Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 95
Filter
1.
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
2.
Birth Defects Res ; 115(17): 1598-1607, 2023 10 15.
Article in English | MEDLINE | ID: mdl-37565787

ABSTRACT

BACKGROUND: Sugammadex is a modified gamma-cyclodextrin that has been developed with the goal of reversing the steroidal neuromuscular blocking agents. The aim of the present study is to investigate the effects of different sugammadex doses on embryologic and neural tube development in an early-stage chick embryo model. METHODS: A total of 100 specific pathogen-free, fertilized domestic chicken eggs were randomly divided into five groups (n = 20, each), and placed in an automatic cycle incubator. The eggs in the "control (C)" group were incubated without administration of any drug till the end of the experiment. Sub-blastodermic administration of 0.9% NaCl as vehicle control (VC) and different doses of sugammadex solutions prepared with the latter [2 mg/mL (LD), 4 mg/mL (MD), 16 mg/mL (HD)] were performed at 30 hr of incubation. All embryos were removed from the eggs at 72 hr when they were expected to reach Hamburger-Hamilton (HH) stages 19-20, then they were fixed, and evaluated histo-morphologically. RESULTS: Embryonic development was not observed in 11 eggs (1 in C, 1 in VC; 3 in LD, 3 in MD, and 3 in HD). All the developed embryos were compatible with the HH stages 19-20. A neural tube closure defect was detected in one embryo in the HD group. No statistically significant difference was found between the groups in terms of embryonic and neural tube developments. CONCLUSIONS: No significant association was found between the drug and adverse outcomes; however, a trend with dosing was seen. Further studies are required before conclude on safety and extrapolate these results to human beings.


Subject(s)
Neural Tube Defects , Neural Tube , Sugammadex , Animals , Chick Embryo , Chickens , Embryonic Development/drug effects , Neural Tube/growth & development , Neural Tube Defects/chemically induced , Sugammadex/adverse effects
3.
Genesis ; 59(12): e23453, 2021 12.
Article in English | MEDLINE | ID: mdl-34664392

ABSTRACT

The vertebrate Six (Sine oculis homeobox) family of homeodomain transcription factors plays critical roles in the development of several organs. Six1 plays a central role in cranial placode development, including the precursor tissues of the inner ear, as well as other cranial sensory organs and the kidney. In humans, mutations in SIX1 underlie some cases of Branchio-oto-renal (BOR) syndrome, which is characterized by moderate-to-severe hearing loss. We utilized CRISPR/Cas9 technology to establish a six1 mutant line in Xenopus tropicalis that is available to the research community. We demonstrate that at larval stages, the six1-null animals show severe disruptions in gene expression of putative Six1 target genes in the otic vesicle, cranial ganglia, branchial arch, and neural tube. At tadpole stages, six1-null animals display dysmorphic Meckel's, ceratohyal, and otic capsule cartilage morphology. This mutant line will be of value for the study of the development of several organs as well as congenital syndromes that involve these tissues.


Subject(s)
Branchio-Oto-Renal Syndrome/genetics , Congenital Abnormalities/genetics , Hearing Loss/genetics , Homeodomain Proteins/genetics , Xenopus Proteins/genetics , Animals , Branchial Region/growth & development , Branchial Region/pathology , Branchio-Oto-Renal Syndrome/physiopathology , CRISPR-Cas Systems/genetics , Congenital Abnormalities/pathology , Embryonic Development/genetics , Ganglia, Parasympathetic/growth & development , Ganglia, Parasympathetic/pathology , Gene Expression , Gene Expression Regulation, Developmental/genetics , Hearing Loss/physiopathology , Humans , Neural Tube/growth & development , Neural Tube/pathology , Skull/growth & development , Skull/pathology , Transcription Factors/genetics , Xenopus/genetics , Xenopus/growth & development
4.
Nat Commun ; 12(1): 3277, 2021 06 02.
Article in English | MEDLINE | ID: mdl-34078907

ABSTRACT

Generating properly differentiated embryonic structures in vitro from pluripotent stem cells remains a challenge. Here we show that instruction of aggregates of mouse embryonic stem cells with an experimentally engineered morphogen signalling centre, that functions as an organizer, results in the development of embryo-like entities (embryoids). In situ hybridization, immunolabelling, cell tracking and transcriptomic analyses show that these embryoids form the three germ layers through a gastrulation process and that they exhibit a wide range of developmental structures, highly similar to neurula-stage mouse embryos. Embryoids are organized around an axial chordamesoderm, with a dorsal neural plate that displays histological properties similar to the murine embryo neuroepithelium and that folds into a neural tube patterned antero-posteriorly from the posterior midbrain to the tip of the tail. Lateral to the chordamesoderm, embryoids display somitic and intermediate mesoderm, with beating cardiac tissue anteriorly and formation of a vasculature network. Ventrally, embryoids differentiate a primitive gut tube, which is patterned both antero-posteriorly and dorso-ventrally. Altogether, embryoids provide an in vitro model of mammalian embryo that displays extensive development of germ layer derivatives and that promises to be a powerful tool for in vitro studies and disease modelling.


Subject(s)
Body Patterning/genetics , Embryoid Bodies/metabolism , Embryonic Development/genetics , Mouse Embryonic Stem Cells/metabolism , Signal Transduction/genetics , Animals , Ectoderm/cytology , Ectoderm/growth & development , Ectoderm/metabolism , Embryo, Mammalian , Embryoid Bodies/cytology , Endoderm/cytology , Endoderm/growth & development , Endoderm/metabolism , GATA6 Transcription Factor/genetics , GATA6 Transcription Factor/metabolism , Gastrula/cytology , Gastrula/growth & development , Gastrula/metabolism , Gastrulation/genetics , Gene Expression Regulation, Developmental , HMGB Proteins/genetics , HMGB Proteins/metabolism , Mice , Mouse Embryonic Stem Cells/cytology , Nanog Homeobox Protein/genetics , Nanog Homeobox Protein/metabolism , Neural Tube/cytology , Neural Tube/growth & development , Neural Tube/metabolism , Notochord/cytology , Notochord/growth & development , Notochord/metabolism , SOXF Transcription Factors/genetics , SOXF Transcription Factors/metabolism
5.
Turk Neurosurg ; 31(4): 594-600, 2021.
Article in English | MEDLINE | ID: mdl-33978217

ABSTRACT

AIM: To investigate the effects of pregabalin on neural tube closure, and other potential effects on other organ systems in a chick embryo model. MATERIAL AND METHODS: Fertilized chicken eggs were divided into groups, and different doses of pregabalin was administered. All embryos were harvested in the 8th day of incubation, and investigated both macroscopically and microscopically against any developmental malformations caused by Pregabalin. RESULTS: Macroscopically not any malformations were detected but macrosomia was statistically significant in medium and high dose groups. Microscopically, vertebral lamina ossification was delayed in some embryos in high dose group but not interpreted as midline closure defect and also not statistically significant. Decrease in the number of renal glomerulus and increase in the tubular damage was statistically significant in medium and high dose groups. Cardiomegaly was also found in some embryos in middle and high dose groups but not statistically significant. CONCLUSION: The use of pregabalin does not cause neural tube closure defect in the embryo unless not exceed recommended maximum dose. Causing macrosomia instead of developmental retardation by Pregabalin is in conflict with the literature. This study revealed that Pregabalin causes fetal nephrotoxicity and macrosomia. These findings indicate that the use of Pregabalin in pregnancy still needs to be accounted as suspicious.


Subject(s)
Embryonic Development/drug effects , Neural Tube/drug effects , Pregabalin/toxicity , Teratogenesis/drug effects , Animals , Chick Embryo , Chickens/growth & development , Dose-Response Relationship, Drug , Neural Tube/embryology , Neural Tube/growth & development , Neural Tube Defects/chemically induced , Pregabalin/pharmacology , Toxicity Tests
6.
Int J Mol Sci ; 22(8)2021 Apr 10.
Article in English | MEDLINE | ID: mdl-33920095

ABSTRACT

Research on the development of the dorsal neural tube is particularly challenging. In this highly dynamic domain, a temporal transition occurs between early neural crest progenitors that undergo an epithelial-to-mesenchymal transition and exit the neural primordium, and the subsequent roof plate, a resident epithelial group of cells that constitutes the dorsal midline of the central nervous system. Among other functions, the roof plate behaves as an organizing center for the generation of dorsal interneurons. Despite extensive knowledge of the formation, emigration and migration of neural crest progenitors, little is known about the mechanisms leading to the end of neural crest production and the transition into a roof plate stage. Are these two mutually dependent or autonomously regulated processes? Is the generation of roof plate and dorsal interneurons induced by neural tube-derived factors throughout both crest and roof plate stages, respectively, or are there differences in signaling properties and responsiveness as a function of time? In this review, we discuss distinctive characteristics of each population and possible mechanisms leading to the shift between the above cell types.


Subject(s)
Cell Differentiation/genetics , Central Nervous System/growth & development , Neural Crest/growth & development , Neural Tube/growth & development , Animals , Bone Morphogenetic Proteins/genetics , Central Nervous System/metabolism , Embryonic Development/genetics , Gene Expression Regulation, Developmental/genetics , Interneurons/metabolism , Signal Transduction/genetics , Wnt Proteins/genetics
7.
Mol Med Rep ; 23(3)2021 03.
Article in English | MEDLINE | ID: mdl-33655333

ABSTRACT

Neural tube defects (NTDs) are the most serious and common birth defects in the clinical setting. The Notch signaling pathway has been implicated in different processes of the embryonic neural stem cells (NSCs) during neural tube development. The aim of the present study was to investigate the expression pattern and function of Notch1 (N1) in all­trans retinoic acid (atRA)­induced NTDs and NSC differentiation. A mouse model of brain abnormality was established by administering 28 mg/kg atRA, and then brain development was examined using hematoxylin and eosin (H&E) staining. The N1 expression pattern was detected in the brain of mice embryos via immunohistochemistry and western blotting. NSCs were extracted from the fetal brain of C57 BL/6 embryos at 18.5 days of pregnancy. N1, Nestin, neurofilament (NF), glial fibrillary acidic protein (GFAP) and galactocerebroside (GALC) were identified using immunohistochemistry. Moreover, N1, presenilin 1 (PS1), Nestin, NF, GFAP and GALC were detected via western blotting at different time points in the NSCs with control media or atRA media. H&E staining identified that the embryonic brain treated with atRA was more developed compared with the control group. N1 was downregulated in the embryonic mouse brain between days 11 and 17 in the atRA­treated group compared with the untreated group. The distribution of N1, Nestin, NF, GFAP and GALC was positively detected using immunofluorescence staining. Western blotting results demonstrated that there were significantly, synchronous decreased expression levels of N1 and PS1, but increased expression levels of NF, GFAP and GALC in NSCs treated with atRA compared with those observed in the controls (P<0.05). The results suggested that the N1 signaling pathway inhibited brain development and NSC differentiation. Collectively, it was found that atRA promoted mouse embryo brain development and the differentiation of NSCs by inhibiting the N1 pathway.


Subject(s)
Cell Differentiation/genetics , Embryonic Development/genetics , Neural Tube Defects/genetics , Receptor, Notch1/genetics , Animals , Cell Differentiation/drug effects , Gene Expression Regulation, Developmental/drug effects , Humans , Mice , Neural Stem Cells/metabolism , Neural Tube/growth & development , Neural Tube/metabolism , Neural Tube Defects/pathology , Tretinoin/pharmacology
8.
Reprod Toxicol ; 99: 160-167, 2021 01.
Article in English | MEDLINE | ID: mdl-32926990

ABSTRACT

In recent years, the development and implementation of animal-free approaches to chemical and pharmaceutical hazard and risk assessment has taken off. Alternative approaches are being developed starting from the perspective of human biology and physiology. Neural tube closure is a vital step that occurs early in human development. Correct closure of the neural tube depends on a complex interplay between proteins along a number of protein concentration gradients. The sensitivity of neural tube closure to chemical disturbance of signalling pathways such as the retinoid pathway, is well known. To map the pathways underlying neural tube closure, literature data on the molecular regulation of neural tube closure were collected. As the process of neural tube closure is highly conserved in vertebrates, the extensive literature available for the mouse was used whilst considering its relevance for humans. Thus, important cell compartments, regulatory pathways, and protein interactions essential for neural tube closure under physiological circumstances were identified and mapped. An understanding of aberrant processes leading to neural tube defects (NTDs) requires detailed maps of neural tube embryology, including the complex genetic signals and responses underlying critical cellular dynamical and biomechanical processes. The retinoid signaling pathway serves as a case study for this ontology because of well-defined crosstalk with the genetic control of neural tube patterning and morphogenesis. It is a known target for mechanistically-diverse chemical structures that disrupt neural tube closure The data presented in this manuscript will set the stage for constructing mathematical models and computer simulation of neural tube closure for human-relevant AOPs and predictive toxicology.


Subject(s)
Models, Biological , Neural Tube/growth & development , Animals , Computer Simulation , Ectoderm , Embryonic Development , Humans , Mesoderm , Mice , Neural Crest , Neural Plate , Neural Tube Defects , Notochord , Systems Biology , Tretinoin/metabolism
9.
Methods Mol Biol ; 2179: 107-114, 2021.
Article in English | MEDLINE | ID: mdl-32939716

ABSTRACT

Live embryo imaging may provide a wealth of information on intact cell and tissue dynamics, but can be technically challenging to sustain embryo orientation and health for long periods under a microscope. In this protocol, we describe an in vivo method to mount and image cell movements during the epithelial-to-mesenchymal transition (EMT) of neural crest cells within the chick dorsal neural tube. We focus on describing the collection of images and data preparation for image analysis throughout the developmental stages HH15-21 in the chick trunk. Trunk neural crest cell EMT is crucial to development of the peripheral nervous system and pigment cell patterning. The methods we describe may also be applied to other cell and tissue phenomena at various chick developmental stages with some modifications.


Subject(s)
Epithelial-Mesenchymal Transition/genetics , Molecular Imaging/methods , Neural Crest/ultrastructure , Neural Tube/ultrastructure , Animals , Cell Movement/genetics , Chick Embryo , Neural Tube/growth & development
10.
Elife ; 92020 12 21.
Article in English | MEDLINE | ID: mdl-33345773

ABSTRACT

Spinal commissural axon navigation across the midline in the floor plate requires repulsive forces from local Slit repellents. The long-held view is that Slits push growth cones forward and prevent them from turning back once they became sensitized to these cues after midline crossing. We analyzed with fluorescent reporters Slits distribution and FP glia morphology. We observed clusters of Slit-N and Slit-C fragments decorating a complex architecture of glial basal process ramifications. We found that PC2 proprotein convertase activity contributes to this pattern of ligands. Next, we studied Slit-C acting via PlexinA1 receptor shared with another FP repellent, the Semaphorin3B, through generation of a mouse model baring PlexinA1Y1815F mutation abrogating SlitC but not Sema3B responsiveness, manipulations in the chicken embryo, and ex vivo live imaging. This revealed a guidance mechanism by which SlitC constantly limits growth cone exploration, imposing ordered and forward-directed progression through aligned corridors formed by FP basal ramifications.


Subject(s)
Commissural Interneurons/physiology , Spinal Cord/growth & development , Animals , Axons/physiology , Blotting, Western , Chick Embryo , Growth Cones/physiology , Mice , Microscopy, Fluorescence , Neural Tube/embryology , Neural Tube/growth & development , Spinal Cord/embryology
11.
Development ; 147(24)2020 12 23.
Article in English | MEDLINE | ID: mdl-33361092

ABSTRACT

Using the zebrafish neural tube as a model, we uncover the in vivo mechanisms allowing the generation of two opposing apical epithelial surfaces within the centre of an initially unpolarised, solid organ. We show that Mpp5a and Rab11a play a dual role in coordinating the generation of ipsilateral junctional belts whilst simultaneously releasing contralateral adhesions across the centre of the tissue. We show that Mpp5a- and Rab11a-mediated resolution of cell-cell adhesions are both necessary for midline lumen opening and contribute to later maintenance of epithelial organisation. We propose that these roles for both Mpp5a and Rab11a operate through the transmembrane protein Crumbs. In light of a recent conflicting publication, we also clarify that the junction-remodelling role of Mpp5a is not specific to dividing cells.


Subject(s)
Guanylate Cyclase/genetics , Morphogenesis/genetics , Zebrafish Proteins/genetics , Zebrafish/growth & development , rab GTP-Binding Proteins/genetics , Animals , Cell Polarity/genetics , Epithelial Cells/metabolism , Gene Expression Regulation, Developmental/genetics , Intercellular Junctions/genetics , Membrane Proteins , Neural Tube/growth & development , Zebrafish/genetics
12.
Elife ; 92020 10 26.
Article in English | MEDLINE | ID: mdl-33103996

ABSTRACT

Neural tube closure defects are a major cause of infant mortality, with exencephaly accounting for nearly one-third of cases. However, the mechanisms of cranial neural tube closure are not well understood. Here, we show that this process involves a tissue-wide pattern of apical constriction controlled by Sonic hedgehog (Shh) signaling. Midline cells in the mouse midbrain neuroepithelium are flat with large apical surfaces, whereas lateral cells are taller and undergo synchronous apical constriction, driving neural fold elevation. Embryos lacking the Shh effector Gli2 fail to produce appropriate midline cell architecture, whereas embryos with expanded Shh signaling, including the IFT-A complex mutants Ift122 and Ttc21b and embryos expressing activated Smoothened, display apical constriction defects in lateral cells. Disruption of lateral, but not midline, cell remodeling results in exencephaly. These results reveal a morphogenetic program of patterned apical constriction governed by Shh signaling that generates structural changes in the developing mammalian brain.


Subject(s)
Hedgehog Proteins/physiology , Neural Tube/growth & development , Animals , Brain/embryology , Cell Shape , Hedgehog Proteins/metabolism , Mice , Mice, Inbred C57BL , Neural Crest/embryology , Neural Tube/embryology
13.
Elife ; 92020 03 31.
Article in English | MEDLINE | ID: mdl-32228864

ABSTRACT

An extracellular matrix of Fibronectin adheres the neural tube to the two flanking columns of paraxial mesoderm and is required for normal vertebrate development. Here, we find that the bilaterally symmetric interfaces between the zebrafish neural tube and paraxial mesoderm function as optimally engineered adhesive lap joints with rounded edges, graded Fibronectin 'adhesive' and an arced adhesive spew filet. Fibronectin is a 'smart adhesive' that remodels to the lateral edges of the neural tube-paraxial mesoderm interfaces where shear stress is highest. Fibronectin remodeling is mechanically responsive to contralateral variation morphogenesis, and Fibronectin-mediated inter-tissue adhesion is required for bilaterally symmetric morphogenesis of the paraxial mesoderm. Strikingly, however, perturbation of the Fibronectin matrix rescues the neural tube convergence defect of cadherin 2 mutants. Therefore, Fibronectin-mediated inter-tissue adhesion dynamically coordinates bilaterally symmetric morphogenesis of the vertebrate trunk but predisposes the neural tube to convergence defects that lead to spina bifida.


In embryos, the spinal cord starts out as a flat sheet of cells that curls up to form a closed cylinder called the neural tube. The folding tube is attached to the surrounding tissues through an extracellular matrix of proteins and sugars. Overlapping strands of a protein from the extracellular matrix called Fibronectin connect the neural tube to adjacent tissues, like a kind of biological glue. However, it remained unclear what effect this attachment had on the embryonic development of the spinal cord. Connecting two overlapping objects with glue to form what is known as an 'adhesive lap joint' is common in fields such as woodworking and aeronautical engineering. The glue in these joints comes under shearing stress whenever the two objects it connects try to pull apart. But, thanks to work in engineering, it is possible to predict how different joints will perform under tension. Now, Guillon et al. have deployed these engineering principles to shed light on neural tube development. Using zebrafish embryos and computational models, Guillon et al. investigated what happens when the strength of the adhesive lap joints in the developing spine changes. This revealed that Fibronectin works like a smart adhesive: rather than staying in one place like a conventional glue, it moves around. As the neural tube closes, cells remodel the Fibronectin, concentrating it on the areas under the highest stress. This seemed to both help and hinder neural tube development. On the one hand, by anchoring the tube equally to the left and right sides of the embryo, the Fibronectin glue helped the spine to develop symmetrically. On the other hand, the strength of the adhesive lap joints made it harder for the neural tube to curl up and close. If the neural tube fails to close properly, it can lead to birth defects like spina bifida. One of the best-known causes of these birth defects in humans is a lack of a vitamin known as folic acid. Cell culture experiments suggest that this might have something to do with the mechanics of the cells during development. It may be that faulty neural tubes could close more easily if they were able to unglue themselves from the surrounding tissues. Further use of engineering principles could shed more light on this idea in the future.


Subject(s)
Fibronectins/physiology , Mesoderm/physiology , Morphogenesis , Neural Tube/growth & development , Spine/growth & development , Adhesives , Animals , Extracellular Matrix/physiology , Female , Humans , Male , Spine/anatomy & histology , Zebrafish/physiology
14.
Cell Biochem Funct ; 38(7): 905-920, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32129905

ABSTRACT

MicroRNAs (miRNAs) provide context-dependent transcriptional regulation of genes comprising signalling networks throughout the developing organism including morphogenesis of the embryonic neural tube (NT). Using a high-sensitivity, high-coverage microarray analysis platform, miRNA expression in the murine embryonic NT during the critical stages of its formation was examined. Analysis of a number of differentially expressed (DE) miRNAs enabled identification of several gene targets associated with cellular processes essential for normal NT development. Using computational pathway analysis, interactive biologic networks and functional relationships connecting DE miRNAs with their targeted messenger RNAs (mRNAs) were identified. Potential mRNA targets and a key signal transduction pathway governing critical cellular processes indispensable for normal mammalian neurulation were also identified. RNA preparations were also used to hybridize both miRNA arrays and mRNA arrays allowing miRNA-mRNA target analysis using data of DE miRNAs and DE mRNAs - co-expressed in the same developing NT tissue samples. Identification of these miRNA targets provides key insight into the epigenetic regulation of NT development as well as into potential mechanistic underpinning of NT defects. SIGNIFICANCE OF THE STUDY: This study underscores the premise that microRNAs are potential coordinators of normal neural tube (NT) formation, via regulation of the crucial, planar cell polarity pathway. Any alteration in their expression during neurulation would result in abnormal NT development.


Subject(s)
MicroRNAs/metabolism , Neural Tube/metabolism , Animals , Cell Polarity , Embryonic Development/genetics , Female , Gene Expression Regulation, Developmental , Male , Mice , Mice, Inbred ICR , Neural Tube/growth & development , RNA, Messenger/metabolism , Signal Transduction/genetics , Wnt Signaling Pathway
15.
Bioessays ; 42(3): e1900186, 2020 03.
Article in English | MEDLINE | ID: mdl-32078177

ABSTRACT

The brain ventricular system is a series of connected cavities, filled with cerebrospinal fluid (CSF), that forms within the vertebrate central nervous system (CNS). The hollow neural tube is a hallmark of the chordate CNS, and a closed neural tube is essential for normal development. Development and function of the ventricular system is examined, emphasizing three interdigitating components that form a functional system: ventricle walls, CSF fluid properties, and activity of CSF constituent factors. The cellular lining of the ventricle both can produce and is responsive to CSF. Fluid properties and conserved CSF components contribute to normal CNS development. Anomalies of the CSF/ventricular system serve as diagnostics and may cause CNS disorders, further highlighting their importance. This review focuses on the evolution and development of the brain ventricular system, associated function, and connected pathologies. It is geared as an introduction for scholars with little background in the field.


Subject(s)
Cerebral Ventricles/growth & development , Cerebral Ventricles/metabolism , Cerebrospinal Fluid/metabolism , Animals , Biological Evolution , Brain Diseases/metabolism , Cerebral Ventricles/cytology , Cerebrospinal Fluid Pressure/physiology , Cerebrospinal Fluid Proteins/metabolism , Cilia/metabolism , Epithelium/growth & development , Epithelium/metabolism , Humans , Kinetics , Neural Tube/cytology , Neural Tube/growth & development , Neural Tube/metabolism , Signal Transduction
16.
Stem Cell Res Ther ; 10(1): 389, 2019 12 16.
Article in English | MEDLINE | ID: mdl-31842983

ABSTRACT

OBJECTIVE: Neural tube defects (NTDs) are the most serious and common birth defects in the clinic. The SRY-related HMG box B1 (SoxB1) gene family has been implicated in different processes of early embryogenesis. Sox19b is a maternally expressed gene in the SoxB1 family that is found in the region of the presumptive central nervous system (CNS), but its role and mechanism in embryonic neural stem cells (NSCs) during neural tube development have not yet been explored. Considering that Sox19b is specific to bony fish, we intended to investigate the role and mechanism of Sox19b in neural tube development in zebrafish embryos. MATERIAL AND METHODS: Morpholino (MO) antisense oligonucleotides were used to construct a Sox19b loss-of-function zebrafish model. The phenotype and the expression of related genes were analysed by in situ hybridization and immunolabelling. Epigenetic modifications were detected by western blot and chromatin immunoprecipitation. RESULTS: In this study, we found that zebrafish embryos exhibited a reduced or even deleted forebrain phenotype after the expression of the Sox19b gene was inhibited. Moreover, we found for the first time that knockdown of Sox19b reduced the proliferation of NSCs; increased the transcription levels of Ngn1, Ascl1, HuC, Islet1, and cyclin-dependent kinase (CDK) inhibitors; and led to premature differentiation of NSCs. Finally, we found that knockdown of Sox19b decreased the levels of EZH2/H3K27me3 and decreased the level of H3K27me3 at the promoters of Ngn1 and ascl1a. CONCLUSION: Together, our data demonstrate that Sox19b plays an essential role in early NSC proliferation and differentiation through EZH2-mediated histone methylation in neural tube development. This study established the role of transcription factor Sox19b and epigenetic factor EZH2 regulatory network on NSC development, which provides new clues and theoretical guidance for the clinical treatment of neural tube defects.


Subject(s)
Enhancer of Zeste Homolog 2 Protein/metabolism , Histones/metabolism , Neural Stem Cells/metabolism , Neural Tube/growth & development , SOX Transcription Factors/metabolism , Zebrafish Proteins/metabolism , Animals , Cell Differentiation/physiology , Disease Models, Animal , Gene Knockdown Techniques , Methylation , Neural Stem Cells/cytology , Neural Tube/cytology , Neural Tube/metabolism , Prosencephalon/embryology , Prosencephalon/metabolism , SOX Transcription Factors/biosynthesis , SOX Transcription Factors/genetics , Zebrafish , Zebrafish Proteins/biosynthesis , Zebrafish Proteins/genetics
17.
Sci Adv ; 5(12): eaax5933, 2019 12.
Article in English | MEDLINE | ID: mdl-31844664

ABSTRACT

Despite its importance in central nervous system development, development of the human neural tube (NT) remains poorly understood, given the challenges of studying human embryos, and the developmental divergence between humans and animal models. We report a human NT development model, in which NT-like tissues, neuroepithelial (NE) cysts, are generated in a bioengineered neurogenic environment through self-organization of human pluripotent stem cells (hPSCs). NE cysts correspond to the neural plate in the dorsal ectoderm and have a default dorsal identity. Dorsal-ventral (DV) patterning of NE cysts is achieved using retinoic acid and/or sonic hedgehog and features sequential emergence of the ventral floor plate, P3, and pMN domains in discrete, adjacent regions and a dorsal territory progressively restricted to the opposite dorsal pole. This hPSC-based, DV patterned NE cyst system will be useful for understanding the self-organizing principles that guide NT patterning and for investigations of neural development and neural disease.


Subject(s)
Body Patterning/genetics , Neural Tube/growth & development , Neurogenesis/genetics , Pluripotent Stem Cells/cytology , Animals , Central Nervous System/growth & development , Central Nervous System/metabolism , Ectoderm/growth & development , Ectoderm/metabolism , Embryonic Development/genetics , Gene Expression Regulation, Developmental/genetics , Hedgehog Proteins/genetics , Humans , Neural Tube/metabolism , Pluripotent Stem Cells/metabolism , Tretinoin/metabolism
18.
Neural Dev ; 14(1): 7, 2019 03 13.
Article in English | MEDLINE | ID: mdl-30867016

ABSTRACT

In the developing neural tube in chicken and mammals, neural stem cells proliferate and differentiate according to a stereotyped spatiotemporal pattern. Several actors have been identified in the control of this process, from tissue-scale morphogens patterning to intrinsic determinants in neural progenitor cells. In a previous study (Bonnet et al. eLife 7, 2018), we have shown that the CDC25B phosphatase promotes the transition from proliferation to differentiation by stimulating neurogenic divisions, suggesting that it acts as a maturating factor for neural progenitors. In this previous study, we set up a mathematical model linking fixed progenitor modes of division to the dynamics of progenitors and differentiated populations. Here, we extend this model over time to propose a complete dynamical picture of this process. We start from the standard paradigm that progenitors are homogeneous and can perform any type of divisions (proliferative division yielding two progenitors, asymmetric neurogenic divisions yielding one progenitor and one neuron, and terminal symmetric divisions yielding two neurons). We calibrate this model using data published by Saade et al. (Cell Reports 4, 2013) about mode of divisions and population dynamics of progenitors/neurons at different developmental stages. Next, we explore the scenarios in which the progenitor population is actually split into two different pools, one of which is composed of cells that have lost the capacity to perform proliferative divisions. The scenario in which asymmetric neurogenic division would induce such a loss of proliferative capacity appears very relevant.


Subject(s)
Cell Differentiation/physiology , Cell Proliferation/physiology , Models, Biological , Neural Stem Cells/physiology , Neural Tube/cytology , Neural Tube/growth & development , Spinal Cord/cytology , Spinal Cord/growth & development , cdc25 Phosphatases/physiology , Animals
19.
Hum Mol Genet ; 28(10): 1726-1737, 2019 05 15.
Article in English | MEDLINE | ID: mdl-30689861

ABSTRACT

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


Subject(s)
DNA-Binding Proteins/genetics , Interferon Regulatory Factors/genetics , Neurulation/genetics , Transcription Factor AP-2/genetics , Transcription Factors/genetics , Animals , Conserved Sequence/genetics , Gene Expression Regulation, Developmental/genetics , Humans , Mice , Mutation , Neural Tube/growth & development , Neural Tube/pathology , Neural Tube Defects/genetics , Neural Tube Defects/pathology , Signal Transduction/genetics , Spinal Dysraphism/genetics , Spinal Dysraphism/pathology
20.
Methods Mol Biol ; 1863: 47-63, 2018.
Article in English | MEDLINE | ID: mdl-30324592

ABSTRACT

Developmental processes are inherently dynamic and understanding them requires quantitative measurements of gene and protein expression levels in space and time. While live imaging is a powerful approach for obtaining such data, it is still a challenge to apply it over long periods of time to large tissues, such as the embryonic spinal cord in mouse and chick. Nevertheless, dynamics of gene expression and signaling activity patterns in this organ can be studied by collecting tissue sections at different developmental stages. In combination with immunohistochemistry, this allows for measuring the levels of multiple developmental regulators in a quantitative manner with high spatiotemporal resolution. The mean protein expression levels over time, as well as embryo-to-embryo variability can be analyzed. A key aspect of the approach is the ability to compare protein levels across different samples. This requires a number of considerations in sample preparation, imaging and data analysis. Here we present a protocol for obtaining time course data of dorsoventral expression patterns from mouse and chick neural tube in the first 3 days of neural tube development. The described workflow starts from embryo dissection and ends with a processed dataset. Software scripts for data analysis are included. The protocol is adaptable and instructions that allow the user to modify different steps are provided. Thus, the procedure can be altered for analysis of time-lapse images and applied to systems other than the neural tube.


Subject(s)
Embryo, Mammalian/cytology , Gene Expression Regulation, Developmental , Morphogenesis , Neural Tube/embryology , Neurogenesis , Time-Lapse Imaging/methods , Animals , Cell Differentiation , Chickens , Embryo, Mammalian/physiology , Embryonic Development , Image Processing, Computer-Assisted , Mice , Neural Tube/growth & development , Neural Tube/physiology , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...