Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 428
Filtrar
1.
Dev Biol ; 516: 114-121, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-39102935

RESUMEN

The lack of a widely accessible method for expressing genes of interest in wild-type embryos is a fundamental obstacle to understanding genetic regulation during embryonic development. In particular, only a few methods are available for introducing gene expression vectors into cells prior to neural tube closure, which is a period of drastic development for many tissues. In this study, we present a simple technique for injecting vectors into the amniotic cavity and allowing them to reach the ectodermal cells and the epithelia of endodermal organs of mouse embryos at E8.0 via in utero injection, using only a widely used optical fiber with an illuminator. Using this technique, retroviruses can be introduced to facilitate the labeling of cells in various tissues, including the brain, spinal cord, epidermis, and digestive and respiratory organs. We also demonstrated in utero electroporation of plasmid DNA into E7.0 and E8.0 embryos. Taking advantage of this method, we reveal the association between Ldb1 and the activity of the Neurog2 transcription factor in the mouse neocortex. This technique can aid in analyzing the roles of genes of interest during endo- and ectodermal development prior to neural tube closure.


Asunto(s)
Ectodermo , Electroporación , Regulación del Desarrollo de la Expresión Génica , Tubo Neural , Animales , Ectodermo/metabolismo , Ectodermo/embriología , Ratones , Tubo Neural/embriología , Tubo Neural/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica/genética , Electroporación/métodos , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Embrión de Mamíferos/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Endodermo/metabolismo , Endodermo/embriología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Neurulación/genética , Vectores Genéticos/genética , Embarazo
2.
Cell Biol Toxicol ; 40(1): 51, 2024 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-38958792

RESUMEN

The implementation of Zinc oxide nanoparticles (ZnO NPs) raises concerns regarding their potential toxic effects on human health. Although more and more researches have confirmed the toxic effects of ZnO NPs, limited attention has been given to their impact on the early embryonic nervous system. This study aimed to explore the impact of exposure to ZnO NPs on early neurogenesis and explore its underlying mechanisms. We conducted experiments here to confirm the hypothesis that exposure to ZnO NPs causes neural tube defects in early embryonic development. We first used mouse and chicken embryos to confirm that ZnO NPs and the Zn2+ they release are able to penetrate the placental barrier, influence fetal growth and result in incomplete neural tube closure. Using SH-SY5Y cells, we determined that ZnO NPs-induced incomplete neural tube closure was caused by activation of various cell death modes, including ferroptosis, apoptosis and autophagy. Moreover, dissolved Zn2+ played a role in triggering widespread cell death. ZnO NPs were accumulated within mitochondria after entering cells, damaging mitochondrial function and resulting in the over production of reactive oxygen species, ultimately inducing cellular oxidative stress. The N-acetylcysteine (NAC) exhibits significant efficacy in mitigating cellular oxidative stress, thereby alleviating the cytotoxicity and neurotoxicity brought about by ZnO NPs. These findings indicated that the exposure of ZnO NPs in early embryonic development can induce cell death through oxidative stress, resulting in a reduced number of cells involved in early neural tube closure and ultimately resulting in incomplete neural tube closure during embryo development. The findings of this study could raise public awareness regarding the potential risks associated with the exposure and use of ZnO NPs in early pregnancy.


Asunto(s)
Desarrollo Embrionario , Defectos del Tubo Neural , Tubo Neural , Estrés Oxidativo , Especies Reactivas de Oxígeno , Óxido de Zinc , Óxido de Zinc/toxicidad , Animales , Estrés Oxidativo/efectos de los fármacos , Embrión de Pollo , Desarrollo Embrionario/efectos de los fármacos , Ratones , Tubo Neural/efectos de los fármacos , Tubo Neural/embriología , Tubo Neural/metabolismo , Humanos , Defectos del Tubo Neural/inducido químicamente , Defectos del Tubo Neural/metabolismo , Defectos del Tubo Neural/embriología , Defectos del Tubo Neural/patología , Especies Reactivas de Oxígeno/metabolismo , Apoptosis/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Femenino , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Nanopartículas del Metal/toxicidad , Autofagia/efectos de los fármacos , Línea Celular Tumoral , Nanopartículas/toxicidad
3.
Sci Adv ; 10(27): eadk1174, 2024 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-38968355

RESUMEN

During angiogenesis, vascular tip cells guide nascent vascular sprouts to form a vascular network. Apelin, an agonist of the G protein-coupled receptor Aplnr, is enriched in vascular tip cells, and it is hypothesized that vascular-derived Apelin regulates sprouting angiogenesis. We identify an apelin-expressing neural progenitor cell population in the dorsal neural tube. Vascular tip cells exhibit directed elongation and migration toward and along the apelin-expressing neural progenitor cells. Notably, restoration of neural but not vascular apelin expression in apelin mutants remedies the angiogenic defects of mutants. By functional analyses, we show the requirement of Apelin signaling for tip cell behaviors, like filopodia formation and cell elongation. Through genetic interaction studies and analysis of transgenic activity reporters, we identify Apelin signaling as a modulator of phosphoinositide 3-kinase and extracellular signal-regulated kinase signaling in tip cells in vivo. Our results suggest a previously unidentified neurovascular cross-talk mediated by Apelin signaling that is important for tip cell function during sprouting angiogenesis.


Asunto(s)
Apelina , Neovascularización Fisiológica , Células-Madre Neurales , Transducción de Señal , Animales , Apelina/metabolismo , Apelina/genética , Células-Madre Neurales/metabolismo , Células-Madre Neurales/citología , Pez Cebra , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Péptidos y Proteínas de Señalización Intercelular/genética , Movimiento Celular , Tubo Neural/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Quimiocinas , Proteínas de Pez Cebra
4.
Dev Growth Differ ; 66(5): 320-328, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38925637

RESUMEN

During the formation of the neural tube, the primordium of the vertebrate central nervous system, the actomyosin activity of cells in different regions drives neural plate bending. However, how the stiffness of the neural plate and surrounding tissues is regulated and mechanically influences neural plate bending has not been elucidated. Here, we used atomic force microscopy to reveal the relationship between the stiffness of the neural plate and the mesoderm during Xenopus neural tube formation. Measurements with intact embryos revealed that the stiffness of the neural plate was consistently higher compared with the non-neural ectoderm and that it increased in an actomyosin activity-dependent manner during neural plate bending. Interestingly, measurements of isolated tissue explants also revealed that the relationship between the stiffness of the apical and basal sides of the neural plate was reversed during bending and that the stiffness of the mesoderm was lower than that of the basal side of the neural plate. The experimental elevation of mesoderm stiffness delayed neural plate bending, suggesting that low mesoderm stiffness mechanically supports neural tube closure. This study provides an example of mechanical interactions between tissues during large-scale morphogenetic movements.


Asunto(s)
Placa Neural , Tubo Neural , Xenopus laevis , Animales , Tubo Neural/embriología , Tubo Neural/citología , Tubo Neural/metabolismo , Placa Neural/embriología , Placa Neural/metabolismo , Placa Neural/citología , Xenopus laevis/embriología , Mesodermo/citología , Mesodermo/embriología , Mesodermo/metabolismo , Ectodermo/citología , Ectodermo/metabolismo , Microscopía de Fuerza Atómica , Embrión no Mamífero/citología , Embrión no Mamífero/metabolismo , Embrión no Mamífero/embriología
5.
Int J Mol Sci ; 25(11)2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38891776

RESUMEN

Neural tube defects (NTDs), which are caused by impaired embryonic neural tube closure, are one of the most serious and common birth defects. Peptidyl-prolyl cis/trans isomerase 1 (Pin1) is a prolyl isomerase that uniquely regulates cell signaling by manipulating protein conformation following phosphorylation, although its involvement in neuronal development remains unknown. In this study, we explored the involvement of Pin1 in NTDs and its potential mechanisms both in vitro and in vivo. The levels of Pin1 expression were reduced in NTD models induced by all-trans retinoic acid (Atra). Pin1 plays a significant role in regulating the apoptosis, proliferation, differentiation, and migration of neurons. Moreover, Pin1 knockdown significantly was found to exacerbate oxidative stress (OS) and endoplasmic reticulum stress (ERs) in neuronal cells. Further studies showed that the Notch1-Nrf2 signaling pathway may participate in Pin1 regulation of NTDs, as evidenced by the inhibition and overexpression of the Notch1-Nrf2 pathway. In addition, immunofluorescence (IF), co-immunoprecipitation (Co-IP), and GST pull-down experiments also showed that Pin1 interacts directly with Notch1 and Nrf2. Thus, our study suggested that the knocking down of Pin1 promotes NTD progression by inhibiting the activation of the Notch1-Nrf2 signaling pathway, and it is possible that this effect is achieved by disrupting the interaction of Pin1 with Notch1 and Nrf2, affecting their proteostasis. Our research identified that the regulation of Pin1 by retinoic acid (RA) and its involvement in the development of NTDs through the Notch1-Nrf2 axis could enhance our comprehension of the mechanism behind RA-induced brain abnormalities.


Asunto(s)
Peptidilprolil Isomerasa de Interacción con NIMA , Defectos del Tubo Neural , Tretinoina , Animales , Femenino , Humanos , Ratones , Apoptosis/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Tubo Neural/metabolismo , Tubo Neural/efectos de los fármacos , Defectos del Tubo Neural/metabolismo , Defectos del Tubo Neural/genética , Defectos del Tubo Neural/inducido químicamente , Neuronas/metabolismo , Neuronas/efectos de los fármacos , Factor 2 Relacionado con NF-E2/metabolismo , Factor 2 Relacionado con NF-E2/genética , Peptidilprolil Isomerasa de Interacción con NIMA/metabolismo , Peptidilprolil Isomerasa de Interacción con NIMA/genética , Estrés Oxidativo/efectos de los fármacos , Receptor Notch1/metabolismo , Receptor Notch1/genética , Transducción de Señal/efectos de los fármacos , Tretinoina/metabolismo , Tretinoina/farmacología
6.
Sci Bull (Beijing) ; 69(14): 2260-2272, 2024 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-38702277

RESUMEN

Heparan sulfate proteoglycan 2 (HSPG2) gene encodes the matrix protein Perlecan, and genetic inactivation of this gene creates mice that are embryonic lethal with severe neural tube defects (NTDs). We discovered rare genetic variants of HSPG2 in 10% cases compared to only 4% in controls among a cohort of 369 NTDs. Endorepellin, a peptide cleaved from the domain V of Perlecan, is known to promote angiogenesis and autophagy in endothelial cells. The roles of enderepellin in neurodevelopment remain unclear so far. Our study revealed that endorepellin can migrate to the neuroepithelial cells and then be recognized and bind with the neuroepithelia receptor neurexin in vivo. Through the endocytic pathway, the interaction of endorepellin and neurexin physiologically triggers autophagy and appropriately modulates the differentiation of neural stem cells into neurons as a blocker, which is necessary for normal neural tube closure. We created knock-in (KI) mouse models with human-derived HSPG2 variants, using sperm-like stem cells that had been genetically edited by CRISPR/Cas9. We realized that any HSPG2 variants that affected the function of endorepellin were considered pathogenic causal variants for human NTDs given that the severe NTD phenotypes exhibited by these KI embryos occurred in a significantly higher response frequency compared to wildtype embryos. Our study provides a paradigm for effectively confirming pathogenic mutations in other genetic diseases. Furthermore, we demonstrated that using autophagy inhibitors at a cellular level can repress neuronal differentiation. Therefore, autophagy agonists may prevent NTDs resulting from failed autophagy maintenance and neuronal over-differentiation caused by deleterious endorepellin variants.


Asunto(s)
Autofagia , Proteoglicanos de Heparán Sulfato , Defectos del Tubo Neural , Animales , Ratones , Proteoglicanos de Heparán Sulfato/metabolismo , Proteoglicanos de Heparán Sulfato/genética , Humanos , Defectos del Tubo Neural/genética , Defectos del Tubo Neural/metabolismo , Defectos del Tubo Neural/patología , Tubo Neural/metabolismo , Tubo Neural/embriología , Tubo Neural/patología , Células-Madre Neurales/metabolismo , Células Neuroepiteliales/metabolismo , Femenino , Masculino , Modelos Animales de Enfermedad
7.
Dev Cell ; 59(15): 1940-1953.e10, 2024 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-38776925

RESUMEN

During neural tube (NT) development, the notochord induces an organizer, the floorplate, which secretes Sonic Hedgehog (SHH) to pattern neural progenitors. Conversely, NT organoids (NTOs) from embryonic stem cells (ESCs) spontaneously form floorplates without the notochord, demonstrating that stem cells can self-organize without embryonic inducers. Here, we investigated floorplate self-organization in clonal mouse NTOs. Expression of the floorplate marker FOXA2 was initially spatially scattered before resolving into multiple clusters, which underwent competition and sorting, resulting in a stable "winning" floorplate. We identified that BMP signaling governed long-range cluster competition. FOXA2+ clusters expressed BMP4, suppressing FOXA2 in receiving cells while simultaneously expressing the BMP-inhibitor NOGGIN, promoting cluster persistence. Noggin mutation perturbed floorplate formation in NTOs and in the NT in vivo at mid/hindbrain regions, demonstrating how the floorplate can form autonomously without the notochord. Identifying the pathways governing organizer self-organization is critical for harnessing the developmental plasticity of stem cells in tissue engineering.


Asunto(s)
Proteína Morfogenética Ósea 4 , Tubo Neural , Notocorda , Organoides , Animales , Ratones , Organoides/metabolismo , Organoides/citología , Tubo Neural/metabolismo , Tubo Neural/citología , Notocorda/metabolismo , Notocorda/citología , Proteína Morfogenética Ósea 4/metabolismo , Transducción de Señal , Factor Nuclear 3-beta del Hepatocito/metabolismo , Factor Nuclear 3-beta del Hepatocito/genética , Proteínas Hedgehog/metabolismo , Proteínas Hedgehog/genética , Proteínas Portadoras/metabolismo , Proteínas Portadoras/genética , Regulación del Desarrollo de la Expresión Génica , Proteínas Morfogenéticas Óseas/metabolismo
8.
Curr Top Dev Biol ; 159: 168-231, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38729676

RESUMEN

The development of the vertebrate spinal cord involves the formation of the neural tube and the generation of multiple distinct cell types. The process starts during gastrulation, combining axial elongation with specification of neural cells and the formation of the neuroepithelium. Tissue movements produce the neural tube which is then exposed to signals that provide patterning information to neural progenitors. The intracellular response to these signals, via a gene regulatory network, governs the spatial and temporal differentiation of progenitors into specific cell types, facilitating the assembly of functional neuronal circuits. The interplay between the gene regulatory network, cell movement, and tissue mechanics generates the conserved neural tube pattern observed across species. In this review we offer an overview of the molecular and cellular processes governing the formation and patterning of the neural tube, highlighting how the remarkable complexity and precision of vertebrate nervous system arises. We argue that a multidisciplinary and multiscale understanding of the neural tube development, paired with the study of species-specific strategies, will be crucial to tackle the open questions.


Asunto(s)
Tipificación del Cuerpo , Regulación del Desarrollo de la Expresión Génica , Tubo Neural , Transducción de Señal , Tubo Neural/embriología , Tubo Neural/metabolismo , Tubo Neural/citología , Animales , Tipificación del Cuerpo/genética , Humanos , Redes Reguladoras de Genes , Médula Espinal/embriología , Médula Espinal/citología , Médula Espinal/metabolismo , Diferenciación Celular , Movimiento Celular
9.
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
10.
Development ; 151(10)2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38682273

RESUMEN

Neurulation is a highly synchronized biomechanical process leading to the formation of the brain and spinal cord, and its failure leads to neural tube defects (NTDs). Although we are rapidly learning the genetic mechanisms underlying NTDs, the biomechanical aspects are largely unknown. To understand the correlation between NTDs and tissue stiffness during neural tube closure (NTC), we imaged an NTD murine model using optical coherence tomography (OCT), Brillouin microscopy and confocal fluorescence microscopy. Here, we associate structural information from OCT with local stiffness from the Brillouin signal of embryos undergoing neurulation. The stiffness of neuroepithelial tissues in Mthfd1l null embryos was significantly lower than that of wild-type embryos. Additionally, exogenous formate supplementation improved tissue stiffness and gross embryonic morphology in nullizygous and heterozygous embryos. Our results demonstrate the significance of proper tissue stiffness in normal NTC and pave the way for future studies on the mechanobiology of normal and abnormal embryonic development.


Asunto(s)
Tubo Neural , Neurulación , Tomografía de Coherencia Óptica , Animales , Femenino , Ratones , Fenómenos Biomecánicos , Embrión de Mamíferos/metabolismo , Formiato-Tetrahidrofolato Ligasa/genética , Formiato-Tetrahidrofolato Ligasa/metabolismo , Formiatos/metabolismo , Metilenotetrahidrofolato Deshidrogenasa (NADP)/genética , Metilenotetrahidrofolato Deshidrogenasa (NADP)/metabolismo , Ratones Noqueados , Microscopía Confocal , Mutación/genética , Tubo Neural/metabolismo , Defectos del Tubo Neural/genética , Defectos del Tubo Neural/metabolismo , Defectos del Tubo Neural/patología , Neurulación/genética , Tomografía de Coherencia Óptica/métodos
11.
Reprod Toxicol ; 125: 108576, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38479591

RESUMEN

Folate is a micronutrient essential for DNA synthesis, cell division, fetal growth and development. Folate deficiency leads to genomic instability. Inadequate intake of folate during conception may lead to neural tube defects (NTDs) in the offspring. Folate influences the DNA methylation, histone methylation and homocysteine mediated gene methylation. DNA methylation influences the expression of microRNAs (miRNAs). Folate deficiency may be associated with miRNAs misregulation leading to NTDs. Mitochondrial epigenetics and folate metabolism has proved to be involved in embryogenesis and neural tube development. Folate related genetic variants also cause the occurrence of NTDs. Unmetabolized excessive folate may affect health adversely. Hence estimation of folate levels in the blood plays an important role in high-risk cases.


Asunto(s)
Deficiencia de Ácido Fólico , MicroARNs , Defectos del Tubo Neural , Humanos , Ácido Fólico , Defectos del Tubo Neural/genética , Defectos del Tubo Neural/metabolismo , Deficiencia de Ácido Fólico/complicaciones , Deficiencia de Ácido Fólico/genética , Deficiencia de Ácido Fólico/metabolismo , Epigénesis Genética , Metilación de ADN , MicroARNs/genética , Tubo Neural/metabolismo
12.
Food Chem Toxicol ; 186: 114538, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38387523

RESUMEN

Arsenic exposure is a significant risk factor for folate-resistant neural tube defects (NTDs), but the potential mechanism is unclear. In this study, a mouse model of arsenic-induced NTDs was established to investigate how arsenic affects early neurogenesis leading to malformations. The results showed that in utero exposure to arsenic caused a decline in the normal embryos, an elevated embryo resorption, and a higher incidence of malformed embryos. Cranial and spinal deformities were the main malformation phenotypes observed. Meanwhile, arsenic-induced NTDs were accompanied by an oxidant/antioxidant imbalance manifested by elevated levels of reactive oxygen species (ROS) and decreased antioxidant activities. In addition, changes in the expression of autophagy-related genes and proteins (ULK1, Atg5, LC3B, p62) as well as an increase in autophagosomes were observed in arsenic-induced aberrant brain vesicles. Also, the components of the upstream pathway regulating autophagy (AMPK, PKB, mTOR, Raptor) were altered accordingly after arsenic exposure. Collectively, our findings propose a mechanism for arsenic-induced NTDs involving AMPK/PKB-mTORC1-mediated autophagy. Blocking autophagic cell death due to excessive autophagy provides a novel strategy for the prevention of folate-resistant NTDs, especially for arsenic-exposed populations.


Asunto(s)
Arsénico , Defectos del Tubo Neural , Ratones , Animales , Arsénico/toxicidad , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina , Antioxidantes , Tubo Neural/metabolismo , Autofagia/fisiología , Ácido Fólico/efectos adversos , Defectos del Tubo Neural/inducido químicamente
13.
Nat Commun ; 15(1): 1642, 2024 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-38388461

RESUMEN

Folate supplementation reduces the occurrence of neural tube defects (NTDs), birth defects consisting in the failure of the neural tube to form and close. The mechanisms underlying NTDs and their prevention by folate remain unclear. Here we show that folate receptor 1 (FOLR1) is necessary for the formation of neural tube-like structures in human-cell derived neural organoids. FOLR1 knockdown in neural organoids and in Xenopus laevis embryos leads to NTDs that are rescued by pteroate, a folate precursor that is unable to participate in metabolism. We demonstrate that FOLR1 interacts with and opposes the function of CD2-associated protein, molecule essential for apical endocytosis and turnover of C-cadherin in neural plate cells. In addition, folates increase Ca2+ transient frequency, suggesting that folate and FOLR1 signal intracellularly to regulate neural plate folding. This study identifies a mechanism of action of folate distinct from its vitamin function during neural tube formation.


Asunto(s)
Ácido Fólico , Defectos del Tubo Neural , Humanos , Ácido Fólico/metabolismo , Tubo Neural/metabolismo , Receptor 1 de Folato/genética , Receptor 1 de Folato/metabolismo , Defectos del Tubo Neural/genética , Defectos del Tubo Neural/metabolismo , Placa Neural/metabolismo
14.
Development ; 151(5)2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-38300806

RESUMEN

Defective tissue fusion during mammalian embryogenesis results in congenital anomalies, such as exencephaly, spina bifida and cleft lip and/or palate. The highly conserved transcription factor grainyhead-like 2 (Grhl2) is a crucial regulator of tissue fusion, with mouse models lacking GRHL2 function presenting with a fully penetrant open cranial neural tube, facial and abdominal clefting (abdominoschisis), and an open posterior neuropore. Here, we show that GRHL2 interacts with the soluble morphogen protein and bone morphogenetic protein (BMP) inhibitor noggin (NOG) to impact tissue fusion during development. The maxillary prominence epithelium in embryos lacking Grhl2 shows substantial morphological abnormalities and significant upregulation of NOG expression, together with aberrantly distributed pSMAD5-positive cells within the neural crest cell-derived maxillary prominence mesenchyme, indicative of disrupted BMP signalling. Reducing this elevated NOG expression (by generating Grhl2-/-;Nog+/- embryos) results in delayed embryonic lethality, partial tissue fusion rescue, and restoration of tissue form within the craniofacial epithelia. These data suggest that aberrant epithelial maintenance, partially regulated by noggin-mediated regulation of BMP-SMAD pathways, may underpin tissue fusion defects in Grhl2-/- mice.


Asunto(s)
Labio Leporino , Fisura del Paladar , Defectos del Tubo Neural , Animales , Ratones , Proteínas Morfogenéticas Óseas/metabolismo , Mamíferos/metabolismo , Tubo Neural/metabolismo , Receptores Nogo/metabolismo
15.
Cell ; 187(2): 276-293.e23, 2024 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-38171360

RESUMEN

During development, morphogens pattern tissues by instructing cell fate across long distances. Directly visualizing morphogen transport in situ has been inaccessible, so the molecular mechanisms ensuring successful morphogen delivery remain unclear. To tackle this longstanding problem, we developed a mouse model for compromised sonic hedgehog (SHH) morphogen delivery and discovered that endocytic recycling promotes SHH loading into signaling filopodia called cytonemes. We optimized methods to preserve in vivo cytonemes for advanced microscopy and show endogenous SHH localized to cytonemes in developing mouse neural tubes. Depletion of SHH from neural tube cytonemes alters neuronal cell fates and compromises neurodevelopment. Mutation of the filopodial motor myosin 10 (MYO10) reduces cytoneme length and density, which corrupts neuronal signaling activity of both SHH and WNT. Combined, these results demonstrate that cytoneme-based signal transport provides essential contributions to morphogen dispersion during mammalian tissue development and suggest MYO10 is a key regulator of cytoneme function.


Asunto(s)
Estructuras de la Membrana Celular , Miosinas , Tubo Neural , Transducción de Señal , Animales , Ratones , Transporte Biológico , Estructuras de la Membrana Celular/metabolismo , Proteínas Hedgehog/metabolismo , Miosinas/metabolismo , Seudópodos/metabolismo , Tubo Neural/citología , Tubo Neural/metabolismo
16.
Zool Res ; 45(2): 233-241, 2024 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-38287904

RESUMEN

Neural tube defects (NTDs) are severe congenital neurodevelopmental disorders arising from incomplete neural tube closure. Although folate supplementation has been shown to mitigate the incidence of NTDs, some cases, often attributable to genetic factors, remain unpreventable. The SHROOM3 gene has been implicated in NTD cases that are unresponsive to folate supplementation; at present, however, the underlying mechanism remains unclear. Neural tube morphogenesis is a complex process involving the folding of the planar epithelium of the neural plate. To determine the role of SHROOM3 in early developmental morphogenesis, we established a neuroepithelial organoid culture system derived from cynomolgus monkeys to closely mimic the in vivo neural plate phase. Loss of SHROOM3 resulted in shorter neuroepithelial cells and smaller nuclei. These morphological changes were attributed to the insufficient recruitment of cytoskeletal proteins, namely fibrous actin (F-actin), myosin II, and phospho-myosin light chain (PMLC), to the apical side of the neuroepithelial cells. Notably, these defects were not rescued by folate supplementation. RNA sequencing revealed that differentially expressed genes were enriched in biological processes associated with cellular and organ morphogenesis. In summary, we established an authentic in vitro system to study NTDs and identified a novel mechanism for NTDs that are unresponsive to folate supplementation.


Asunto(s)
Proteínas del Citoesqueleto , Defectos del Tubo Neural , Animales , Proteínas del Citoesqueleto/metabolismo , Tubo Neural/metabolismo , Macaca fascicularis , Defectos del Tubo Neural/genética , Defectos del Tubo Neural/metabolismo , Defectos del Tubo Neural/veterinaria , Células Neuroepiteliales/metabolismo , Ácido Fólico/metabolismo , Organoides , Citoesqueleto
17.
J Environ Sci (China) ; 138: 572-584, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38135421

RESUMEN

Birth defects have become a public health concern. The hazardous environmental factors exposure to embryos could increase the risk of birth defects. Cadmium, a toxic environmental factor, can cross the placental barrier during pregnancy. Pregnant woman may be subjected to cadmium before taking precautionary protective actions. However, the link between birth defects and cadmium remains obscure. Cadmium exposure can induce excessive apoptosis in neuroepithelium during embryonic development progresses. Cadmium exposure activated the p53 via enhancing the adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) and reactive oxygen species' (ROS) level. And cadmium decreases the level of Paired box 3 (Pax3) and murine double minute 2 (Mdm2), disrupting the process of p53 ubiquitylation. And p53 accumulation induced excessive apoptosis in neuroepithelium during embryonic development progresses. Excessive apoptosis led to the failure of neural tube closure. The study emphasizes that environmental materials may increase the health risk for embryos. Cadmium caused the failure of neural tube closure during early embryotic day. Pregnant women may be exposed by cadmium before taking precautionary protective actions, because of cadmium concentration-containing foods and environmental tobacco smoking. This suggests that prenatal cadmium exposure is a threatening risk factor for birth defects.


Asunto(s)
Defectos del Tubo Neural , Femenino , Embarazo , Humanos , Animales , Ratones , Defectos del Tubo Neural/inducido químicamente , Defectos del Tubo Neural/metabolismo , Cadmio/toxicidad , Cadmio/metabolismo , Tubo Neural/metabolismo , Factor de Transcripción PAX3/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Placenta/metabolismo , Apoptosis
18.
Dis Model Mech ; 16(11)2023 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-37885410

RESUMEN

Sonic hedgehog (Shh) signaling is the morphogen signaling that regulates embryonic craniofacial and neural tube development. G protein-coupled receptor 161 (Gpr161) is a negative regulator of Shh signaling, and its inactivation in mice results in embryo lethality associated with craniofacial defects and neural tube defects. However, the structural defects of later embryonic stages and cell lineages underlying abnormalities have not been well characterized due to the limited lifespan of Gpr161 null mice. We found that embryos with Pax3 lineage-specific deletion of Gpr161 presented with tectal hypertrophy (anterior dorsal neuroepithelium), cranial vault and facial bone hypoplasia (cranial neural crest), vertebral abnormalities (somite) and the closed form of spina bifida (posterior dorsal neuroepithelium). In particular, the closed form of spina bifida was partly due to reduced Pax3 and Cdx4 gene expression in the posterior dorsal neural tubes of Gpr161 mutant embryos with decreased Wnt signaling, whereas Shh signaling was increased. We describe a previously unreported role for Gpr161 in the development of posterior neural tubes and confirm its role in cranial neural crest- and somite-derived skeletogenesis and midbrain morphogenesis in mice.


Asunto(s)
Tubo Neural , Disrafia Espinal , Ratones , Animales , Tubo Neural/metabolismo , Proteínas Hedgehog/metabolismo , Factores de Transcripción/metabolismo , Desarrollo Embrionario , Vía de Señalización Wnt , Neurogénesis , Columna Vertebral
19.
Biol Rev Camb Philos Soc ; 98(6): 2271-2283, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37534608

RESUMEN

Neurulation transforms the neuroectoderm into the neural tube. This transformation relies on reorganising the configurational relationships between the orientations of intrinsic polarities of neighbouring cells. These orientational intercellular relationships are established, maintained, and modulated by orientational cell adhesions (OCAs). Here, using zebrafish (Danio rerio) neurulation as a major model, we propose a new perspective on how OCAs contribute to the parallel, antiparallel, and opposing intercellular relationships that underlie the neural plate-keel-rod-tube transformation, a stepwise process of cell aggregation followed by cord hollowing. We also discuss how OCAs in neurulation may be regulated by various adhesion molecules, including cadherins, Eph/Ephrins, Claudins, Occludins, Crumbs, Na+ /K+ -ATPase, and integrins. By comparing neurulation among species, we reveal that antiparallel OCAs represent a conserved mechanism for the fusion of the neural tube. Throughout, we highlight some outstanding questions regarding OCAs in neurulation. Answers to these questions will help us understand better the mechanisms of tubulogenesis of many tissues.


Asunto(s)
Neurulación , Pez Cebra , Animales , Adhesión Celular , Tubo Neural/metabolismo , Placa Neural/metabolismo
20.
Trends Endocrinol Metab ; 34(9): 539-553, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37468429

RESUMEN

Changes in maternal nutrient availability due to diet or disease significantly increase the risk of neural tube defects (NTDs). Because the incidence of metabolic disease continues to rise, it is urgent that we better understand how altered maternal nutrient levels can influence embryonic neural tube development. Furthermore, primary neurulation occurs before placental function during a period of histiotrophic nutrient exchange. In this review we detail how maternal metabolites are transported by the yolk sac to the developing embryo. We discuss recent advances in understanding how altered maternal levels of essential nutrients disrupt development of the neuroepithelium, and identify points of intersection between metabolic pathways that are crucial for NTD prevention.


Asunto(s)
Ácido Fólico , Defectos del Tubo Neural , Humanos , Femenino , Embarazo , Ácido Fólico/metabolismo , Tubo Neural/metabolismo , Neurulación , Placenta/metabolismo , Defectos del Tubo Neural/etiología , Defectos del Tubo Neural/metabolismo , Defectos del Tubo Neural/prevención & control
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA