Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 95
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Development ; 149(17)2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-35950911

RESUMEN

Coordinated migration of the mesoderm is essential for accurate organization of the body plan during embryogenesis. However, little is known about how mesoderm migration influences posterior neural tube closure in mammals. Here, we show that spinal neural tube closure and lateral migration of the caudal paraxial mesoderm depend on transmembrane protein 132A (TMEM132A), a single-pass type I transmembrane protein, the function of which is not fully understood. Our study in Tmem132a-null mice and cell models demonstrates that TMEM132A regulates several integrins and downstream integrin pathway activation as well as cell migration behaviors. Our data also implicates mesoderm migration in elevation of the caudal neural folds and successful closure of the caudal neural tube. These results suggest a requirement for paraxial mesodermal cell migration during spinal neural tube closure, disruption of which may lead to spina bifida.


Asunto(s)
Proteínas de la Membrana , Defectos del Tubo Neural , Tubo Neural , Animales , Integrinas/metabolismo , Proteínas de la Membrana/genética , Mesodermo/metabolismo , Ratones , Ratones Noqueados , Tubo Neural/metabolismo , Defectos del Tubo Neural/genética , Defectos del Tubo Neural/metabolismo
2.
Genesis ; 62(3): e23602, 2024 06.
Artículo en Inglés | MEDLINE | ID: mdl-38721990

RESUMEN

Cilia play a key role in the regulation of signaling pathways required for embryonic development, including the proper formation of the neural tube, the precursor to the brain and spinal cord. Forward genetic screens were used to generate mouse lines that display neural tube defects (NTD) and secondary phenotypes useful in interrogating function. We describe here the L3P mutant line that displays phenotypes of disrupted Sonic hedgehog signaling and affects the initiation of cilia formation. A point mutation was mapped in the L3P line to the gene Rsg1, which encodes a GTPase-like protein. The mutation lies within the GTP-binding pocket and disrupts the highly conserved G1 domain. The mutant protein and other centrosomal and IFT proteins still localize appropriately to the basal body of cilia, suggesting that RSG1 GTPase activity is not required for basal body maturation but is needed for a downstream step in axonemal elongation.


Asunto(s)
Cilios , Tubo Neural , Animales , Ratones , Cilios/metabolismo , Cilios/genética , Proteínas Hedgehog/metabolismo , Proteínas Hedgehog/genética , Tubo Neural/embriología , Tubo Neural/metabolismo , Defectos del Tubo Neural/genética , Defectos del Tubo Neural/metabolismo , Mutación Puntual , Transducción de Señal
3.
Development ; 147(22)2020 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-33214242

RESUMEN

Disruptions in neural tube (NT) closure result in neural tube defects (NTDs). To understand the molecular processes required for mammalian NT closure, we investigated the role of Snx3, a sorting nexin gene. Snx3-/- mutant mouse embryos display a fully-penetrant cranial NTD. In vivo, we observed decreased canonical WNT target gene expression in the cranial neural epithelium of the Snx3-/- embryos and a defect in convergent extension of the neural epithelium. Snx3-/- cells show decreased WNT secretion, and live cell imaging reveals aberrant recycling of the WNT ligand-binding protein WLS and mis-trafficking to the lysosome for degradation. The importance of SNX3 in WNT signaling regulation is demonstrated by rescue of NT closure in Snx3-/- embryos with a WNT agonist. The potential for SNX3 to function in human neurulation is revealed by a point mutation identified in an NTD-affected individual that results in functionally impaired SNX3 that does not colocalize with WLS and the degradation of WLS in the lysosome. These data indicate that Snx3 is crucial for NT closure via its role in recycling WLS in order to control levels of WNT signaling.


Asunto(s)
Lisosomas/metabolismo , Defectos del Tubo Neural/embriología , Tubo Neural/embriología , Receptores Acoplados a Proteínas G/metabolismo , Nexinas de Clasificación/metabolismo , Vía de Señalización Wnt , Animales , Humanos , Lisosomas/genética , Lisosomas/patología , Ratones , Ratones Noqueados , Tubo Neural/patología , Defectos del Tubo Neural/genética , Defectos del Tubo Neural/patología , Receptores Acoplados a Proteínas G/genética , Nexinas de Clasificación/genética
4.
Annu Rev Genet ; 48: 583-611, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25292356

RESUMEN

The formation of the embryonic brain and spinal cord begins as the neural plate bends to form the neural folds, which meet and adhere to close the neural tube. The neural ectoderm and surrounding tissues also coordinate proliferation, differentiation, and patterning. This highly orchestrated process is susceptible to disruption, leading to neural tube defects (NTDs), a common birth defect. Here, we highlight genetic and epigenetic contributions to neural tube closure. We describe an online database we created as a resource for researchers, geneticists, and clinicians. Neural tube closure is sensitive to environmental influences, and we discuss disruptive causes, preventative measures, and possible mechanisms. New technologies will move beyond candidate genes in small cohort studies toward unbiased discoveries in sporadic NTD cases. This will uncover the genetic complexity of NTDs and critical gene-gene interactions. Animal models can reveal the causative nature of genetic variants, the genetic interrelationships, and the mechanisms underlying environmental influences.


Asunto(s)
Encéfalo/crecimiento & desarrollo , Epigénesis Genética , Tubo Neural/crecimiento & desarrollo , Médula Espinal/crecimiento & desarrollo , Animales , Encéfalo/embriología , Femenino , Cresta Neural/embriología , Cresta Neural/crecimiento & desarrollo , Placa Neural/embriología , Placa Neural/crecimiento & desarrollo , Tubo Neural/embriología , Médula Espinal/embriología
5.
Dev Dyn ; 250(5): 732-744, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33378081

RESUMEN

BACKGROUND: The transcription factor Grainyhead-like 3 (GRHL3) has multiple roles in a variety of tissues during development including epithelial patterning and actin cytoskeletal regulation. During neural tube closure (NTC) in the mouse embryo, GRHL3 is expressed and functions in the non-neural ectoderm (NNE). Two important functions of GRHL3 are regulating the actin cytoskeleton during NTC and regulating the boundary between the NNE and neural ectoderm. However, an open question that remains is whether these functions explain the caudally restricted neural tube defect (NTD) of spina bifida observed in Grhl3 mutants. RESULTS: Using scanning electron microscopy and immunofluorescence based imaging on Grhl3 mutants and wildtype controls, we show that GRHL3 is dispensable for NNE identity or epithelial maintenance in the caudal NNE but is needed for regulation of cellular protrusions during NTC. Grhl3 mutants have decreased lamellipodia relative to wildtype embryos during caudal NTC, first observed at the onset of delays when lamellipodia become prominent in wildtype embryos. At the axial level of NTD, half of the mutants show increased and disorganized filopodia and half lack cellular protrusions. CONCLUSION: These data suggest that altered cellular protrusions during NTC contribute to the etiology of NTD in Grhl3 mutants.


Asunto(s)
Extensiones de la Superficie Celular , Proteínas de Unión al ADN/fisiología , Ectodermo/fisiología , Tubo Neural/ultraestructura , Neurulación , Factores de Transcripción/fisiología , Animales , Ectodermo/ultraestructura , Femenino , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados
6.
Genesis ; 59(11): e23455, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34665506

RESUMEN

Neural tube defects (NTDs) are among the most common birth defects, with a prevalence of close to 19 per 10,000 births worldwide. The etiology of NTDs is complex involving the interplay of genetic and environmental factors. Since nutrient deficiency is a risk factor and dietary changes are the major preventative measure to reduce the risk of NTDs, a more detailed understanding of how common micronutrient imbalances contribute to NTDs is crucial. While folic acid has been the most discussed environmental factor due to the success that population-wide fortification has had on prevention of NTDs, folic acid supplementation does not prevent all NTDs. The imbalance of several other micronutrients has been implicated as risks for NTDs by epidemiological studies and in vivo studies in animal models. In this review, we highlight recent literature deciphering the multifactorial mechanisms underlying NTDs with an emphasis on mouse and human data. Specifically, we focus on advances in our understanding of how too much or too little retinoic acid, zinc, and iron alter gene expression and cellular processes contributing to the pathobiology of NTDs. Synthesis of the discussed literature reveals common cellular phenotypes found in embryos with NTDs resulting from several micronutrient imbalances. The goal is to combine knowledge of these common cellular phenotypes with mechanisms underlying micronutrient imbalances to provide insights into possible new targets for preventative measures against NTDs.


Asunto(s)
Micronutrientes/metabolismo , Defectos del Tubo Neural/metabolismo , Animales , Interacción Gen-Ambiente , Humanos , Defectos del Tubo Neural/genética
7.
Hum Mutat ; 42(4): 392-407, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33382518

RESUMEN

Idiopathic scoliosis (IS) is a spinal disorder affecting up to 3% of otherwise healthy children. IS has a strong familial genetic component and is believed to be genetically complex due to significant variability in phenotype and heritability. Previous studies identified putative loci and variants possibly contributing to IS susceptibility, including within extracellular matrix, cilia, and actin networks, but the genetic architecture and underlying mechanisms remain unresolved. Here, we used whole-exome sequencing from three affected individuals in a multigenerational family with IS and identified 19 uncommon variants (minor allele frequency < 0.05). Genotyping of additional family members identified a candidate heterozygous variant (H1115Q, G>C, rs142032413) within the ciliary gene KIF7, a regulator within the hedgehog (Hh) signaling pathway. Resequencing of the second cohort of unrelated IS individuals and controls identified several severe mutations in KIF7 in affected individuals only. Subsequently, we generated a mutant zebrafish model of kif7 using CRISPR-Cas9. kif7co63/co63 zebrafish displayed severe scoliosis, presenting in juveniles and progressing through adulthood. We observed no deformities in the brain, Reissner fiber, or central canal cilia in kif7co63/co63 embryos, although alterations were seen in Hh pathway gene expression. This study suggests defects in KIF7-dependent Hh signaling, which may drive pathogenesis in a subset of individuals with IS.


Asunto(s)
Cinesinas , Escoliosis , Pez Cebra , Animales , Cilios/metabolismo , Humanos , Cinesinas/genética , Mutación , Escoliosis/genética , Pez Cebra/genética , Proteínas de Pez Cebra
8.
Dev Biol ; 464(1): 24-34, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32446700

RESUMEN

Development of the craniofacial structures requires the precise differentiation of cranial neural crest cells into osteoblasts or chondrocytes. Here, we explore the epigenetic and non-epigenetic mechanisms that are required for the development of craniofacial chondrocytes. We previously demonstrated that the acetyltransferase activity of the highly conserved acetyltransferase GCN5, or KAT2A, is required for murine craniofacial development. We show that Gcn5 is required cell autonomously in the cranial neural crest. Moreover, GCN5 is required for chondrocyte development following the arrival of the cranial neural crest within the pharyngeal arches. Using a combination of in vivo and in vitro inhibition of GCN5 acetyltransferase activity, we demonstrate that GCN5 is a potent activator of chondrocyte maturation, acting to control chondrocyte maturation and size increase during pre-hypertrophic maturation to hypertrophic chondrocytes. Rather than acting as an epigenetic regulator of histone H3K9 acetylation, our findings suggest GCN5 primarily acts as a non-histone acetyltransferase to regulate chondrocyte development. Here, we investigate the contribution of GCN5 acetylation to the activity of the mTORC1 pathway. Our findings indicate that GCN5 acetylation is required for activation of this pathway, either via direct activation of mTORC1 or through indirect mechanisms. We also investigate one possibility of how mTORC1 activity is regulated through RAPTOR acetylation, which is hypothesized to enhance mTORC1 downstream phosphorylation. This study contributes to our understanding of the specificity of acetyltransferases, and the cell type specific roles in which these enzymes function.


Asunto(s)
Movimiento Celular , Condrocitos/enzimología , Transducción de Señal , Cráneo/embriología , Factores de Transcripción p300-CBP/metabolismo , Acetilación , Animales , Condrocitos/citología , Histonas/genética , Histonas/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones , Ratones Transgénicos , Cresta Neural/citología , Cresta Neural/embriología , Cráneo/citología , Factores de Transcripción p300-CBP/genética
9.
Development ; 145(24)2018 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-30545932

RESUMEN

Micronutrition is essential for neural tube closure, and zinc deficiency is associated with human neural tube defects. Here, we modeled zinc deficiency in mouse embryos, and used live imaging and molecular studies to determine how zinc deficiency affects neural tube closure. Embryos cultured with the zinc chelator TPEN failed to close the neural tube and showed excess apoptosis. TPEN-induced p53 protein stabilization in vivo and in neuroepithelial cell cultures and apoptosis was dependent on p53. Mechanistically, zinc deficiency resulted in disrupted interaction between p53 and the zinc-dependent E3 ubiquitin ligase Mdm2, and greatly reduced p53 ubiquitylation. Overexpression of human CHIP, a zinc-independent E3 ubiquitin ligase that targets p53, relieved TPEN-induced p53 stabilization and reduced apoptosis. Expression of p53 pro-apoptotic target genes was upregulated by zinc deficiency. Correspondingly, embryos cultured with p53 transcriptional activity inhibitor pifithrin-α could overcome TPEN-induced apoptosis and failure of neural tube closure. Our studies indicate that zinc deficiency disrupts neural tube closure through decreased p53 ubiquitylation, increased p53 stabilization and excess apoptosis.


Asunto(s)
Defectos del Tubo Neural/metabolismo , Defectos del Tubo Neural/patología , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitinación , Zinc/deficiencia , Animales , Apoptosis , Línea Celular , Ratones , Mitocondrias/metabolismo , Modelos Biológicos , Tubo Neural/anomalías , Tubo Neural/patología , Células Neuroepiteliales/metabolismo , Transducción de Señal , Transcripción Genética , Activación Transcripcional/genética
10.
Genes Dev ; 26(1): 69-81, 2012 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-22215812

RESUMEN

Hirschsprung disease (HSCR) is caused by a reduction of enteric neural crest cells (ENCCs) in the gut and gastrointestinal blockage. Knowledge of the genetics underlying HSCR is incomplete, particularly genes that control cellular behaviors of ENCC migration. Here we report a novel regulator of ENCC migration in mice. Disruption of the Phactr4 gene causes an embryonic gastrointestinal defect due to colon hypoganglionosis, which resembles human HSCR. Time-lapse imaging of ENCCs within the embryonic gut demonstrates a collective cell migration defect. Mutant ENCCs show undirected cellular protrusions and disrupted directional and chain migration. Phactr4 acts cell-autonomously in ENCCs and colocalizes with integrin and cofilin at cell protrusions. Mechanistically, we show that Phactr4 negatively regulates integrin signaling through the RHO/ROCK pathway and coordinates protein phosphatase 1 (PP1) with cofilin activity to regulate cytoskeletal dynamics. Strikingly, lamellipodia formation and in vivo ENCC chain migration defects are rescued by inhibition of ROCK or integrin function. Our results demonstrate a previously unknown pathway in ENCC collective migration in vivo and provide new candidate genes for human genetic studies of HSCR.


Asunto(s)
Factores Despolimerizantes de la Actina/metabolismo , Sistema Nervioso Entérico , Integrinas/fisiología , Cresta Neural , Proteínas Nucleares/metabolismo , Proteína Fosfatasa 1/metabolismo , Transducción de Señal , Animales , Movimiento Celular , Células Cultivadas , Proteínas del Citoesqueleto , Sistema Nervioso Entérico/citología , Sistema Nervioso Entérico/embriología , Tracto Gastrointestinal/citología , Tracto Gastrointestinal/embriología , Ratones , Cresta Neural/citología , Cresta Neural/embriología , Proteínas Nucleares/genética , Proteína Fosfatasa 1/genética
11.
Genes Dev ; 26(8): 803-15, 2012 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-22508726

RESUMEN

How self-renewal versus differentiation of neural progenitor cells is temporally controlled during early development remains ill-defined. We show that mouse Lin41 (mLin41) is highly expressed in neural progenitor cells and its expression declines during neural differentiation. Loss of mLin41 function in mice causes reduced proliferation and premature differentiation of embryonic neural progenitor cells. mLin41 was recently implicated as the E3 ubiquitin ligase that mediates degradation of Argonaute 2 (AGO2), a key effector of the microRNA pathway. However, our mechanistic studies of neural progenitor cells indicate mLin41 is not required for AGO2 ubiquitination or stability. Instead, mLin41-deficient neural progenitors exhibit hyposensitivity for fibroblast growth factor (FGF) signaling. We show that mLin41 promotes FGF signaling by directly binding to and enhancing the stability of Shc SH2-binding protein 1 (SHCBP1) and that SHCBP1 is an important component of FGF signaling in neural progenitor cells. Thus, mLin41 acts as a temporal regulator to promote neural progenitor cell maintenance, not via the regulation of AGO2 stability, but through FGF signaling.


Asunto(s)
Diferenciación Celular , Factores de Crecimiento de Fibroblastos/metabolismo , Células-Madre Neurales/citología , Factores de Transcripción/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Proteínas Argonautas/metabolismo , Línea Celular , Proliferación Celular , Ratones , Células-Madre Neurales/enzimología , Estabilidad Proteica , Transducción de Señal , Factores de Transcripción/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación
12.
Development ; 143(7): 1192-204, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-26903501

RESUMEN

The transcription factor grainyhead-like 2 (GRHL2) is expressed in non-neural ectoderm (NNE) and Grhl2 loss results in fully penetrant cranial neural tube defects (NTDs) in mice. GRHL2 activates expression of several epithelial genes; however, additional molecular targets and functional processes regulated by GRHL2 in the NNE remain to be determined, as well as the underlying cause of the NTDs in Grhl2 mutants. Here, we find that Grhl2 loss results in abnormal mesenchymal phenotypes in the NNE, including aberrant vimentin expression and increased cellular dynamics that affects the NNE and neural crest cells. The resulting loss of NNE integrity contributes to an inability of the cranial neural folds to move toward the midline and results in NTD. Further, we identified Esrp1, Sostdc1, Fermt1, Tmprss2 and Lamc2 as novel NNE-expressed genes that are downregulated in Grhl2 mutants. Our in vitro assays show that they act as suppressors of the epithelial-to-mesenchymal transition (EMT). Thus, GRHL2 promotes the epithelial nature of the NNE during the dynamic events of neural tube formation by both activating key epithelial genes and actively suppressing EMT through novel downstream EMT suppressors.


Asunto(s)
Transición Epitelial-Mesenquimal/genética , Cresta Neural/embriología , Tubo Neural/embriología , Factores de Transcripción/genética , Animales , Cadherinas/metabolismo , Línea Celular , Movimiento Celular , Proteínas de Unión al ADN/metabolismo , Ectodermo/embriología , Ectodermo/metabolismo , Técnicas de Cultivo de Embriones , Mesodermo/citología , Mesodermo/embriología , Ratones , Ratones Noqueados , Defectos del Tubo Neural/genética , Neurulación/fisiología , Factores de Transcripción/biosíntesis , Vimentina/biosíntesis
13.
J Surg Res ; 235: 227-236, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30691800

RESUMEN

BACKGROUND: Myelomeningocele (MMC) results in lifelong neurologic and functional deficits. Currently, prenatal repair of MMC closes the defect, resulting in a 50% reduction in postnatal ventriculoperitoneal shunting. However, this invasive fetal surgery is associated with significant morbidities to mother and baby. We have pioneered a novel reverse thermal gel (RTG) to cover MMC defects in a minimally invasive manner. Here, we test in-vitro RTG long-term stability in amniotic fluid and in vivo application in the Grainy head-like 3 (Grhl3) mouse MMC model. MATERIALS AND METHODS: RTG stability in amniotic fluid (in-vitro) was monitored for 6 mo and measured using gel permeation chromatography and solution-gel transition temperature (lower critical solution temperature). E16.5 Grhl3 mouse fetuses were injected with the RTG or saline and harvested on E19.5. Tissue was assessed for RTG coverage of the gross defect and inflammatory response by immunohistochemistry for macrophages. RESULTS: Polymer backbone molecular weight and lower critical solution temperature remain stable in amniotic fluid after 6 mo. Needle injection over the MMC of Grhl3 fetuses successfully forms a stable gel that covers the entire defect. On harvest, some animals demonstrate >50% RTG coverage. RTG injection is not associated with inflammation. CONCLUSIONS: Our results demonstrate that the RTG is a promising candidate for a minimally invasive approach to patch MMC. We are now poised to test our RTG patch in the large preclinical ovine model used to evaluate prenatal repair of MMC.


Asunto(s)
Materiales Biocompatibles/uso terapéutico , Fetoscopía , Meningomielocele/cirugía , Resinas Acrílicas , Animales , Femenino , Masculino , Ensayo de Materiales , Ratones , Procedimientos Quirúrgicos Mínimamente Invasivos , Embarazo
14.
J Neurosci ; 37(10): 2565-2579, 2017 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-28154153

RESUMEN

Diencephalic defects underlie an array of neurological diseases. Previous studies have suggested that retinoic acid (RA) signaling is involved in diencephalic development at late stages of embryonic development, but its roles and mechanisms of action during early neural development are still unclear. Here we demonstrate that mice lacking enzymatic activity of the acetyltransferase GCN5 ((Gcn5hat/hat )), which were previously characterized with respect to their exencephalic phenotype, exhibit significant diencephalic expansion, decreased diencephalic RA signaling, and increased diencephalic WNT and SHH signaling. Using a variety of molecular biology techniques in both cultured neuroepithelial cells treated with a GCN5 inhibitor and forebrain tissue from (Gcn5hat/hat ) embryos, we demonstrate that GCN5, RARα/γ, and the poorly characterized protein TACC1 form a complex in the nucleus that binds specific retinoic acid response elements in the absence of RA. Furthermore, RA triggers GCN5-mediated acetylation of TACC1, which results in dissociation of TACC1 from retinoic acid response elements and leads to transcriptional activation of RA target genes. Intriguingly, RA signaling defects caused by in vitro inhibition of GCN5 can be rescued through RA-dependent mechanisms that require RARß. Last, we demonstrate that the diencephalic expansion and transcriptional defects seen in (Gcn5hat/hat ) mutants can be rescued with gestational RA supplementation, supporting a direct link between GCN5, TACC1, and RA signaling in the developing diencephalon. Together, our studies identify a novel, nonhistone substrate for GCN5 whose modification regulates a previously undescribed, tissue-specific mechanism of RA signaling that is required to restrict diencephalic size during early forebrain development.SIGNIFICANCE STATEMENT Changes in diencephalic size and shape, as well as SNPs associated with retinoic acid (RA) signaling-associated genes, have been linked to neuropsychiatric disorders. However, the mechanisms that regulate diencephalic morphogenesis and the involvement of RA signaling in this process are poorly understood. Here we demonstrate a novel role of the acetyltransferase GCN5 in a previously undescribed mechanism of RA signaling in the developing forebrain that is required to maintain the appropriate size of the diencephalon. Together, our experiments identify a novel nonhistone substrate of GCN5, highlight an essential role for both GCN5 and RA signaling in early diencephalic development, and elucidate a novel molecular regulatory mechanism for RA signaling that is specific to the developing forebrain.


Asunto(s)
Diencéfalo/anatomía & histología , Diencéfalo/metabolismo , Transducción de Señal/fisiología , Tretinoina/metabolismo , Factores de Transcripción p300-CBP/metabolismo , Animales , Diencéfalo/embriología , Activación Enzimática , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Tamaño de los Órganos/fisiología
15.
Hum Mutat ; 39(4): 550-562, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29297599

RESUMEN

Rare variants are considered underlying causes of complex diseases. The complex and severe group of disorders called neural tube defects (NTDs) results from failure of the neural tube to close during early embryogenesis. Neural tube closure requires the coordination of numerous signaling pathways, including the precise regulation of retinoic acid (RA) concentration, which is controlled by enzymes involved in RA synthesis and degradation. Here, we used a case-control mutation screen study to reveal rare variants in retinoid-related genes in a Han Chinese NTD population by sequencing six genes in 355 NTD cases and 225 controls. More specific rare variants were found in exonic and upstream regions in NTD cases. The RA-responsive genes CYP26A1, CRABP1, and ALDH1A2 harbored NTD-specific rare variants in their upstream regions. Unexpectedly, the majority of missense variants in NTD cases were found in CYP26B1, which encodes a RA degradation enzyme, whereas no missense variants in this gene were found in controls. Functional analysis indicated that the CYP26B1 NTD variants were inefficient in the degradation of RA using assays of RA-induced transcription and RA-initiated neuronal differentiation. Our study supports the contribution of rare variants in RA-related genes to the etiology of human NTDs.


Asunto(s)
Defectos del Tubo Neural/genética , Receptores de Ácido Retinoico/genética , Retinal-Deshidrogenasa/genética , Ácido Retinoico 4-Hidroxilasa/genética , Tretinoina/metabolismo , Familia de Aldehído Deshidrogenasa 1 , Estudios de Casos y Controles , Niño , Preescolar , China/epidemiología , Estudios de Cohortes , Desarrollo Embrionario , Humanos , Lactante , Recién Nacido , Mutación
16.
Development ; 142(9): 1616-27, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25922525

RESUMEN

Neural progenitor cells (NPCs) have distinct proliferation capacities at different stages of brain development. Lin28 is an RNA-binding protein with two homologs in mice: Lin28a and Lin28b. Here we show that Lin28a/b are enriched in early NPCs and their expression declines during neural differentiation. Lin28a single-knockout mice show reduced NPC proliferation, enhanced cell cycle exit and a smaller brain, whereas mice lacking both Lin28a alleles and one Lin28b allele display similar but more severe phenotypes. Ectopic expression of Lin28a in mice results in increased NPC proliferation, NPC numbers and brain size. Mechanistically, Lin28a physically and functionally interacts with Imp1 (Igf2bp1) and regulates Igf2-mTOR signaling. The function of Lin28a/b in NPCs could be attributed, at least in part, to the regulation of their mRNA targets that encode Igf1r and Hmga2. Thus, Lin28a and Lin28b have overlapping functions in temporally regulating NPC proliferation during early brain development.


Asunto(s)
Encéfalo/embriología , Proliferación Celular/fisiología , Proteínas de Unión al ADN/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Células-Madre Neurales/fisiología , Proteínas de Unión al ARN/metabolismo , Animales , Encéfalo/citología , Bromodesoxiuridina , Proteínas de Unión al ADN/genética , Electroporación , Regulación del Desarrollo de la Expresión Génica/genética , Proteína HMGA2/metabolismo , Inmunoprecipitación , Ratones , Ratones Noqueados , Proteínas de Unión al ARN/genética , Reacción en Cadena en Tiempo Real de la Polimerasa
17.
Dev Biol ; 416(2): 279-85, 2016 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-27343896

RESUMEN

The embryonic brain and spinal cord initially form through the process of neural tube closure (NTC). NTC is thought to be highly similar between rodents and humans, and studies of mouse genetic mutants have greatly increased our understanding of the molecular basis of NTC with relevance for human neural tube defects. In addition, studies using amphibian and chick embryos have shed light into the cellular and tissue dynamics underlying NTC. However, the dynamics of mammalian NTC has been difficult to study due to in utero development until recently when advances in mouse embryo ex vivo culture techniques along with confocal microscopy have allowed for imaging of mouse NTC in real time. Here, we have performed live imaging of mouse embryos with a particular focus on the non-neural ectoderm (NNE). Previous studies in multiple model systems have found that the NNE is important for proper NTC, but little is known about the behavior of these cells during mammalian NTC. Here we utilized a NNE-specific genetic labeling system to assess NNE dynamics during murine NTC and identified different NNE cell behaviors as the cranial region undergoes NTC. These results bring valuable new insight into regional differences in cellular behavior during NTC that may be driven by different molecular regulators and which may underlie the various positional disruptions of NTC observed in humans with neural tube defects.


Asunto(s)
Ectodermo/fisiología , Tubo Neural/embriología , Animales , Membrana Celular/fisiología , Membrana Celular/ultraestructura , Técnicas de Cultivo de Embriones , Células Epiteliales/ultraestructura , Femenino , Microscopía Intravital , Proteínas Luminiscentes/análisis , Proteínas Luminiscentes/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Neurulación/fisiología , Seudópodos/ultraestructura
18.
Dev Biol ; 472: 125-126, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33618188
19.
Dev Biol ; 409(2): 406-19, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26658218

RESUMEN

Mutations in the microtubule cytoskeleton are linked to cognitive and locomotor defects during development, and neurodegeneration in adults. How these mutations impact microtubules, and how this alters function at the level of neurons is an important area of investigation. Using a forward genetic screen in mice, we identified a missense mutation in Tuba1a α-tubulin that disrupts cortical and motor neuron development. Homozygous mutant mice exhibit cortical dysgenesis reminiscent of human tubulinopathies. Motor neurons fail to innervate target muscles in the limbs and show synapse defects at proximal targets. To directly examine effects on tubulin function, we created analogous mutations in the α-tubulin isotypes in budding yeast. These mutations sensitize yeast cells to microtubule stresses including depolymerizing drugs and low temperatures. Furthermore, we find that mutant α-tubulin is depleted from the cell lysate and from microtubules, thereby altering ratios of α-tubulin isotypes. Tubulin-binding cofactors suppress the effects of the mutation, indicating an important role for these cofactors in regulating the quality of the α-tubulin pool. Together, our results give new insights into the functions of Tuba1a, mechanisms for regulating tubulin proteostasis, and how compromising these may lead to neural defects.


Asunto(s)
Mutación/genética , Sistema Nervioso/embriología , Tubulina (Proteína)/genética , Secuencia de Aminoácidos , Animales , Axones/metabolismo , Modelos Animales de Enfermedad , Embrión de Mamíferos/patología , Epistasis Genética , Femenino , Pruebas Genéticas , Masculino , Ratones Endogámicos C57BL , Microtúbulos/metabolismo , Datos de Secuencia Molecular , Actividad Motora , Placa Motora/patología , Neuronas Motoras/patología , Sistema Nervioso/patología , Unión Neuromuscular/patología , Fenotipo , Polimerizacion , Multimerización de Proteína , Estabilidad Proteica , Saccharomyces cerevisiae/metabolismo , Sinapsis/patología , Tubulina (Proteína)/química
20.
Dev Biol ; 415(2): 278-295, 2016 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-26746790

RESUMEN

The cranial base is a component of the neurocranium and has a central role in the structural integration of the face, brain and vertebral column. Consequently, alteration in the shape of the human cranial base has been intimately linked with primate evolution and defective development is associated with numerous human facial abnormalities. Here we describe a novel recessive mutant mouse strain that presented with a domed head and fully penetrant cleft secondary palate coupled with defects in the formation of the underlying cranial base. Mapping and non-complementation studies revealed a specific mutation in Memo1 - a gene originally associated with cell migration. Expression analysis of Memo1 identified robust expression in the perichondrium and periosteum of the developing cranial base, but only modest expression in the palatal shelves. Fittingly, although the palatal shelves failed to elevate in Memo1 mutants, expression changes were modest within the shelves themselves. In contrast, the cranial base, which forms via endochondral ossification had major reductions in the expression of genes responsible for bone formation, notably matrix metalloproteinases and markers of the osteoblast lineage, mirrored by an increase in markers of cartilage and extracellular matrix development. Concomitant with these changes, mutant cranial bases showed an increased zone of hypertrophic chondrocytes accompanied by a reduction in both vascular invasion and mineralization. Finally, neural crest cell-specific deletion of Memo1 caused a failure of anterior cranial base ossification indicating a cell autonomous role for MEMO1 in the development of these neural crest cell derived structures. However, palate formation was largely normal in these conditional mutants, suggesting a non-autonomous role for MEMO1 in palatal closure. Overall, these findings assign a new function to MEMO1 in driving endochondral ossification in the cranium, and also link abnormal development of the cranial base with more widespread effects on craniofacial shape relevant to human craniofacial dysmorphology.


Asunto(s)
Fisura del Paladar/genética , Péptidos y Proteínas de Señalización Intracelular/fisiología , Desarrollo Maxilofacial/fisiología , Osteogénesis/fisiología , Hueso Paladar/embriología , Base del Cráneo/embriología , Animales , Cartílago/embriología , Cartílago/patología , Fisura del Paladar/embriología , Etilnitrosourea , Exones , Regulación del Desarrollo de la Expresión Génica , Genes Recesivos , Humanos , Masculino , Mesodermo/citología , Mesodermo/embriología , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Mutagénesis , Cresta Neural/citología , Cresta Neural/embriología , Hueso Paladar/metabolismo , Hueso Paladar/patología , Mutación Puntual , Base del Cráneo/metabolismo , Base del Cráneo/patología , Especificidad de la Especie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA