, -and 34a were expressed at a higher level. To confirm that differences in the expression levels of the five miRs in the six tumor biopsies were statistically significant, the acquisition of a larger sample size is required. Data previously generated in ccRCC cell lines demonstrating that miR-210, miR-155, and HIFs are druggable targets using a defined dose and schedule of selenium-containing molecules support the concept that simultaneous and concurrent downregulation of miR-210, miR-155, and HIFs, which regulate target genes associated with increased tumor angiogenesis and drug resistance, may offer the potential for the development of a novel mechanism-based strategy for the treatment of patients with advanced ccRCC.


Asunto(s)
Carcinoma de Células Renales , Neoplasias Renales , MicroARNs , Biopsia , Carcinoma de Células Renales/patología , Línea Celular Tumoral , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Renales/metabolismo , MicroARNs/metabolismo
12.
Dev Biol ; 458(2): 246-256, 2020 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-31765609

RESUMEN

In this study, we investigated the role of the transcription factor Six2 in palate development. Six2 was selected using the SysFACE tool to predict genes from the 2p21 locus, a region associated with clefting in humans by GWAS, that are likely to be involved in palatogenesis. We functionally validated the predicted role of Six2 in palatogenesis by showing that 22% of Six2 null embryos develop cleft palate. Six2 contributes to palatogenesis by promoting mesenchymal cell proliferation and regulating bone formation. The clefting phenotype in Six2-/- embryos is similar to Pax9 null embryos, so we examined the functional relationship of these two genes. Mechanistically, SIX2 binds to a PAX9 5' upstream regulatory element and activates PAX9 expression. In addition, we identified a human SIX2 coding variant (p.Gly264Glu) in a proband with cleft palate. We show this missense mutation affects the stability of the SIX2 protein and leads to decreased PAX9 expression. The low penetrance of clefting in the Six2 null mouse combined with the mutation in one patient with cleft palate underscores the potential combinatorial interactions of other genes in clefting. Our study demonstrates that Six2 interacts with the developmental gene regulatory network in the developing palate.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Factor de Transcripción PAX9/genética , Factores de Transcripción/metabolismo , Animales , Fisura del Paladar/embriología , Fisura del Paladar/genética , Anomalías Craneofaciales/embriología , Femenino , Regulación del Desarrollo de la Expresión Génica/genética , Genes Homeobox , Proteínas de Homeodominio/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Morfogénesis , Proteínas del Tejido Nervioso/metabolismo , Osteogénesis , Factor de Transcripción PAX9/metabolismo , Factores de Transcripción Paired Box , Hueso Paladar/metabolismo , Transducción de Señal/genética , Factores de Transcripción/genética
13.
Hum Mol Genet ; 28(10): 1726-1737, 2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-30689861

RESUMEN

Mutations in IRF6, TFAP2A and GRHL3 cause orofacial clefting syndromes in humans. However, Tfap2a and Grhl3 are also required for neurulation in mice. Here, we found that homeostasis of Irf6 is also required for development of the neural tube and associated structures. Over-expression of Irf6 caused exencephaly, a rostral neural tube defect, through suppression of Tfap2a and Grhl3 expression. Conversely, loss of Irf6 function caused a curly tail and coincided with a reduction of Tfap2a and Grhl3 expression in tail tissues. To test whether Irf6 function in neurulation was conserved, we sequenced samples obtained from human cases of spina bifida and anencephaly. We found two likely disease-causing variants in two samples from patients with spina bifida. Overall, these data suggest that the Tfap2a-Irf6-Grhl3 genetic pathway is shared by two embryologically distinct morphogenetic events that previously were considered independent during mammalian development. In addition, these data suggest new candidates to delineate the genetic architecture of neural tube defects and new therapeutic targets to prevent this common birth defect.


Asunto(s)
Proteínas de Unión al ADN/genética , Factores Reguladores del Interferón/genética , Neurulación/genética , Factor de Transcripción AP-2/genética , Factores de Transcripción/genética , Animales , Secuencia Conservada/genética , Regulación del Desarrollo de la Expresión Génica/genética , Humanos , Ratones , Mutación , Tubo Neural/crecimiento & desarrollo , Tubo Neural/patología , Defectos del Tubo Neural/genética , Defectos del Tubo Neural/patología , Transducción de Señal/genética , Disrafia Espinal/genética , Disrafia Espinal/patología
14.
Development ; 145(1)2018 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-29180573

RESUMEN

In mice, the incisors grow throughout the animal's life, and this continuous renewal is driven by dental epithelial and mesenchymal stem cells. Sox2 is a principal marker of the epithelial stem cells that reside in the mouse incisor stem cell niche, called the labial cervical loop, but relatively little is known about the role of the Sox2+ stem cell population. In this study, we show that conditional deletion of Sox2 in the embryonic incisor epithelium leads to growth defects and impairment of ameloblast lineage commitment. Deletion of Sox2 specifically in Sox2+ cells during incisor renewal revealed cellular plasticity that leads to the relatively rapid restoration of a Sox2-expressing cell population. Furthermore, we show that Lgr5-expressing cells are a subpopulation of dental Sox2+ cells that also arise from Sox2+ cells during tooth formation. Finally, we show that the embryonic and adult Sox2+ populations are regulated by distinct signalling pathways, which is reflected in their distinct transcriptomic signatures. Together, our findings demonstrate that a Sox2+ stem cell population can be regenerated from Sox2- cells, reinforcing its importance for incisor homeostasis.


Asunto(s)
Ameloblastos/metabolismo , Antígenos de Diferenciación/biosíntesis , Regulación del Desarrollo de la Expresión Génica , Incisivo/embriología , Factores de Transcripción SOXB1/biosíntesis , Células Madre/metabolismo , Ameloblastos/citología , Animales , Antígenos de Diferenciación/genética , Incisivo/citología , Ratones , Ratones Transgénicos , Factores de Transcripción SOXB1/genética , Células Madre/citología
15.
PLoS Genet ; 14(10): e1007675, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30286078

RESUMEN

The mechanisms that regulate post-natal growth of the craniofacial complex and that ultimately determine the size and shape of our faces are not well understood. Hippo signaling is a general mechanism to control tissue growth and organ size, and although it is known that Hippo signaling functions in neural crest specification and patterning during embryogenesis and before birth, its specific role in postnatal craniofacial growth remains elusive. We have identified the transcription factor FoxO6 as an activator of Hippo signaling regulating neonatal growth of the face. During late stages of mouse development, FoxO6 is expressed specifically in craniofacial tissues and FoxO6-/- mice undergo expansion of the face, frontal cortex, olfactory component and skull. Enlargement of the mandible and maxilla and lengthening of the incisors in FoxO6-/- mice are associated with increases in cell proliferation. In vitro and in vivo studies demonstrated that FoxO6 activates Lats1 expression, thereby increasing Yap phosphorylation and activation of Hippo signaling. FoxO6-/- mice have significantly reduced Hippo Signaling caused by a decrease in Lats1 expression and decreases in Shh and Runx2 expression, suggesting that Shh and Runx2 are also linked to Hippo signaling. In vitro, FoxO6 activates Hippo reporter constructs and regulates cell proliferation. Furthermore PITX2, a regulator of Hippo signaling is associated with Axenfeld-Rieger Syndrome causing a flattened midface and we show that PITX2 activates FoxO6 expression. Craniofacial specific expression of FoxO6 postnatally regulates Hippo signaling and cell proliferation. Together, these results identify a FoxO6-Hippo regulatory pathway that controls skull growth, odontogenesis and face morphology.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Desarrollo Maxilofacial/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Cráneo/crecimiento & desarrollo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Vía de Señalización Hippo , Proteínas de Homeodominio/metabolismo , Desarrollo Maxilofacial/genética , Ratones , Cresta Neural/citología , Tamaño de los Órganos , Fosforilación , Transducción de Señal , Cráneo/metabolismo , Factores de Transcripción/metabolismo , Proteína del Homeodomínio PITX2
16.
Development ; 143(22): 4115-4126, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27660324

RESUMEN

Sox2 marks dental epithelial stem cells (DESCs) in both mammals and reptiles, and in this article we demonstrate several Sox2 transcriptional mechanisms that regulate dental stem cell fate and incisor growth. Conditional Sox2 deletion in the oral and dental epithelium results in severe craniofacial defects, including impaired dental stem cell proliferation, arrested incisor development and abnormal molar development. The murine incisor develops initially but is absorbed independently of apoptosis owing to a lack of progenitor cell proliferation and differentiation. Tamoxifen-induced inactivation of Sox2 demonstrates the requirement of Sox2 for maintenance of the DESCs in adult mice. Conditional overexpression of Lef-1 in mice increases DESC proliferation and creates a new labial cervical loop stem cell compartment, which produces rapidly growing long tusk-like incisors, and Lef-1 epithelial overexpression partially rescues the tooth arrest in Sox2 conditional knockout mice. Mechanistically, Pitx2 and Sox2 interact physically and regulate Lef-1, Pitx2 and Sox2 expression during development. Thus, we have uncovered a Pitx2-Sox2-Lef-1 transcriptional mechanism that regulates DESC homeostasis and dental development.


Asunto(s)
Autorrenovación de las Células/genética , Proteínas de Homeodominio , Incisivo/embriología , Factor de Unión 1 al Potenciador Linfoide , Odontogénesis/genética , Factores de Transcripción SOXB1 , Células Madre/fisiología , Factores de Transcripción , Animales , Células Cultivadas , Embrión de Mamíferos , Epitelio/crecimiento & desarrollo , Epitelio/metabolismo , Regulación del Desarrollo de la Expresión Génica , Redes Reguladoras de Genes , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Incisivo/crecimiento & desarrollo , Incisivo/metabolismo , Factor de Unión 1 al Potenciador Linfoide/genética , Factor de Unión 1 al Potenciador Linfoide/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Unión Proteica , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteína del Homeodomínio PITX2
17.
Dev Biol ; 429(1): 44-55, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28746823

RESUMEN

The Iroquois genes (Irx) appear to regulate fundamental processes that lead to cell proliferation, differentiation, and maturation during development. In this report, the Iroquois homeobox 1 (Irx1) transcription factor was functionally disrupted using a LacZ insert and LacZ expression demonstrated stage-specific expression during embryogenesis. Irx1 is highly expressed in the brain, lung, digits, kidney, testis and developing teeth. Irx1 null mice are neonatal lethal and this lethality it due to pulmonary immaturity. Irx1-/- mice show delayed lung maturation characterized by defective surfactant protein secretion and Irx1 marks a population of SP-C expressing alveolar type II cells. Irx1 is specifically expressed in the outer enamel epithelium (OEE), stellate reticulum (SR) and stratum intermedium (SI) layers of the developing tooth. Irx1 mediates dental epithelial cell differentiation in the lower incisors resulting in delayed growth of the lower incisors. Irx1 is specifically and temporally expressed during developmental stages and we have focused on lung and dental development in this report. Irx1+ cells are unique to the development of the incisor outer enamel epithelium, patterning of Lef-1+ and Sox2+ cells as well as a new marker for lung alveolar type II cells. Mechanistically, Irx1 regulates Foxj1 and Sox9 to control cell differentiation during development.


Asunto(s)
Células Epiteliales Alveolares/citología , Diferenciación Celular , Esmalte Dental/citología , Células Epiteliales/citología , Células Epiteliales/metabolismo , Proteínas de Homeodominio/metabolismo , Factores de Transcripción/metabolismo , Células Epiteliales Alveolares/metabolismo , Animales , Animales Recién Nacidos , Cruzamientos Genéticos , Embrión de Mamíferos/metabolismo , Desarrollo Embrionario/genética , Femenino , Factores de Transcripción Forkhead/metabolismo , Regulación del Desarrollo de la Expresión Génica , Genotipo , Células HEK293 , Proteínas de Homeodominio/genética , Humanos , Incisivo/embriología , Incisivo/metabolismo , Factor de Unión 1 al Potenciador Linfoide/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Asociadas a Surfactante Pulmonar/metabolismo , Ratas , Factor de Transcripción SOX9/metabolismo , Factores de Transcripción SOXB1/metabolismo , Factores de Transcripción/genética
18.
Biochim Biophys Acta ; 1860(11 Pt B): 2613-8, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27288587

RESUMEN

BACKGROUND: Developmental dental anomalies are common forms of congenital defects. The molecular mechanisms of dental anomalies are poorly understood. Systematic approaches such as clustering genes based on similar expression patterns could identify novel genes involved in dental anomalies and provide a framework for understanding molecular regulatory mechanisms of these genes during tooth development (odontogenesis). METHODS: A python package (pySAPC) of sparse affinity propagation clustering algorithm for large datasets was developed. Whole genome pair-wise similarity was calculated based on expression pattern similarity based on 45 microarrays of several stages during odontogenesis. RESULTS: pySAPC identified 743 gene clusters based on expression pattern similarity during mouse tooth development. Three clusters are significantly enriched for genes associated with dental anomalies (with FDR <0.1). The three clusters of genes have distinct expression patterns during odontogenesis. CONCLUSIONS: Clustering genes based on similar expression profiles recovered several known regulatory relationships for genes involved in odontogenesis, as well as many novel genes that may be involved with the same genetic pathways as genes that have already been shown to contribute to dental defects. GENERAL SIGNIFICANCE: By using sparse similarity matrix, pySAPC use much less memory and CPU time compared with the original affinity propagation program that uses a full similarity matrix. This python package will be useful for many applications where dataset(s) are too large to use full similarity matrix. This article is part of a Special Issue entitled "System Genetics" Guest Editor: Dr. Yudong Cai and Dr. Tao Huang.


Asunto(s)
Expresión Génica/genética , Genoma/genética , Familia de Multigenes/genética , Odontogénesis/genética , Algoritmos , Animales , Análisis por Conglomerados , Bases de Datos Genéticas , Perfilación de la Expresión Génica/métodos , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Diente/crecimiento & desarrollo
19.
Hum Mol Genet ; 24(8): 2330-48, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25556186

RESUMEN

T-box transcription factor TBX1 is the major candidate gene for 22q11.2 deletion syndrome (22q11.2DS, DiGeorge syndrome/Velo-cardio-facial syndrome), whose phenotypes include craniofacial malformations such as dental defects and cleft palate. In this study, Tbx1 was conditionally deleted or over-expressed in the oral and dental epithelium to establish its role in odontogenesis and craniofacial developmental. Tbx1 lineage tracing experiments demonstrated a specific region of Tbx1-positive cells in the labial cervical loop (LaCL, stem cell niche). We found that Tbx1 conditional knockout (Tbx1(cKO)) mice featured microdontia, which coincides with decreased stem cell proliferation in the LaCL of Tbx1(cKO) mice. In contrast, Tbx1 over-expression increased dental epithelial progenitor cells in the LaCL. Furthermore, microRNA-96 (miR-96) repressed Tbx1 expression and Tbx1 repressed miR-96 expression, suggesting that miR-96 and Tbx1 work in a regulatory loop to maintain the correct levels of Tbx1. Cleft palate was observed in both conditional knockout and over-expression mice, consistent with the craniofacial/tooth defects associated with TBX1 deletion and the gene duplication that leads to 22q11.2DS. The biochemical analyses of TBX1 human mutations demonstrate functional differences in their transcriptional regulation of miR-96 and co-regulation of PITX2 activity. TBX1 interacts with PITX2 to negatively regulate PITX2 transcriptional activity and the TBX1 N-terminus is required for its repressive activity. Overall, our results indicate that Tbx1 regulates the proliferation of dental progenitor cells and craniofacial development through miR-96-5p and PITX2. Together, these data suggest a new molecular mechanism controlling pathogenesis of dental anomalies in human 22q11.2DS.


Asunto(s)
Proliferación Celular , Síndrome de DiGeorge/metabolismo , Huesos Faciales/metabolismo , MicroARNs/metabolismo , Proteínas de Dominio T Box/metabolismo , Diente/metabolismo , Animales , Anomalías Craneofaciales , Síndrome de DiGeorge/embriología , Síndrome de DiGeorge/genética , Síndrome de DiGeorge/fisiopatología , Huesos Faciales/embriología , Femenino , Regulación del Desarrollo de la Expresión Génica , Humanos , Masculino , Ratones , MicroARNs/genética , Regiones Promotoras Genéticas , Unión Proteica , Proteínas de Dominio T Box/genética , Diente/embriología
20.
Hum Mol Genet ; 24(14): 3895-907, 2015 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-25652407

RESUMEN

Three common diseases, isolated cleft lip and cleft palate (CLP), hypothyroidism and thyroid cancer all map to the FOXE1 locus, but causative variants have yet to be identified. In patients with CLP, the frequency of coding mutations in FOXE1 fails to account for the risk attributable to this locus, suggesting that the common risk alleles reside in nearby regulatory elements. Using a combination of zebrafish and mouse transgenesis, we screened 15 conserved non-coding sequences for enhancer activity, identifying three that regulate expression in a tissue specific pattern consistent with endogenous foxe1 expression. These three, located -82.4, -67.7 and +22.6 kb from the FOXE1 start codon, are all active in the oral epithelium or branchial arches. The -67.7 and +22.6 kb elements are also active in the developing heart, and the -67.7 kb element uniquely directs expression in the developing thyroid. Within the -67.7 kb element is the SNP rs7850258 that is associated with all three diseases. Quantitative reporter assays in oral epithelial and thyroid cell lines show that the rs7850258 allele (G) associated with CLP and hypothyroidism has significantly greater enhancer activity than the allele associated with thyroid cancer (A). Moreover, consistent with predicted transcription factor binding differences, the -67.7 kb element containing rs7850258 allele G is significantly more responsive to both MYC and ARNT than allele A. By demonstrating that this common non-coding variant alters FOXE1 expression, we have identified at least in part the functional basis for the genetic risk of these seemingly disparate disorders.


Asunto(s)
Labio Leporino/genética , Fisura del Paladar/genética , Factores de Transcripción Forkhead/genética , Hipotiroidismo/genética , Polimorfismo de Nucleótido Simple , Neoplasias de la Tiroides/genética , Alelos , Animales , Línea Celular , Clonación Molecular , Células Epiteliales/citología , Células Epiteliales/metabolismo , Epitelio/metabolismo , Factores de Transcripción Forkhead/metabolismo , Regulación del Desarrollo de la Expresión Génica , Sitios Genéticos , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Ratones , Ratones Transgénicos , Hueso Paladar/metabolismo , Regiones Promotoras Genéticas , Glándula Tiroides/citología , Glándula Tiroides/metabolismo , Pez Cebra/embriología , Pez Cebra/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA