Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.150
Filter
1.
Dev Biol ; 516: 114-121, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39102935

ABSTRACT

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.


Subject(s)
Ectoderm , Electroporation , Gene Expression Regulation, Developmental , Neural Tube , Animals , Ectoderm/metabolism , Ectoderm/embryology , Mice , Neural Tube/embryology , Neural Tube/metabolism , Female , Gene Expression Regulation, Developmental/genetics , Electroporation/methods , Transcription Factors/genetics , Transcription Factors/metabolism , Embryo, Mammalian/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Endoderm/metabolism , Endoderm/embryology , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Neurulation/genetics , Genetic Vectors/genetics , Pregnancy
2.
Turk Neurosurg ; 34(5): 827-832, 2024.
Article in English | MEDLINE | ID: mdl-39087289

ABSTRACT

AIM: To evaluate tenoxicam's effects on embryonic neural tube formation to identify potential teratogenicity and determine the underlying mechanisms leading to neural tube defects (NTDs). MATERIAL AND METHODS: This study was conducted at our University's Neuro-embryology Laboratory. A total of 100 fertile chicken eggs were opened using the windowing method after 24 hours of incubation. The embryo models were divided into four groups based on tenoxicam dosage: 0.01, 0.02, 0.10 µg, and control group (0.9% SF was administered). The tenoxicam groups were administered 20 µL volume sub-blastodermally. The eggs were incubated for another 24 hours after being covered with sterile draping. All the eggs were opened at the 48th hour, and the embryos were evaluated. RESULTS: Each group consisted of 25 chicken embryos. Normal neural tube development was observed in Group 1 (0.01µg) with 23 out of 25 embryos, Group 2 (0.02 µg) with 20 out of 25 embryos, Group 3 (0.10µg) with 16 out of 25 embryos, and Group 4 (control group) with 24 out of 25 embryos. Additionally, the rates of absence of embryo development were 8%, 8%, 12%, and 4% in Groups 1, 2, and 3 and the control group, respectively. CONCLUSION: We observed that tenoxicam use caused midline closure defects in early chicken embryos in a dose-dependent manner. Further studies are required to determine the mechanisms underlying the embryonic damage and teratogenic effects due to genetic and environmental factors and minimize the development of congenital defects.


Subject(s)
Neural Tube Defects , Piroxicam , Animals , Piroxicam/analogs & derivatives , Chick Embryo , Neural Tube Defects/chemically induced , Neural Tube Defects/embryology , Neural Tube/drug effects , Neural Tube/embryology , Disease Models, Animal , Embryonic Development/drug effects , Anti-Inflammatory Agents, Non-Steroidal
3.
Cell Biol Toxicol ; 40(1): 51, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38958792

ABSTRACT

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.


Subject(s)
Embryonic Development , Neural Tube Defects , Neural Tube , Oxidative Stress , Reactive Oxygen Species , Zinc Oxide , Zinc Oxide/toxicity , Animals , Oxidative Stress/drug effects , Chick Embryo , Embryonic Development/drug effects , Mice , Neural Tube/drug effects , Neural Tube/embryology , Neural Tube/metabolism , Humans , Neural Tube Defects/chemically induced , Neural Tube Defects/metabolism , Neural Tube Defects/embryology , Neural Tube Defects/pathology , Reactive Oxygen Species/metabolism , Apoptosis/drug effects , Cell Death/drug effects , Female , Mitochondria/drug effects , Mitochondria/metabolism , Metal Nanoparticles/toxicity , Autophagy/drug effects , Cell Line, Tumor , Nanoparticles/toxicity
4.
Sci Adv ; 10(27): eadk1174, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38968355

ABSTRACT

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.


Subject(s)
Apelin , Neovascularization, Physiologic , Neural Stem Cells , Signal Transduction , Animals , Apelin/metabolism , Apelin/genetics , Neural Stem Cells/metabolism , Neural Stem Cells/cytology , Zebrafish , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Cell Movement , Neural Tube/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Chemokines , Zebrafish Proteins
5.
Dev Cell ; 59(12): 1487-1488, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38889690

ABSTRACT

In this issue of Developmental Cell, Bolondi et al. systematically assesses neuro-mesodermal progenitor (NMP) dynamics by combining a mouse stem-cell-based embryo model with molecular recording of single cells, shedding light on the dynamics of neural tube and paraxial mesoderm formation during mammalian development.


Subject(s)
Mesoderm , Animals , Mice , Mesoderm/cytology , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Neural Tube/cytology , Neural Tube/embryology , Cell Differentiation/physiology , Stem Cells/cytology , Stem Cells/metabolism , Body Patterning
6.
Toxicol Appl Pharmacol ; 489: 117009, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38906509

ABSTRACT

INTRODUCTION: Aripiprazole (ARI) is a recently developed antipsychotic medication that belongs to the second generation of antipsychotics. The literature has contradictory information regarding ARI, which has been classified as pregnant use category C by the FDA. METHODS: 125 pathogen-free fertilized eggs were incubated for 28 h and divided into five groups of 25 eggs each (including the control group), and 18 eggs with intact integrity were selected from each group. After the experimental groups were divided, ARI was administered subblastodermally with a Hamilton micro-injector at 4 different doses (1 mg/kg, 5 mg/kg, 10 mg/kg, 20 mg/kg). At the 48th hour of incubation, all eggs were hatched and embryos were removed from the embryonic membranes. And then morphologic (position of the neural tube (open or closed), crown-rump length, number of somites, embryological development status), histopathologic (apoptosis (caspase 3), cell proliferation (PCNA), in situ recognition of DNA breaks (tunnel)), genetic (BRE gene expression) analyzes were performed. RESULTS: According to the results of the morphological analysis, when the frequency of neural tube patency was evaluated among the experimental groups, a statistically significant difference was determined between the control group and all groups (p < 0.001). In addition, the mean crown-rump length and somite number of the embryos decreased in a dose-dependent manner compared to the control group. It was determined that mRNA levels of the BRE gene decreased in embryos exposed to ARI compared to the control group (p < 0.001). CONCLUSION: Morphologically, histopathologically, and genetically, aripiprazole exposure delayed neurogenesis and development in early chick embryos. These findings suggest its use in pregnant women may be teratogenic. We note that these results are preliminary for pregnant women, but they should be expanded and studied with additional and other samples.


Subject(s)
Aripiprazole , Neural Tube , Animals , Aripiprazole/toxicity , Neural Tube/drug effects , Chick Embryo , Antipsychotic Agents/toxicity , Apoptosis/drug effects , Cell Proliferation/drug effects , Embryonic Development/drug effects , Dose-Response Relationship, Drug , Gene Expression Regulation, Developmental/drug effects , Proliferating Cell Nuclear Antigen/metabolism , Caspase 3/metabolism , Caspase 3/genetics
7.
Toxicol Appl Pharmacol ; 489: 117011, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38906510

ABSTRACT

The critical developmental stages of the embryo are strongly influenced by the dietary composition of the mother. Acrylamide is a food contaminant that can form in carbohydrate-rich foods that are heat-treated. The aim of this study was to investigate the toxicity of a relatively low dose of acrylamide on the development of the neural tube in the early stage chick embryos. Specific pathogen-free fertilized eggs (n = 100) were treated with acrylamide (0.1, 0.5, 2.5, 12.5 mg/kg) between 28-30th hours of incubation and dissected at 48th hours. In addition to morphological and histopathological examinations, proliferating cell nuclear antigen (PCNA) and caspase 3 were analyzed immunohistochemically. The brain and reproductive expression gene (BRE) was analyzed by RT-PCR. Acrylamide exposure had a negative effect on neural tube status even at a very low dose (0.1 mg/kg) (p < 0.05). Doses of 0.5 mg/kg and above caused a delay in neural tube development (p < 0.05). Crown-rump length and somite count decreased dose-dependently, while this decrease was not significant in the very low dose group (p > 0.05), which was most pronounced at doses of 2.5 and 12.5 mg/kg (p < 0.001). Acrylamide exposure dose-dependently decreased PCNA and increased caspase 3, with this change being significant at doses of 0.5 mg/kg and above (p < 0.001). BRE was downregulated at all acrylamide exposures except in the very low dose group (0.1 mg/kg). In conclusion, we find that acrylamide exposure (at 0.5 mg/kg and above) in post-gastrulation delays neural tube closure in chicken embryos by suppressing proliferation and apoptosis induction and downregulating BRE gene expression.


Subject(s)
Acrylamide , Dose-Response Relationship, Drug , Embryonic Development , Proliferating Cell Nuclear Antigen , Animals , Chick Embryo , Acrylamide/toxicity , Proliferating Cell Nuclear Antigen/metabolism , Embryonic Development/drug effects , Neural Tube/drug effects , Neural Tube/embryology , Caspase 3/metabolism , Caspase 3/genetics , Gene Expression Regulation, Developmental/drug effects
8.
Dev Growth Differ ; 66(5): 320-328, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38925637

ABSTRACT

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.


Subject(s)
Neural Plate , Neural Tube , Xenopus laevis , Animals , Neural Tube/embryology , Neural Tube/cytology , Neural Tube/metabolism , Neural Plate/embryology , Neural Plate/metabolism , Neural Plate/cytology , Xenopus laevis/embryology , Mesoderm/cytology , Mesoderm/embryology , Mesoderm/metabolism , Ectoderm/cytology , Ectoderm/metabolism , Microscopy, Atomic Force , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Embryo, Nonmammalian/embryology
9.
Brief Bioinform ; 25(4)2024 May 23.
Article in English | MEDLINE | ID: mdl-38851297

ABSTRACT

The development of the human central nervous system initiates in the early embryonic period until long after delivery. It has been shown that several neurological and neuropsychiatric diseases originate from prenatal incidents. Mathematical models offer a direct way to understand neurodevelopmental processes better. Mathematical modelling of neurodevelopment during the embryonic period is challenging in terms of how to 'Approach', how to initiate modelling and how to propose the appropriate equations that fit the underlying dynamics of neurodevelopment during the embryonic period while including the variety of elements that are built-in naturally during the process of neurodevelopment. It is imperative to answer where and how to start modelling; in other words, what is the appropriate 'Approach'? Therefore, one objective of this study was to tackle the mathematical issue broadly from different aspects and approaches. The approaches were divided into three embryonic categories: cell division, neural tube growth and neural plate growth. We concluded that the neural plate growth approach provides a suitable platform for simulation of brain formation/neurodevelopment compared to cell division and neural tube growth. We devised a novel equation and designed algorithms that include geometrical and topological algorithms that could fit most of the necessary elements of the neurodevelopmental process during the embryonic period. Hence, the proposed equations and defined mathematical structure would be a platform to generate an artificial neural network that autonomously grows and develops.


Subject(s)
Models, Biological , Neural Tube , Animals , Humans , Algorithms , Cell Division , Embryonic Development , Models, Neurological , Neural Networks, Computer , Neural Plate/cytology , Neural Plate/embryology , Neural Tube/embryology , Neurogenesis , Neurons/cytology
10.
Development ; 151(13)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38856082

ABSTRACT

A major challenge in biology is to understand how mechanical interactions and cellular behavior affect the shapes of tissues and embryo morphology. The extension of the neural tube and paraxial mesoderm, which form the spinal cord and musculoskeletal system, respectively, results in the elongated shape of the vertebrate embryonic body. Despite our understanding of how each of these tissues elongates independently of the others, the morphogenetic consequences of their simultaneous growth and mechanical interactions are still unclear. Our study investigates how differential growth, tissue biophysical properties and mechanical interactions affect embryonic morphogenesis during axial extension using a 2D multi-tissue continuum-based mathematical model. Our model captures the dynamics observed in vivo by time-lapse imaging of bird embryos, and reveals the underestimated influence of differential tissue proliferation rates. We confirmed this prediction in quail embryos by showing that decreasing the rate of cell proliferation in the paraxial mesoderm affects long-term tissue dynamics, and shaping of both the paraxial mesoderm and the neighboring neural tube. Overall, our work provides a new theoretical platform upon which to consider the long-term consequences of tissue differential growth and mechanical interactions on morphogenesis.


Subject(s)
Cell Proliferation , Mesoderm , Models, Biological , Morphogenesis , Neural Tube , Animals , Mesoderm/embryology , Mesoderm/cytology , Neural Tube/embryology , Neural Tube/cytology , Quail/embryology , Embryo, Nonmammalian/cytology , Embryonic Development/physiology , Viscosity
11.
Int J Mol Sci ; 25(11)2024 May 21.
Article in English | MEDLINE | ID: mdl-38891776

ABSTRACT

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.


Subject(s)
NIMA-Interacting Peptidylprolyl Isomerase , Neural Tube Defects , Tretinoin , Animals , Female , Humans , Mice , Apoptosis/drug effects , Cell Differentiation/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Down-Regulation/drug effects , Endoplasmic Reticulum Stress/drug effects , Neural Tube/metabolism , Neural Tube/drug effects , Neural Tube Defects/metabolism , Neural Tube Defects/genetics , Neural Tube Defects/chemically induced , Neurons/metabolism , Neurons/drug effects , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , NIMA-Interacting Peptidylprolyl Isomerase/metabolism , NIMA-Interacting Peptidylprolyl Isomerase/genetics , Oxidative Stress/drug effects , Receptor, Notch1/metabolism , Receptor, Notch1/genetics , Signal Transduction/drug effects , Tretinoin/metabolism , Tretinoin/pharmacology
12.
Dev Cell ; 59(15): 1940-1953.e10, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-38776925

ABSTRACT

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.


Subject(s)
Bone Morphogenetic Protein 4 , Neural Tube , Notochord , Organoids , Animals , Mice , Organoids/metabolism , Organoids/cytology , Neural Tube/metabolism , Neural Tube/cytology , Notochord/metabolism , Notochord/cytology , Bone Morphogenetic Protein 4/metabolism , Signal Transduction , Hepatocyte Nuclear Factor 3-beta/metabolism , Hepatocyte Nuclear Factor 3-beta/genetics , Hedgehog Proteins/metabolism , Hedgehog Proteins/genetics , Carrier Proteins/metabolism , Carrier Proteins/genetics , Gene Expression Regulation, Developmental , Bone Morphogenetic Proteins/metabolism
13.
Curr Top Dev Biol ; 159: 168-231, 2024.
Article in English | MEDLINE | ID: mdl-38729676

ABSTRACT

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.


Subject(s)
Body Patterning , Gene Expression Regulation, Developmental , Neural Tube , Signal Transduction , Neural Tube/embryology , Neural Tube/metabolism , Neural Tube/cytology , Animals , Body Patterning/genetics , Humans , Gene Regulatory Networks , Spinal Cord/embryology , Spinal Cord/cytology , Spinal Cord/metabolism , Cell Differentiation , Cell Movement
14.
Genesis ; 62(3): e23602, 2024 06.
Article in English | MEDLINE | ID: mdl-38721990

ABSTRACT

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.


Subject(s)
Cilia , Neural Tube , Animals , Mice , Cilia/metabolism , Cilia/genetics , Hedgehog Proteins/metabolism , Hedgehog Proteins/genetics , Neural Tube/embryology , Neural Tube/metabolism , Neural Tube Defects/genetics , Neural Tube Defects/metabolism , Point Mutation , Signal Transduction
15.
Sci Bull (Beijing) ; 69(14): 2260-2272, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-38702277

ABSTRACT

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.


Subject(s)
Autophagy , Heparan Sulfate Proteoglycans , Neural Tube Defects , Animals , Mice , Heparan Sulfate Proteoglycans/metabolism , Heparan Sulfate Proteoglycans/genetics , Humans , Neural Tube Defects/genetics , Neural Tube Defects/metabolism , Neural Tube Defects/pathology , Neural Tube/metabolism , Neural Tube/embryology , Neural Tube/pathology , Neural Stem Cells/metabolism , Neuroepithelial Cells/metabolism , Female , Male , Disease Models, Animal
16.
Dev Biol ; 511: 26-38, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38580174

ABSTRACT

In a developing embryo, formation of tissues and organs is remarkably precise in both time and space. Through cell-cell interactions, neighboring progenitors coordinate their activities, sequentially generating distinct types of cells. At present, we only have limited knowledge, rather than a systematic understanding, of the underlying logic and mechanisms responsible for cell fate transitions. The formation of the dorsal aspect of the spinal cord is an outstanding model to tackle these dynamics, as it first generates the peripheral nervous system and is later responsible for transmitting sensory information from the periphery to the brain and for coordinating local reflexes. This is reflected first by the ontogeny of neural crest cells, progenitors of the peripheral nervous system, followed by formation of the definitive roof plate of the central nervous system and specification of adjacent interneurons, then a transformation of roof plate into dorsal radial glia and ependyma lining the forming central canal. How do these peripheral and central neural branches segregate from common progenitors? How are dorsal radial glia established concomitant with transformation of the neural tube lumen into a central canal? How do the dorsal radial glia influence neighboring cells? This is only a partial list of questions whose clarification requires the implementation of experimental paradigms in which precise control of timing is crucial. Here, we outline some available answers and still open issues, while highlighting the contributions of avian models and their potential to address mechanisms of neural patterning and function.


Subject(s)
Neural Tube , Spinal Cord , Animals , Spinal Cord/embryology , Neural Tube/embryology , Neural Crest/embryology , Neural Crest/cytology , Neural Crest/physiology , Cell Differentiation/physiology , Neuroglia/physiology , Neuroepithelial Cells/cytology , Neuroepithelial Cells/physiology , Humans
17.
Development ; 151(10)2024 May 15.
Article in English | MEDLINE | ID: mdl-38682273

ABSTRACT

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.


Subject(s)
Neural Tube , Neurulation , Tomography, Optical Coherence , Animals , Female , Mice , Biomechanical Phenomena , Embryo, Mammalian/metabolism , Formate-Tetrahydrofolate Ligase/genetics , Formate-Tetrahydrofolate Ligase/metabolism , Formates/metabolism , Methylenetetrahydrofolate Dehydrogenase (NADP)/genetics , Methylenetetrahydrofolate Dehydrogenase (NADP)/metabolism , Mice, Knockout , Microscopy, Confocal , Mutation/genetics , Neural Tube/metabolism , Neural Tube Defects/genetics , Neural Tube Defects/metabolism , Neural Tube Defects/pathology , Neurulation/genetics , Tomography, Optical Coherence/methods
18.
Reprod Toxicol ; 125: 108576, 2024 04.
Article in English | MEDLINE | ID: mdl-38479591

ABSTRACT

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.


Subject(s)
Folic Acid Deficiency , MicroRNAs , Neural Tube Defects , Humans , Folic Acid , Neural Tube Defects/genetics , Neural Tube Defects/metabolism , Folic Acid Deficiency/complications , Folic Acid Deficiency/genetics , Folic Acid Deficiency/metabolism , Epigenesis, Genetic , DNA Methylation , MicroRNAs/genetics , Neural Tube/metabolism
19.
Curr Opin Genet Dev ; 86: 102179, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38490162

ABSTRACT

The chronologically ordered generation of distinct cell types is essential for the establishment of neuronal diversity and the formation of neuronal circuits. Recently, single-cell transcriptomic analyses of various areas of the developing vertebrate nervous system have provided evidence for the existence of a shared temporal patterning program that partitions neurons based on the timing of neurogenesis. In this review, I summarize the findings that lead to the proposal of this shared temporal program before focusing on the developing spinal cord to discuss how temporal patterning in general and this program specifically contributes to the ordered formation of neuronal circuits.


Subject(s)
Body Patterning , Gene Expression Regulation, Developmental , Neural Tube , Neurogenesis , Spinal Cord , Vertebrates , Animals , Neural Tube/growth & development , Neurogenesis/genetics , Vertebrates/growth & development , Vertebrates/genetics , Vertebrates/embryology , Body Patterning/genetics , Gene Expression Regulation, Developmental/genetics , Spinal Cord/growth & development , Spinal Cord/embryology , Neurons/cytology , Neurons/metabolism , Humans
20.
Am J Ophthalmol ; 261: 141-164, 2024 05.
Article in English | MEDLINE | ID: mdl-38311154

ABSTRACT

PURPOSE: To compare the prevalence, location and magnitude of optic nerve head (ONH) OCT-detected, exposed neural canal (ENC), externally oblique choroidal border tissue (EOCBT) and exposed scleral flange (ESF) regions in 122 highly myopic (Hi-Myo) versus 362 nonhighly myopic healthy (Non-Hi-Myo-Healthy) eyes. DESIGN: Cross-sectional study. METHODS: After OCT radial B-scan, ONH imaging, Bruch's membrane opening (BMO), the anterior scleral canal opening (ASCO), and the scleral flange opening (SFO) were manually segmented in each B-scan and projected to BMO reference plane. The direction and magnitude of BMO/ASCO offset and BMO/SFO offset as well as the location and magnitude of ENC, EOCBT and ESF regions, perineural canal (pNC) retinal nerve fiber layer thickness (RNFLT) and pNC choroidal thickness (CT) were calculated within 30° sectors relative to the Foveal-BMO (FoBMO) axis. Hi-ESF eyes were defined to be those with an ESF region ≥100 µms in at least 1 sector. RESULTS: Hi-Myo eyes more frequently demonstrated Hi-ESF regions (87/122) than Non-Hi-myo-Healthy eyes (73/362) and contained significantly larger ENC, EOCBT, and ESF regions (P < .001) which were greatest in magnitude and prevalence within the inferior-temporal FoBMO sectors where Hi-Myo pNC-RNFLT and pNCCT were thinnest. BMO/ASCO offset and the BMO/SFO offset were both significantly increased (P < .001) in the Hi-Myo eyes, with the latter demonstrating a greater increase. CONCLUSIONS: ENC region tissue remodeling that includes the scleral flange is enhanced in Hi-Myo compared to Non-Hi-Myo-Healthy eyes. Longitudinal studies are necessary to determine whether the presence of an ENC region influences ONH susceptibility to aging and/or glaucoma.


Subject(s)
Myopia , Optic Disk , Humans , Optic Disk/anatomy & histology , Tomography, Optical Coherence/methods , Neural Tube , Cross-Sectional Studies , Myopia/diagnosis , Bruch Membrane/anatomy & histology , Intraocular Pressure
SELECTION OF CITATIONS
SEARCH DETAIL