Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
Nat Commun ; 15(1): 5136, 2024 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-38879605

RESUMEN

Coordination of neuronal differentiation with expansion of the neuroepithelial/neural progenitor cell (NEPC/NPC) pool is essential in early brain development. Our in vitro and in vivo studies identify independent and opposing roles for two neural-specific and differentially expressed non-coding RNAs derived from the same locus: the evolutionarily conserved lncRNA Rncr3 and the embedded microRNA miR124a-1. Rncr3 regulates NEPC/NPC proliferation and controls the biogenesis of miR124a, which determines neuronal differentiation. Rncr3 conserved exons 2/3 are cytosine methylated and bound by methyl-CpG binding protein MeCP2, which restricts expression of miR124a embedded in exon 4 to prevent premature neuronal differentiation, and to orchestrate proper brain growth. MeCP2 directly binds cytosine-methylated Rncr3 through previously unrecognized lysine residues and suppresses miR124a processing by recruiting PTBP1 to block access of DROSHA-DGCR8. Thus, miRNA processing is controlled by lncRNA m5C methylation along with the defined m5C epitranscriptomic RNA reader protein MeCP2 to coordinate brain development.


Asunto(s)
Proteína 2 de Unión a Metil-CpG , MicroARNs , Células-Madre Neurales , Neurogénesis , ARN Largo no Codificante , MicroARNs/metabolismo , MicroARNs/genética , Proteína 2 de Unión a Metil-CpG/metabolismo , Proteína 2 de Unión a Metil-CpG/genética , Neurogénesis/genética , Animales , Ratones , ARN Largo no Codificante/metabolismo , ARN Largo no Codificante/genética , Células-Madre Neurales/metabolismo , Células-Madre Neurales/citología , Encéfalo/metabolismo , Encéfalo/embriología , Humanos , Diferenciación Celular , Metilación de ADN , Proteína de Unión al Tracto de Polipirimidina/metabolismo , Proteína de Unión al Tracto de Polipirimidina/genética , Proliferación Celular , Ratones Endogámicos C57BL , 5-Metilcitosina/metabolismo , 5-Metilcitosina/análogos & derivados , Masculino , Exones/genética , Neuronas/metabolismo , Ribonucleasa III
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 ; 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
4.
WIREs Mech Dis ; 14(5): e1559, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35504597

RESUMEN

Neural tube closure (NTC) is crucial for proper development of the brain and spinal cord and requires precise morphogenesis from a sheet of cells to an intact three-dimensional structure. NTC is dependent on successful regulation of hundreds of genes, a myriad of signaling pathways, concentration gradients, and is influenced by epigenetic and environmental cues. Failure of NTC is termed a neural tube defect (NTD) and is a leading class of congenital defects in the United States and worldwide. Though NTDs are all defined as incomplete closure of the neural tube, the pathogenesis of an NTD determines the type, severity, positioning, and accompanying phenotypes. In this review, we survey pathogenesis of NTDs relating to disruption of cellular processes arising from genetic mutations, altered epigenetic regulation, and environmental influences by micronutrients and maternal condition. This article is categorized under: Congenital Diseases > Genetics/Genomics/Epigenetics Neurological Diseases > Genetics/Genomics/Epigenetics Neurological Diseases > Stem Cells and Development.


Asunto(s)
Defectos del Tubo Neural , Tubo Neural , Epigénesis Genética , Ácido Fólico/metabolismo , Humanos , Tubo Neural/anomalías , Defectos del Tubo Neural/genética , Neurulación/genética
5.
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
6.
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
7.
Neural Dev ; 15(1): 8, 2020 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-32650820

RESUMEN

BACKGROUND: Neural tube defects (NTDs) are failure of neural tube closure, which includes multiple central nervous system phenotypes. More than 300 mouse mutant strains exhibits NTDs phenotypes and give us some clues to establish association between biological functions and subphenotypes. However, the knowledge about association in human remains still very poor. METHODS: High throughput targeted genome DNA sequencing were performed on 280 neural tube closure-related genes in 355 NTDs cases and 225 ethnicity matched controls, RESULTS: We explored that potential damaging rare variants in genes functioning in chromatin modification, apoptosis, retinoid metabolism and lipid metabolism are associated with human NTDs. Importantly, our data indicate that except for planar cell polarity pathway, craniorachischisis is also genetically related with chromatin modification and retinoid metabolism. Furthermore, single phenotype in cranial or spinal regions displays significant association with specific biological function, such as anencephaly is associated with potentially damaging rare variants in genes functioning in chromatin modification, encephalocele is associated with apoptosis, retinoid metabolism and one carbon metabolism, spina bifida aperta and spina bifida cystica are associated with apoptosis; lumbar sacral spina bifida aperta and spina bifida occulta are associated with lipid metabolism. By contrast, complex phenotypes in both cranial and spinal regions display association with various biological functions given the different phenotypes. CONCLUSIONS: Our study links genetic variant to subphenotypes of human NTDs and provides a preliminary but direct clue to investigate pathogenic mechanism for human NTDs.


Asunto(s)
Estudios de Asociación Genética , Defectos del Tubo Neural/clasificación , Defectos del Tubo Neural/genética , Niño , Preescolar , Estudios de Cohortes , Femenino , Feto , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Lactante , Recién Nacido , Masculino , Defectos del Tubo Neural/metabolismo , Defectos del Tubo Neural/fisiopatología , Fenotipo , Embarazo , Análisis de Secuencia de ADN
8.
Epigenomics ; 12(1): 5-18, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31769301

RESUMEN

Aim: To know the cause of sequence variants in neural tube defect (NTD). Materials & methods: We sequenced genes implicated in neural tube closure (NTC) in a Chinese cohort and elucidated the molecular mechanism-driving mutations. Results: In NTD cases, an increase in specific variants was identified, potentially deleterious rare variants harbored in H3K36me3 occupancy regions that recruits mismatch repair (MMR) machinery. Lower folate concentrations in local brain tissues were also observed. In neuroectoderm cells, folic acid insufficiency attenuated association of Msh6 to H3K36me3, and reduced bindings to NTC genes. Rare variants in human NTDs were featured by MMR deficiency and more severe microsatellite instability. Conclusion: Our work suggests a mechanistic link between folate insufficiency and MMR deficiency that correlates with an increase of rare variants in NTC genes.


Asunto(s)
Reparación de la Incompatibilidad de ADN/genética , Susceptibilidad a Enfermedades , Deficiencia de Ácido Fólico/complicaciones , Defectos del Tubo Neural/etiología , Defectos del Tubo Neural/metabolismo , Ácido Fólico/metabolismo , Deficiencia de Ácido Fólico/metabolismo , Variación Genética , Histonas/metabolismo , Humanos
9.
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
10.
Sci Rep ; 8(1): 18002, 2018 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-30573757

RESUMEN

Intratumoral genetic heterogeneity is a widely accepted characteristic of human cancer, including the most common primary malignant brain tumor, glioblastoma. However, the variability in biological behaviors amongst cells within individual tumors is not well described. Invasion into unaffected brain parenchyma is one such behavior, and a leading mechanism of tumor recurrence unaddressed by the current therapeutic armamentarium. Further, providing insight into variability of tumor cell migration within individual tumors may inform discovery of novel anti-invasive therapeutics. In this study, ex vivo organotypic slice cultures from EGFR-wild type and EGFR-amplified patient tumors were treated with the EGFR inhibitor gefitinib to evaluate potential sub-population restricted intratumoral drug-specific responses. High-resolution time-lapse microscopy and quantitative path tracking demonstrated migration of individual cells are punctuated by intermittent bursts of movement. Elevation of population aggregate mean speeds were driven by subpopulations of cells exhibiting frequent high-amplitude bursts, enriched within EGFR-amplified tumors. Treatment with gefitinib specifically targeted high-burst cell subpopulations only in EGFR-amplified tumors, decreasing bursting frequency and amplitude. We provide evidence of intratumoral subpopulations of cells with enhanced migratory behavior in human glioblastoma, selectively targeted via EGFR inhibition. These data justify use of direct human tumor slice cultures to investigate patient-specific therapies designed to limit tumor invasion.


Asunto(s)
Neoplasias Encefálicas/patología , Glioblastoma/patología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Gefitinib/farmacología , Humanos , Invasividad Neoplásica , Cultivo Primario de Células , Técnicas de Cultivo de Tejidos , Células Tumorales Cultivadas
11.
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
13.
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
14.
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
15.
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
16.
Dev Dyn ; 244(6): 736-47, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25774014

RESUMEN

BACKGROUND: Cilia are important for Hedgehog signaling in vertebrates and many genes that encode proteins involved in ciliogenesis have been studied for their roles in embryonic development. Null mutations in many of these genes cause early embryonic lethality, hence an understanding of their roles in postnatal development is limited. RESULTS: The Inturned (Intu) gene is required for ciliogenesis and here we report a recessive hypomorphic mutation, resulting in substitution of a conserved hydrophobic residue (I813N) near the C-terminus, that sheds light on later functions of Intu. Mice homozygous for this Double-thumb (Intu(Dtm)) allele exhibit polydactyly, retarded growth, and reduced survival. There is a moderate loss of cilia in Intu(Dtm/Dtm) mutants, and Intu(I813N) exhibits compromised ability to increase ciliogenesis in cultured Intu null mutant cells. Intu(Dtm) mutants show rib defects and delay of endochondral ossification in long bones, digits, vertebrae, and the sternum. These skeletal defects correlate with a decrease in Hh signaling. However, patterning of the neural tube and planar cell polarity appear to be normal. CONCLUSIONS: This hypomorphic Intu allele highlights an important role of Intu in mouse skeletal development.


Asunto(s)
Anomalías Múltiples/genética , Proteínas de la Membrana/fisiología , Mutación Missense , Osteogénesis/genética , Mutación Puntual , Anomalías Múltiples/embriología , Alelos , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Huesos/anomalías , Huesos/embriología , Polaridad Celular , Células Cultivadas , Cilios/ultraestructura , Trastornos del Crecimiento/genética , Proteínas Hedgehog/fisiología , Proteínas de la Membrana/genética , Ratones , Datos de Secuencia Molecular , Defectos del Tubo Neural/genética , Receptores Patched , Polidactilia/embriología , Polidactilia/genética , Estructura Terciaria de Proteína , Receptores de Superficie Celular/biosíntesis , Receptores de Superficie Celular/genética , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Transducción de Señal/genética
17.
Elife ; 42015 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-25564733

RESUMEN

Myopathies decrease muscle functionality. Mutations in ryanodine receptor 1 (RyR1) are often associated with myopathies with microscopic core-like structures in the muscle fiber. In this study, we identify a mouse RyR1 model in which heterozygous animals display clinical and pathological hallmarks of myopathy with core-like structures. The RyR1 mutation decreases sensitivity to activated calcium release and myoplasmic calcium levels, subsequently affecting mitochondrial calcium and ATP production. Mutant muscle shows a persistent potassium leak and disrupted expression of regulators of potassium homeostasis. Inhibition of KATP channels or increasing interstitial potassium by diet or FDA-approved drugs can reverse the muscle weakness, fatigue-like physiology and pathology. We identify regulators of potassium homeostasis as biomarkers of disease that may reveal therapeutic targets in human patients with myopathy of central core disease (CCD). Altogether, our results suggest that amelioration of potassium leaks through potassium homeostasis mechanisms may minimize muscle damage of myopathies due to certain RyR1 mutations.


Asunto(s)
Enfermedades Musculares/patología , Potasio/metabolismo , Animales , Transporte Biológico/efectos de los fármacos , Biomarcadores/metabolismo , Biopsia , Calcio/metabolismo , Dieta , Etilnitrosourea , Regulación de la Expresión Génica/efectos de los fármacos , Gliburida/farmacología , Heterocigoto , Homeostasis/efectos de los fármacos , Humanos , Canales KATP/metabolismo , Ratones Endogámicos C57BL , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/patología , Músculo Esquelético/ultraestructura , Enfermedades Musculares/genética , Mutación/genética , Miopatía del Núcleo Central/genética , Miopatía del Núcleo Central/patología , NAD/metabolismo , Fenotipo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/genética , Canal Liberador de Calcio Receptor de Rianodina/metabolismo
18.
Artículo en Inglés | MEDLINE | ID: mdl-24902834

RESUMEN

The neural tube (NT), the embryonic precursor of the vertebrate brain and spinal cord, is generated by a complex and highly dynamic morphological process. In mammals, the initially flat neural plate bends and lifts bilaterally to generate the neural folds followed by fusion of the folds at the midline during the process of neural tube closure (NTC). Failures in any step of this process can lead to neural tube defects (NTDs), a common class of birth defects that occur in approximately 1 in 1000 live births. These severe birth abnormalities include spina bifida, a failure of closure at the spinal level; craniorachischisis, a failure of NTC along the entire body axis; and exencephaly, a failure of the cranial neural folds to close which leads to degeneration of the exposed brain tissue termed anencephaly. The mouse embryo presents excellent opportunities to explore the genetic basis of NTC in mammals; however, its in utero development has also presented great challenges in generating a deeper understanding of how gene function regulates the cell and tissue behaviors that drive this highly dynamic process. Recent technological advances are now allowing researchers to address these questions through visualization of NTC dynamics in the mouse embryo in real time, thus offering new insights into the morphogenesis of mammalian NTC.


Asunto(s)
Morfogénesis , Cresta Neural/crecimiento & desarrollo , Placa Neural/crecimiento & desarrollo , Tubo Neural/crecimiento & desarrollo , Anencefalia/genética , Anencefalia/patología , Animales , Ratones , Placa Neural/patología , Tubo Neural/patología , Defectos del Tubo Neural/genética , Defectos del Tubo Neural/patología , Disrafia Espinal/genética , Disrafia Espinal/patología
19.
Nat Commun ; 5: 3885, 2014 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-24875059

RESUMEN

Human genetic studies have established a link between a class of centrosome proteins and microcephaly. Current studies of microcephaly focus on defective centrosome/spindle orientation. Mutations in WDR62 are associated with microcephaly and other cortical abnormalities in humans. Here we create a mouse model of Wdr62 deficiency and find that the mice exhibit reduced brain size due to decreased neural progenitor cells (NPCs). Wdr62 depleted cells show spindle instability, spindle assembly checkpoint (SAC) activation, mitotic arrest and cell death. Mechanistically, Wdr62 associates and genetically interacts with Aurora A to regulate spindle formation, mitotic progression and brain size. Our results suggest that Wdr62 interacts with Aurora A to control mitotic progression, and loss of these interactions leads to mitotic delay and cell death of NPCs, which could be a potential cause of human microcephaly.


Asunto(s)
Aurora Quinasa A/metabolismo , Encéfalo/embriología , Proteínas de Ciclo Celular/genética , Puntos de Control de la Fase M del Ciclo Celular/genética , Microcefalia/embriología , Proteínas Asociadas a Microtúbulos/genética , Mitosis/genética , Células-Madre Neurales/metabolismo , Huso Acromático/metabolismo , Animales , Encéfalo/metabolismo , Encéfalo/patología , Células Madre Embrionarias/metabolismo , Ratones , Microcefalia/metabolismo , Microcefalia/patología , Mutación , Proteínas del Tejido Nervioso , Tamaño de los Órganos
20.
Dev Neurobiol ; 74(5): 483-97, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24170322

RESUMEN

Failure of embryonic neural tube closure results in the second most common class of birth defects known as neural tube defects (NTDs). While NTDs are likely the result of complex multigenic dysfunction, it is not known whether polymorphisms in epigenetic regulators may be risk factors for NTDs. Here we characterized Baf155(msp3) , a unique ENU-induced allele in mice. Homozygous Baf155(mps3) embryos exhibit highly penetrant exencephaly, allowing us to investigate the roles of an assembled, but malfunctional BAF chromatin remodeling complex in vivo at the time of neural tube closure. Evidence of defects in proliferation and apoptosis were found within the neural tube. RNA-Seq analysis revealed that surprisingly few genes showed altered expression in Baf155 mutant neural tissue, given the broad epigenetic role of the BAF complex, but included genes involved in neural development and cell survival. Moreover, gene expression changes between individual mutants were variable even though the NTD was consistently observed. This suggests that inconsistent gene regulation contributes to failed neural tube closure. These results shed light on the role of the BAF complex in the process of neural tube closure and highlight the importance of studying missense alleles to understand epigenetic regulation during critical phases of development.


Asunto(s)
Mutación Missense , Defectos del Tubo Neural/genética , Defectos del Tubo Neural/patología , Factores de Transcripción/genética , Alelos , Animales , Western Blotting , Muerte Celular/genética , Muerte Celular/fisiología , Expresión Génica , Técnicas de Genotipaje , Inmunoprecipitación , Ratones Endogámicos C57BL , Ratones Endogámicos , Células-Madre Neurales/patología , Células-Madre Neurales/fisiología , Tubo Neural/metabolismo , Tubo Neural/patología , Defectos del Tubo Neural/fisiopatología , Neurogénesis/genética , Neurogénesis/fisiología , Fenotipo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA