Fact-checked by Grok 2 weeks ago

Neuroectoderm

The neuroectoderm is a specialized portion of the ectoderm, one of the three primary germ layers formed during gastrulation in the third week of embryonic development, that serves as the precursor to the nervous system. It arises through a process known as neural induction, where signals from the underlying notochord and mesoderm prompt ectodermal cells to differentiate into neuroepithelial cells, forming the neural plate around 18 days post-fertilization in humans. This structure is critical for establishing the foundational architecture of the central and peripheral nervous systems. During , the folds inward to create neural folds that fuse to form the , which closes by approximately day 28 of embryogenesis, with the neuroectoderm differentiating into the neuroepithelium that lines this tube. The subsequently develops into the (CNS), comprising the , , and , while cells at the junction of the and overlying give rise to cells. These cells migrate extensively to form diverse structures, including the peripheral nervous system (sensory ganglia, autonomic neurons, and Schwann cells), melanocytes, craniofacial and , odontoblasts of teeth, and components of the . The induction and patterning of the neuroectoderm are tightly regulated by signaling molecules, such as fibroblast growth factors (FGF) secreted by the to promote neural fate, and inhibitors like chordin, noggin, and that block (BMP) signaling from the to prevent non-neural ectodermal differentiation. Additional pathways, including Wnt and Sonic hedgehog, contribute to rostral-caudal and dorsal-ventral patterning along the neuroectoderm. Disruptions in these processes can lead to congenital anomalies, such as neural tube defects (e.g., or ), highlighting the neuroectoderm's vulnerability during early development. By the fourth week, the anterior neural tube expands into primary brain vesicles (, , and ), setting the stage for further CNS specialization.

Definition and Overview

Definition

The neuroectoderm is the specialized portion of the that differentiates into neural tissues during early embryogenesis, giving rise to the central and peripheral nervous systems. This layer emerges as part of the trilaminar embryo following , where the forms the outermost . Neuroectodermal cells exhibit distinct morphological and molecular characteristics, including a pseudostratified epithelial with apicobasal , elongated cells, and a high nucleus-to-cytoplasm ratio that supports proliferative capacity. These cells also express neural-specific markers such as the , which maintains progenitor identity and inhibits premature differentiation, and the intermediate filament protein Nestin, indicative of potential. In contrast to the surface ectoderm, which remains external and differentiates into the and associated structures like and nails, the neuroectoderm internalizes through to form the , the precursor to the , while contributing to the for peripheral elements.

Historical Context

The early understanding of neuroectoderm began with observations of embryonic germ layers in the mid-19th century, building on the foundational work of Christian Pander and , who described the as one of the primary germ layers in embryos. In 1874, Wilhelm His provided detailed histological descriptions of the as a thickened region of in and embryos, identifying it as the precursor to neural structures through serial sectioning and three-dimensional reconstructions, which marked a shift toward mechanistic explanations of form in . His's work in Unsere Körperform emphasized the ectodermal origin of the , distinguishing it from surrounding epidermal ectoderm and laying the groundwork for recognizing specialized ectodermal derivatives. The term "neuroectoderm" emerged in the late 19th and early 20th centuries amid efforts to classify ectodermal specializations, with embryologists like Oscar Hertwig contributing through studies on formation and fertilization in sea urchins and amphibians, which highlighted the 's role in neural . Hertwig's comprehensive textbooks on , such as Lehrbuch der Entwicklungsgeschichte des Menschen (1906), integrated these concepts, influencing the for neural-specifying ectoderm. By the early 1900s, the term gained traction in English-language literature to denote the ectodermal population fated for formation, reflecting advances in comparative across species. A pivotal advancement came in 1924 with the experiments of and Hilde Mangold, who transplanted dorsal lip tissue from amphibian gastrulae to induce a secondary neural axis in host embryos, demonstrating that presumptive could be directed to form neuroectoderm through inductive signals from an "organizer" region. This discovery, detailed in their seminal paper, established the inducible nature of neuroectoderm and shifted focus from preformationist views to interactive processes in development, earning Spemann the 1935 in or . Throughout the , studies of neuroectoderm evolved from descriptive —relying on and —to molecular , particularly after the with the integration of and biochemistry. Key transitions included the identification of gradients and signaling molecules in the –1990s, enabling dissection of mechanisms at the ular level, as reviewed in historical analyses of the field. This progression transformed neuroectoderm from observational anatomy to a gene-regulatory framework, underscoring its role in vertebrate specification.

Embryonic Formation

Role in Gastrulation

Gastrulation represents a critical phase in early embryogenesis, transforming the bilaminar disc into a trilaminar structure by establishing the three primary germ layers: , , and . In human embryos, this process unfolds during the third week of development, approximately days 15 to 21 post-fertilization, with the emerging around day 15 to guide cellular rearrangements. Epiblast cells ingress through the , displacing the to form the definitive while contributing to the intraembryonic ; the non-ingressing epiblast cells persist as the presumptive , which covers the dorsal surface of the embryo. Within the , specification toward neuroectoderm occurs as cells along the dorsal aspect of the adopt a neural fate through a mechanism of default . This default pathway posits that ectodermal cells are predisposed to become neuroectoderm unless actively instructed otherwise to pursue alternative fates, such as epidermal . During , this commitment arises from the intrinsic properties of ectodermal progenitors in the absence of inhibitory signals, positioning neuroectoderm as the of ectodermal .81853-X)01620-5) Spatially, the prospective neuroectoderm occupies a medial position within the epiblast-derived ectodermal layer, directly overlying the emerging notochord and adjacent to lateral domains fated for non-neural ectoderm. In vertebrates such as amphibians and mammals, this medial ectoderm undergoes commitment to the neural lineage shortly after gastrulation concludes, typically marked by cellular changes that prepare for further differentiation, with brief influence from inductive signals emanating from the subjacent mesoderm.00113-2)

Neural Induction Process

Neural induction is the process by which presumptive ectodermal cells are specified to become neuroectoderm, primarily through signals from the underlying dorsal mesoderm during gastrulation. The classic model, established in amphibians, originates from the seminal transplantation experiments of Spemann and Mangold, who demonstrated that the dorsal blastopore lip—termed the Spemann organizer—induces the overlying ectoderm to form neural tissue rather than epidermis. This organizer secretes diffusible factors that reprogram competent ectoderm cells toward a neural fate, a mechanism first observed in newt embryos. The process unfolds in sequential steps: first, the acquires , the intrinsic ability to respond to inductive signals, which occurs during blastula stages and is progressively lost by the end of in species like (stages 10-12). Following competence, activation induces the initial neural fate, specifying anterior neural character such as identity, as proposed in Nieuwkoop's model. Subsequent transformation refines this by imposing posterior identities (e.g., and ) through additional signals, ensuring proper anterior-posterior patterning of the neuroectoderm. Central to this process are (BMP) inhibitors secreted by the Spemann organizer, including Noggin, Chordin, and , which bind and sequester BMP ligands like BMP4 in the , thereby blocking their receptor activation and promoting the "default" neural state in . Noggin was identified as a key dorsalizing factor that mimics organizer activity when expressed in ventral , while Chordin and similarly antagonize BMP signaling to ventralize and neuralize tissues.90313-9)00673-4) This inhibition relieves BMP-mediated epidermal promotion, allowing neural gene expression to proceed. While conserved across vertebrates, neural induction shows variations; in mammals like the mouse, Nodal signaling from the and initially specifies pluripotency and prevents precocious neural differentiation in the epiblast, before organizer-derived antagonists initiate neural commitment during . This early Nodal role integrates with the default model, ensuring timed progression to neuroectoderm in the absence of mesendodermal induction.

Derivatives and Differentiation

Neural Plate Development

The neural plate forms through the thickening of the neuroectoderm into a , a process that begins around embryonic day 18 in humans during the third week of . This thickening occurs as ectodermal cells overlying the and prechordal undergo morphological changes, establishing the foundational structure for the . Neural induction, which precedes this stage, primes the for these transformations by inhibiting signaling in the dorsal midline. Key cellular processes drive the neural plate's development, including apical constriction of neuroepithelial , which narrows the apical surface and elongates the basally; convergent extension through mediolateral intercalation, which narrows and elongates the plate; and midline convergence that shapes the median hinge point along the plate's central . These coordinated behaviors, mediated by actomyosin contractility and planar pathways, transform the flat neuroectoderm into a defined midline structure without yet initiating folding. The median hinge point serves as a pivotal region where adopt a wedge-like shape, facilitating subsequent bending. Patterning along the establishes regional fates, with the anterior region destined for development and the posterior region for and , guided by opposing gradients of signaling molecules. High anterior levels of Wnt and FGF inhibitors promote identity, while posterior gradients of Wnt, FGF, and posteriorize the tissue toward and fates, creating a gradient-dependent anterior-posterior axis during . This spatial organization ensures precise allocation of neural progenitors to their prospective domains. Molecular markers confirm neural plate specification, with upregulation of Sox1, Sox2, and Sox3 transcription factors in the neuroepithelial cells, which maintain progenitor identity and promote neural differentiation. Concurrently, N-cadherin expression increases, stabilizing cell-cell adhesions and reinforcing the epithelial integrity of the plate while suppressing non-neural fates. These markers are essential for the plate's coherence and its transition to further morphogenetic stages.

Neural Tube Formation

The , the precursor to the , forms from the through primary , a process that transforms the flat epithelial sheet into a hollow cylindrical structure via and fusion of its edges. This occurs primarily in the and regions during early embryonic development. Primary neurulation begins with the elevation and convergence of the 's lateral edges to form neural folds, driven by bending at specific hinge points. The median hinge point (MHP), located midline over the , and paired dorsolateral hinge points (DLHPs) near the plate's edges, facilitate this folding; neuroepithelial cells at these sites adopt a wedge shape through apical constriction and basal expansion, creating the neural groove between the folds. As the folds elevate and approximate, they fuse dorsally to enclose the , progressing from an open to a closed structure. In human embryos, neural tube closure initiates at the level around day 22 of , with fusion beginning at the site corresponding to the future lower medulla and upper spinal . The process then proceeds bidirectionally like a : the anterior (rostral) neuropore closes by day 25, and the posterior (caudal) neuropore by day 27, completing enclosure of the rostral to the lower sacral region. Failure in this closure sequence can result in neural tube defects, such as or , highlighting the precision required. Underlying these morphological changes are dynamic cellular behaviors in the neuroepithelium. Convergent extension narrows the mediolateral width of the while elongating it along the anterior-posterior axis, achieved through mediolateral intercalation where cells rearrange via polarized protrusions and adhesions. Simultaneously, wedge-shaped neuroepithelial cells at the points drive by contracting apically (via actomyosin) and expanding basally, generating the forces necessary for folding without external pulling from surrounding tissues. Upon closure, the neural tube's lumen establishes the ventricular zone, an inner layer of proliferative neuroepithelial cells surrounding the central cavity. This lumen persists as the brain's ventricles (lateral, third, and fourth) and the spinal cord's , providing a fluid-filled space for circulation and serving as the site for early from the ventricular zone.

Neural Crest Contribution

The neural crest originates from a specialized population of cells at the dorsal border of the neural folds during , positioned between the neuroectoderm and the overlying surface . These cells are induced through interactions involving (BMP) and Wnt signaling pathways emanating from the ectoderm and surface , which specify their multipotent fate and distinguish them from the adjacent neuroectoderm that forms the . Prior to migration, neural crest cells undergo an epithelial-to-mesenchymal transition (), a critical process that transforms their epithelial morphology into a motile mesenchymal state, allowing delamination from the and dispersal throughout the . This is regulated by key transcription factors, including Slug (Snail2), , and , which repress epithelial markers like E-cadherin and promote mesenchymal traits such as increased and invasiveness. Neural crest cells exhibit remarkable multipotency, differentiating into a wide array of cell types that contribute to both neural and non-neural structures. In the peripheral nervous system, they form sensory and autonomic neurons, Schwann cells, and satellite glia; additionally, they generate melanocytes for pigmentation, mesenchymal cells that ossify into craniofacial cartilage and , and chromaffin cells of the . Migration of neural crest cells follows distinct spatiotemporal pathways tailored to their axial level and derivative fates. In the trunk, early emigrating cells travel ventromedially through the anterior sclerotome of somites to populate dorsal root and , while later waves use circumferential intersomitic routes or dorsolateral paths beneath the to reach sites for . In contrast, cranial cells migrate in orderly streams into the branchial arches, contributing to the formation of and associated connective tissues.

Molecular Mechanisms

Key Transcription Factors

The specification and maintenance of neuroectoderm rely on a network of transcription factors that orchestrate programs essential for neural fate commitment and identity. Among these, the SoxB1 family members—Sox1, , and Sox3—play pivotal roles, with overlapping yet distinct functions in establishing and sustaining the neuroectodermal state. Sox2 is a central regulator that maintains the pluripotency of early neuroectodermal progenitors while promoting neural lineage progression. In embryonic stem cells and early neuroepithelium, Sox2 binds to enhancer regions of neural genes, sustaining self-renewal and preventing premature differentiation into non-neural ectoderm. Sox2 interacts with Pou5f1 (also known as Oct4) to form a core pluripotency network in the nascent neuroectoderm, where their cooperative binding activates stemness genes and represses alternative fates. This partnership is crucial during the initial stages of neural induction, helping to lock in the neuroectodermal identity. Additionally, Sox2 mutually represses epidermal transcription factors such as Tfap2a (AP-2α), ensuring exclusive commitment to the neural pathway by inhibiting surface ectoderm markers. In contrast, Sox1 and Sox3 become more prominent during neural plate commitment, marking the transition from multipotent progenitors to committed neural precursors. expression surges in the forming , where it reinforces activity and drives the expression of early neural markers, contributing to the stabilization of the neuroectodermal domain. Sox3, similarly, activates downstream neural genes like itself and geminin, while indirectly suppressing epidermal genes to delineate the neural territory. These SoxB1 factors exhibit functional redundancy in the , as evidenced by studies showing that combined loss of Sox1, , and Sox3 disrupts formation more severely than individual knockouts. Proneural basic helix-loop-helix (bHLH) transcription factors, particularly Neurogenin1 (Neurog1) and Neurogenin2 (Neurog2), initiate within subsets of neuroectodermal cells by promoting neuronal . Expressed in proneural clusters of the neuroectoderm, Neurog1 and Neurog2 activate pan-neuronal genes and drive progenitors out of the , specifying neuronal identities in the developing . Their activity is restricted to neuroectodermal domains, where they integrate with SoxB1 factors to balance proliferation and . Dorsal-ventral patterning of the neural tube is regulated by transcription factors such as and Irx3, which establish progenitor domains along this axis. , expressed in intermediate to regions, promotes the specification of progenitor domains by activating proneural genes like Neurog1 and Neurog2, while restricting ventral markers such as Nkx2.2. In neuroectoderm, acts as a fate determinant, uniformly marking early neural cells. Conversely, Irx3, expressed in intermediate domains, limits the ventralizing influence of signals like Sonic hedgehog by repressing motor neuron markers, helping to segregate progenitor identities along the D-V axis. Together, these factors interact to refine the neuroectodermal architecture into spatially organized domains.

Signaling Pathways Involved

The development of neuroectoderm is orchestrated by a series of extracellular signaling pathways that guide cell fate decisions from initial induction through patterning and differentiation. These pathways, including , Wnt, FGF, , and Shh, operate in a spatially and temporally regulated manner to establish neural identity, anterior-posterior (A-P) and dorsoventral (D-V) axes, and cellular diversity within the . Inhibition or activation of these cascades by secreted antagonists or agonists ensures precise patterning, with disruptions leading to developmental anomalies. The signaling pathway plays a pivotal role in the initial commitment of to a neural fate during . In vertebrates, BMP ligands, such as BMP4, promote epidermal in the presumptive ; however, their inhibition by secreted antagonists like Chordin and Noggin, produced by the Spemann-Mangold organizer, blocks BMP receptor binding and Smad1/5/8 activation, thereby preventing epidermal fate and favoring neuroectoderm specification. This antagonism is essential in early stages, as demonstrated in and chick embryos, where exogenous BMP application suppresses neural markers while Noggin overexpression induces ectopic neural tissue. Along the A-P axis, Wnt and FGF signaling pathways cooperate to posteriorize the neuroectoderm, directing the formation of and identities while restricting anterior fates. Wnt/β-catenin signaling, emanating from posterior , establishes a that is high posteriorly and low anteriorly, activating posterior and repressing anterior determinants like Otx2; in and models, Wnt inhibition expands anterior neural structures, whereas activation shifts fates caudally. Similarly, FGF signaling from the and posterior tissues reinforces this posteriorization through ERK/MAPK activation, promoting genes such as Cdx and Hox in a dose-dependent manner, as evidenced by FGF8 knockdown in chicks resulting in anteriorized neural plates. These pathways often intersect, with Wnt inducing FGF expression to amplify the posterior model. Notch signaling mediates within proneural clusters of the neuroectoderm, ensuring stochastic selection of neural progenitors amid a field of equivalent cells during early . In and neuroectoderm, Delta-like ligands on proneural cells activate receptors on neighbors, leading to cleavage and release of the NICD intracellular domain, which represses proneural genes like achaete-scute complex (ASC) via Enhancer of split repressors; this feedback amplifies differences, resulting in spaced neural precursors surrounded by epidermal or glial cells. Genetic studies in and mice show that mutants exhibit overproduction of neurons due to failed inhibition, highlighting its role in balancing proliferation and . Post-formation of the , the Shh signaling pathway drives ventralization, specifying floor plate and identities along the D-V axis through a ventral-to-dorsal gradient. Secreted from the and induced floor plate, Shh binds Patched receptors, relieving inhibition of and activating transcription factors (primarily Gli2/3 activators ventrally); concentration thresholds pattern progenitors, with high Shh inducing floor plate (via Nkx2.2) and intermediate levels specifying (via Olig2). In mouse knockouts, Shh absence abolishes ventral domains, while ectopic expression ventralizes dorsal regions, underscoring its morphogen-like function. These pathways converge on downstream transcription factors such as and to execute neural-specific gene programs.

Clinical and Research Significance

Associated Disorders

Defects in neuroectoderm development during embryogenesis can lead to a range of congenital disorders, primarily affecting the and derivatives. These conditions arise from disruptions in critical processes such as closure and , resulting in structural malformations with significant clinical implications. Neural tube defects (NTDs) represent one of the most common classes of neuroectoderm-related disorders, occurring due to incomplete closure of the during early development. results from failure of the posterior neuropore to close, leading to exposure or tethering of the and associated meninges, while stems from anterior neuropore defects, causing absence of the and cerebral hemispheres. The global incidence of NTDs is approximately 1 in 1,000 live births, with accounting for a substantial portion in regions without widespread folate fortification. A key environmental risk factor is maternal during periconception, which increases NTD risk by impairing and in neuroectodermal cells; supplementation with folic acid has been shown to prevent up to 70% of cases. Holoprosencephaly (HPE) is another severe disorder linked to neuroectoderm anomalies, characterized by incomplete division and ventral midline facial defects such as or . This condition arises from disruptions in Sonic hedgehog (Shh) signaling within the ventral neuroectoderm, which is essential for patterning the midline structures during . Genetic affecting Shh pathway components, including SHH itself, account for a significant proportion of HPE cases, highlighting the pathway's role in maintaining neuroectodermal integrity. Disorders originating from neural crest cells, a neuroectodermal derivative, further illustrate the broad impact of these developmental failures. Hirschsprung disease involves aganglionosis of the distal bowel due to incomplete migration and differentiation of enteric neural crest cells, resulting in functional obstruction from absent neurons in the myenteric and submucosal plexuses. DiGeorge syndrome (22q11.2 deletion syndrome) encompasses conotruncal heart defects, thymic hypoplasia, hypocalcemia, and craniofacial anomalies, stemming from impaired migration or survival of pharyngeal neural crest cells that contribute to outflow tract septation and branchial arch structures. The of these neuroectoderm-associated disorders often involves a interplay of genetic and environmental factors. For NTDs, polymorphisms in the MTHFR gene, such as the C677T variant, elevate risk by reducing metabolism efficiency, with homozygous carriers facing up to a fourfold increase in susceptibility. Environmental contributors, including , , and exposure, compound these genetic predispositions, underscoring the multifactorial nature of neuroectodermal vulnerabilities.

Current Research Directions

Recent research in neuroectoderm biology has leveraged induced pluripotent stem cells (iPSCs) to generate neuroectoderm models for studying defects (NTDs). Patient-specific iPSC-derived neuroepithelial models demonstrate reproducible differentiation into neuroectoderm-like structures, revealing deficiencies in cell shape regulation and differentiation in NTD cases, such as and . For instance, iPSC-based organoids (NTOs) have identified mutations in genes like NUAK2, which impair Hippo-YAP signaling and lead to failed closure, providing platforms for high-throughput drug screening. These models, developed since 2020, enable ethical, human-relevant simulations of neuroectoderm formation, surpassing limitations of animal models in capturing species-specific timing. Single-cell RNA sequencing (scRNA-seq) has advanced the mapping of neuroectoderm heterogeneity and differentiation trajectories in human embryos and organoids. Integrated scRNA-seq atlases of human pre-gastrulation embryos (over 3,300 cells) delineate neuroectoderm lineage emergence from epiblast cells, identifying dynamic networks like NANOG downregulation during specification. In hESC-derived models, scRNA-seq at days 26 and 54 of neuronal differentiation highlights 539 differentially expressed genes, including NEUROD1 and TBR1, enriched in pathways and revealing regulatory footprints for early neuroectoderm commitment. Post-2018 organoid advances use scRNA-seq to profile cell clusters in embryo-like structures, confirming neuroectoderm trajectories akin to Carnegie stage 12 . These techniques uncover subtype-specific markers, aiding in dissecting neuroectoderm diversification. Comparative studies across phyla illuminate evolutionary of neural in neuroectoderm. Gene expression mapping reveals shared neuroectoderm anlagen between insects (e.g., ) and s, with the insect head boundary homologous to the vertebrate mid-hindbrain organizer, supporting conserved BMP/SHH signaling for patterning. In non-s like , recent analyses of highlight gene regulatory networks for neural fate acquisition, paralleling vertebrate neuroectoderm via default mechanisms without overt organizers. These insights, from 2020 onward, underscore modular , informing human neuroectoderm models through cross-species validation. Therapeutic research targets neuroectoderm regeneration using gene editing and bioengineering. CRISPR-Cas9 editing of , a key neuroectoderm , has shown its essential role in repair; knockouts in axolotls impair proliferation post-injury, while targeted upregulation promotes maintenance for regeneration. In the , bioengineered neural tubes from hPSCs via the method fuse aggregates to impose BMP/SHH gradients, yielding dorsoventral-patterned organoids with 13 subtypes, mimicking for transplantable repair constructs. NTOs further model neuroectoderm timing, with versions exhibiting 2.5–3.7-fold slower maturation than murine, offering platforms to test CRISPR-enhanced grafts for NTD correction and injury recovery.

References

  1. [1]
    Embryology, Ectoderm - StatPearls - NCBI Bookshelf - NIH
    The neuro-ectoderm will form the neural tube and the neural crest. The neural tube will form the central nervous system (CNS: brain and spinal cord) and control ...Missing: neuroectoderm | Show results with:neuroectoderm
  2. [2]
    Neuroanatomy, Neural Tube Development and Stages - NCBI - NIH
    Notochord induces the neuroectoderm to become the neural plate by increasing the fibroblast growth factor(FGF) and inhibiting the bone morphogenic protein (BMP ...Structure And Function · Embryology · Clinical SignificanceMissing: definition | Show results with:definition
  3. [3]
    Neuroectoderm - an overview | ScienceDirect Topics
    Neuroectoderm refers to the embryonic tissue that gives rise to the central nervous system and neural components, including glial cells and brain tumors, which ...Nervous System... · Nervous Systems · The Neuron
  4. [4]
    14.1 Embryonic Development – Anatomy & Physiology 2e
    A specialized region of this layer, the neuroectoderm, becomes a groove that folds in and becomes the neural tube beneath the dorsal surface of the embryo. The ...The Neural Tube · Relating Embryonic... · Disorders Of The Nervous...
  5. [5]
    The Initial Formation of the Nervous System: Gastrulation and ... - NCBI
    The ectoderm that lies immediately above the notochord is called the neuroectoderm, and gives rise to the entire nervous system. In addition to specifying the ...
  6. [6]
    Embryology, Gastrulation - StatPearls - NCBI Bookshelf - NIH
    Apr 23, 2023 · The ectoderm is the outer layer of the embryo, which gives rise to the external ectoderm (epidermis, hair, nails) and the neuroectoderm (neural ...
  7. [7]
    [PDF] Pseudostratified epithelia – cell biology, diversity and roles in organ ...
    Apr 28, 2017 · While cells in PSE, like other epithelial cells, feature apico-basal polarity, they generally are more elongated and their nuclei are more ...Missing: ratio Sox2
  8. [8]
    Stepwise Maturation of Apicobasal Polarity of the Neuroepithelium Is ...
    Here, we show that N-Cad/ZO-1 complex-initiated apicobasal polarity is stabilized by the late-onsetting Lin7c/Nok complex after the extensive morphogenetic cell ...Missing: pseudostratified Sox2
  9. [9]
    SOX2 Functions to Maintain Neural Progenitor Identity - ScienceDirect
    We show here that constitutive expression of SOX2 inhibits neuronal differentiation and results in the maintenance of progenitor characteristics.
  10. [10]
    Immunohistochemical Markers of Neural Progenitor Cells in the ...
    The characteristic pseudostratification is mainly due to ... The ventricular zone was intensely immunostained by WT1, Sox2, Nestin and Vimentin.
  11. [11]
    Directed Neural Differentiation of Human Embryonic Stem Cells via ...
    The use of primitive neuroectoderm in this context is intended to define the first cells possessing morphological characteristics and expressing known ...Missing: definition | Show results with:definition
  12. [12]
    The revolutionary developmental biology of Wilhelm His, Sr - PMC
    Feb 22, 2022 · Wilhelm His, Sr. (1831–1904) was the first scientist to study embryos using paraffin histology, serial sectioning and three‐dimensional modelling.
  13. [13]
    Ectoderm | Embryo Project Encyclopedia
    Dec 2, 2013 · The second is the neuroectoderm, which forms the nervous system of the embryo. The neuroectoderm further divides into the neural tube, which ...Missing: definition | Show results with:definition
  14. [14]
    NEUROECTODERM Definition & Meaning | Merriam-Webster Medical
    The meaning of NEUROECTODERM is embryonic ectoderm that gives rise to nervous tissue.
  15. [15]
  16. [16]
    Neural induction and early patterning in vertebrates - PMC
    Jan 7, 2020 · Neural induction is triggered by signals from an early embryo region during gastrulation, where the dorsal ectoderm forms the neural plate.
  17. [17]
    Neural induction: old problem, new findings, yet more questions
    May 1, 2005 · This review summarizes neural induction events in different species and highlights some unanswered questions about this important developmental process.
  18. [18]
  19. [19]
    Neural Induction - FGF Signalling in Vertebrate Development - NCBI
    Chordin and Noggin are both secreted BMP antagonists that bind BMP ligands extracellularly and prevent their association with receptors, thereby blocking BMP ...
  20. [20]
    Proposal of a model of mammalian neural induction - PMC - NIH
    In his seminal work with Hilde Mangold, published in 1924, they proposed the organizer model. Spemann and Mangold found that the dorsal blastopore lip of the ...Missing: paper | Show results with:paper<|separator|>
  21. [21]
  22. [22]
    Formation of the Neural Tube - Developmental Biology - NCBI - NIH
    Shortly after the neural plate has formed, its edges thicken and move upward to form the neural folds, while a U-shaped neural groove appears in the center of ...Missing: 18 pseudostratified epithelium
  23. [23]
    Neural Plate - an overview | ScienceDirect Topics
    The neural plate is defined as a thickening of ectodermal cells on the dorsal surface of the embryo, which initiates the process of neurulation and ...Introduction · Embryological Development... · Molecular and Cellular...
  24. [24]
    Embryology, Neural Tube - StatPearls - NCBI Bookshelf - NIH
    This process is called primary neurulation, and it begins with an open neural plate, then ends with the neural plate bending in specific, distinct steps.[1] ...Missing: 18 pseudostratified
  25. [25]
    Neural tube closure: cellular, molecular and biomechanical ...
    Feb 15, 2017 · The process of neural tube closure is complex and involves cellular events such as convergent extension, apical constriction and interkinetic nuclear migration.
  26. [26]
    Planar Cell Polarity Links Axes of Spatial Dynamics in Neural-Tube ...
    May 25, 2012 · The median hinge point. (MHP) occurs along the midline of the neural plate where neuro- epithelial cells exhibit a wedge-like shape due to their ...
  27. [27]
    Sonic hedgehog signaling directs patterned cell remodeling during ...
    Oct 26, 2020 · In the amphibian neural plate, apical constriction is required to form the median and dorsolateral hinge points, two localized tissue ...
  28. [28]
    Patterning the Vertebrate Neural Plate by Wnt Signaling - NCBI - NIH
    Proposed candidates include FGF and retinoic acid (RA) which can posteriorize anterior neural tissue with virtually no neural inducing activity on their own.-.
  29. [29]
    Wnt signaling regulates neural plate patterning in distinct temporal ...
    Jun 15, 2020 · We show that Wnt-mediated neural patterning in zebrafish can be divided into three phases: (I) a primary AP patterning phase, which occurs during gastrulation.Wnt Signaling Regulates... · 3. Results · 3.2. Timed Wnt...
  30. [30]
    Anterior-Posterior Patterning Precedes Regional Nervous System ...
    Nov 1, 2018 · Later, Nieuwkoop proposed a two-step “activation-transformation” model by which anterior neural plate is induced (activation) and then ...
  31. [31]
    Sequentially acting Sox transcription factors in neural lineage ...
    All three SoxB1 proteins (Sox1, Sox2, and Sox3) are expressed in most neural precursors in both the developing and adult CNS, and studies conducted in chick and ...
  32. [32]
    N-cadherin stabilises neural identity by dampening anti-neural signals
    When exogenous N-cadherin was induced in these cells during neural differentiation (Fig. 2G), an increase in expression of the early neural markers Sox1, Pax6 ...
  33. [33]
    Sox3 regulates both neural fate and differentiation in the zebrafish ...
    Aug 1, 2008 · Sox3 over-expression causes neural plate markers ncad and sox2 to expand locally (G, M, 29/47 embryos, H, N, 32/39). Markers of the neural ...Missing: upregulation | Show results with:upregulation
  34. [34]
    Hinge point emergence in mammalian spinal neurulation - PMC - NIH
    Spinal neural tube folding shows a transition from medial to dorsolateral neural plate bending sites (hinge points) along the embryo's body axis.
  35. [35]
    Neural tube closure in humans initiates at multiple sites - PubMed
    NT closure in human embryos initiates at multiple sites but that the mode of NT closure in humans is different from that in many other animal species.Missing: sequence day 22 bidirectional
  36. [36]
    The cellular dynamics of neural tube formation - PMC
    Feb 16, 2023 · Although the details vary between species, the general process of primary neurulation involves remodelling of the neural plate by convergent ...
  37. [37]
    Tissue Architecture of the Central Nervous System - NCBI - NIH
    A section of a 5-week human neural tube (left) reveals three zones: ventricular (ependymal), intermediate (mantle), and marginal. In the spinal cord and medulla ...Tissue Architecture Of The... · Spinal Chord And Medulla... · Cerebellar Organization<|separator|>
  38. [38]
    Differential requirements of BMP and Wnt signalling during ... - NIH
    We show that the first inductive step requires Wnt activation and BMP inhibition, whereas the later maintenance step requires activation of both pathways.
  39. [39]
    WNT/β-catenin signaling mediates human neural crest induction via ...
    Feb 1, 2016 · We demonstrate a crucial role for WNT/β-catenin signaling in launching NC development, while blocking placodal and surface ectoderm fates.
  40. [40]
    Specification and formation of the neural crest - PubMed Central - NIH
    A human model of neural crest based on embryonic stem cells induced through WNT signaling has also demonstrated that BMP is required for neural crest induction, ...
  41. [41]
    Snail2 is an essential mediator of Twist1-induced epithelial ... - NIH
    Our results show that Twist1 needs to induce Snail2 to suppress the epithelial branch of the EMT program and that Twist1 and Snail2 act together to promote EMT ...
  42. [42]
    Snail2 controls mesodermal BMP/Wnt induction of neural crest - PMC
    It provides a classic example of an epithelial-mesenchymal transition (EMT) ... Snail precedes slug in the genetic cascade required for the specification ...
  43. [43]
    [PDF] New Insights into the Regulation of Epithelial-Mesenchymal ...
    The expression of Snail, Slug, Twist, and SIP1 is increased during. EMT in neural crest cells (Casas et al., 2011; Nieto, 2002), and Twist1 and. Slug are ...
  44. [44]
    The Neural Crest - Developmental Biology - NCBI Bookshelf
    The cardiac neural crest cells can develop into melanocytes, neurons, cartilage, and connective tissue (of the third, fourth, and sixth pharyngeal arches). In ...
  45. [45]
    Neuroanatomy, Neural Crest - StatPearls - NCBI Bookshelf
    The neural crest is a collection of multipotent stem cells located at the side of the neural tube proximal to the epidermal layer after neurulation.
  46. [46]
    Neural Crest Migration: Patterns, Phases and Signals - PMC
    In the trunk, neural crest migration is patterned by the somites. Trunk neural crest cells, which initially migrate between the somites, are later repelled ...
  47. [47]
    Molecular analysis of neural crest migration - PMC - NIH
    NC cells that are specified as neurons and glial cells only migrate ventrally and are prevented from migrating dorsolaterally into the skin, whereas NC cells ...
  48. [48]
    Transcription factors and neural stem cell self-renewal, growth and ...
    Transcription factors (TFs) play prominent roles in developmental processes and have provided excellent tools to understand stem cell-lineage specification. The ...
  49. [49]
    Sox2, a key factor in the regulation of pluripotency and neural ...
    Sox2 is one of the key transcription factors that play an essential role in maintaining pluripotency of stem cells.
  50. [50]
    Early neural specification of stem cells is mediated by a set of SOX2 ...
    Apr 4, 2024 · In summary, we characterized the role of Sox2 in driving NE specification via selective activation of SONAEs, a set of SOX2-dependent neural ...Missing: Sox3 | Show results with:Sox3
  51. [51]
    Interaction of Sox1, Sox2, Sox3 and Oct4 during primary neurogenesis
    Sox1, Sox2 and Sox3 have overlapping and distinct roles in neural specification and differentiation. We demonstrated that overexpression of Sox2 or Sox3 ...
  52. [52]
    Xenopus Sox3 activates sox2 and geminin and indirectly represses ...
    Sox3 functions as an activator to induce expression of the early neural genes, sox2 and geminin in the absence of protein synthesis and to indirectly inhibit ...3.2. Sox3 Activates Geminin... · 3.4. Sox3 Inhibits Epidermal... · 3.6. Sox3 Inhibits Neural...
  53. [53]
    SOX after SOX: SOXession regulates neurogenesis - PMC
    Upon specification to NPCs, Sox2 may be replaced on neural enhancers by other SoxB1 proteins, such as Sox3. Neural enhancers bound by Sox2 or Sox3 (Sox2/3) ...
  54. [54]
    The Sox Family of Transcription Factors: Versatile Regulators of ...
    In the absence of competing factors, Sox2 drives axial stem cells towards a neural plate fate. However, in the presence of Tbx6, a regulator of presomitic ...
  55. [55]
    Regulatory pathways linking progenitor patterning, cell fates and ...
    These proneural genes play an essential role in the decision of progenitors to exit the cell cycle and in specifying the pan-neuronal characteristics of neurons ...
  56. [56]
    Sequential phases of cortical specification involve Neurogenin ...
    Given the central role that Pax6 plays in cortical development, it is of particular interest that Pax6 directly activates Ngn1 and Ngn2, two highly related ...
  57. [57]
    Pax6 is a human neuroectoderm cell fate determinant - PMC - NIH
    The transcriptional regulation of neuroectoderm (NE) specification is unknown. Here we show that Pax6 is uniformly expressed in early NE cells of human fetuses ...
  58. [58]
    Irx3 and Pax6 establish differential competence for Shh-mediated ...
    Sep 24, 2013 · Here we demonstrate that the differential expression of two transcription factors, Irx3 and Pax6, spatially limits the area of competence for these inductions.
  59. [59]
    Establishing and Interpreting Graded Sonic Hedgehog Signaling ...
    The secreted protein Sonic Hedgehog (SHH) acts in graded fashion to pattern the dorsal–ventral axis of the vertebrate neural tube. This is a dynamic process ...
  60. [60]
    Noggin-mediated antagonism of BMP signaling is required for ...
    Noggin is not essential for neural induction but is required for normal growth and patterning of the neural tube and somite. Thus, inhibition of endogenous BMP ...
  61. [61]
    BMP inhibition initiates neural induction via FGF signaling and Zic ...
    Third, the BMP antagonists chordin, noggin, and cerberus are still expressed in FGF4 morphants that lack neural gene expression. Finally, we showed that FGF ...
  62. [62]
    Self-patterning of rostral-caudal neuroectoderm requires dual role of ...
    Nov 7, 2017 · The neuroectoderm is patterned along a rostral-caudal axis in response to localized factors in the embryo, but exactly how these factors act ...Missing: definition | Show results with:definition
  63. [63]
    Integration of Canonical and Noncanonical Wnt Signaling Pathways ...
    Jan 15, 2013 · Patterning the neuroectoderm along the anterior–posterior (AP) axis is a critical event in the early development of deuterostome embryos.
  64. [64]
    Correct anteroposterior patterning of the zebrafish neurectoderm in ...
    May 16, 2011 · Wnt, Nodal, and FGF signaling pathways are all known to be involved in the posteriorization of neural tissue [45,54-56], an effect they ...
  65. [65]
    Mathematical modeling of Notch dynamics in Drosophila neural ...
    A typical feature of Notch signalling is 'lateral inhibition', whereby two neighbouring cells of equivalent state of differentiation acquire different cell ...
  66. [66]
    Notch signaling and proneural genes work together to control the ...
    The other function of Notch is lateral inhibition, whereby a ligand-expressing cell inhibits the expression of the ligand in the neighboring cells, therefore ...
  67. [67]
    Notch Signalling Under Maternal-to-Zygotic Transition - PMC - NIH
    (b) Notch signaling is involved in lateral inhibition and its failure results in a neurogenic phenotype. In Drosophila neuroectoderm, a neuroblast (orange) is ...
  68. [68]
    Ventral neural patterning in the absence of a Shh activity gradient ...
    Vertebrate spinal cord development requires Sonic Hedgehog (Shh) signaling from the floorplate and notochord, where it is thought to act in concentration ...
  69. [69]
    The Many Hats of Sonic Hedgehog Signaling in Nervous System ...
    Early studies demonstrated that blocking Shh signaling impairs the generation of motor neurons through its action during two critical periods; one in which ...
  70. [70]
    Dorsal-ventral patterning of the spinal cord requires Gli3 ...
    However, even in the absence of graded Shh signaling, Gli3 is required for the dorsal-ventral patterning of the intermediate neural tube. Together these data ...
  71. [71]
    Human Neural Tube Defects: Developmental Biology, Epidemiology ...
    Primary neurulation generates the entire neural tube rostral to the caudal neuropore. ... During this process, occurring during the third and fourth weeks of ...
  72. [72]
    Neural Tube Defects - CDC
    May 20, 2025 · The most common NTDs are spina bifida, anencephaly, and encephalocele. Spina bifida affects about 1,300 babies a year in the United States.1 ...
  73. [73]
    Folate fortification for spina bifida: preventing neural tube defects
    Oct 4, 2024 · These conditions are often linked to folic acid deficiency during early pregnancy, a modifiable risk factor.
  74. [74]
    Folic acid and neural tube defects: Discovery, debate and the need ...
    Jun 23, 2022 · The search for a cause was elusive, but in 1991 it was shown that about 8 out of 10 cases are due to a lack of vitamin B9 (folate) and are therefore ...
  75. [75]
    Mutations in the human Sonic Hedgehog gene cause ... - Nature
    Nov 1, 1996 · Disruption of Sonic Hedgehog Signaling Accelerates Age-Related Neurogenesis Decline and Abolishes Stroke-Induced Neurogenesis and Leads to ...
  76. [76]
    Temporal perturbations in sonic hedgehog signaling elicit the ...
    Studies conducted in mammals and birds show that the severity of HPE defects correlates with the stage in which interruption in Shh signaling occurs (9, 28).
  77. [77]
    Loss of Tgif Function Causes Holoprosencephaly by Disrupting the ...
    We show that mutations in Tgif1 and Tgif2 in mice cause HPE by disrupting the Sonic Hedgehog signaling pathway, further emphasizing the importance of this ...
  78. [78]
    The Developmental Etiology and Pathogenesis of Hirschsprung ...
    Extensive research has identified a number of key genes that regulate neural crest cell development in the pathogenesis of Hirschsprung disease including RET, ...Pathogenesis · Gdnf/ret · Abbreviations
  79. [79]
    Hirschsprung's Disease: What It Is, Symptoms & Treatment
    What causes Hirschsprung's disease? During fetal development, neural crest cells typically grow from the top of the small intestine through the large ...Overview · Symptoms And Causes · Management And Treatment
  80. [80]
    The role of neural crest during cardiac development in a mouse ...
    Neural crest cells play a major role in the development of the pharyngeal arches, and defects in these cells are likely responsible for the syndrome. Most ...
  81. [81]
    Genetic Epidemiology of Neural Tube Defects - PMC - NIH
    It has been estimated that 60–70% of neural tube defects (NTDs) have a genetic component, but few causative genes have been identified.Missing: incidence | Show results with:incidence
  82. [82]
    Methylenetetrahydrofolate reductase mutations, a genetic cause for ...
    The best-characterized MTHFR genetic mutation 677C→T is associated with a 2–4 fold increased risk of NTD if patient is homozygous for this mutation.
  83. [83]
    Genetic Variants in the Folate Pathway and the Risk of Neural Tube ...
    Our meta-analysis strongly suggested a significant association of the variant MTHFR C677T and a suggestive association of RFC-1 A80G with increased risk of NTDs ...
  84. [84]
    Patient-specific iPSC models of neural tube defects identify ... - bioRxiv
    May 20, 2025 · We characterise a human iPSC-derived neuroepithelial morphogenesis model which is highly reproducible between lines.Missing: neuroectoderm 2020-2025
  85. [85]
    Progress in Modeling Neural Tube Development and Defects by ...
    Jun 26, 2022 · In this review, we focus on the recent research progress on neural tube organoids and discuss both their challenges and potential solutions.
  86. [86]
    A comprehensive human embryo reference tool using single-cell ...
    Nov 14, 2024 · To authenticate human embryo models, single-cell RNA sequencing has been utilized for unbiased transcriptional profiling. However, an organized ...Missing: neuroectoderm | Show results with:neuroectoderm
  87. [87]
    Single-cell RNA-seq data analysis reveals functionally relevant ...
    May 12, 2024 · By employing scRNA-seq on human embryonic stem cells, we aim to identify differentially expressed genes (DEGs) crucial for early-stage neuronal ...
  88. [88]
    Gene expression mapping of the neuroectoderm across phyla - eLife
    Sep 26, 2023 · In this review, we revisit the molecular mapping of the neuroectoderm of insects and vertebrates to reconsider homology hypotheses.
  89. [89]
    Neurogenesis in Caenorhabditis elegans - PMC - PubMed Central
    Aug 21, 2024 · Here, we review the gene regulatory mechanisms driving neurogenesis and the diversification of neuron classes and subclasses in C. elegans.Missing: 2020-2025 | Show results with:2020-2025
  90. [90]
    Editing SOX Genes by CRISPR-Cas: Current Insights and Future ...
    SOX2 plays a major role in both tumorigenesis and embryogenesis, especially during the development and differentiation of the neuroectodermal layer. Yang et al.
  91. [91]
  92. [92]
    Neural Tube Organoids: A Novel System to Study Developmental ...
    Sep 4, 2024 · Neural tube organoids (NTOs) are derived from stem cells, used to study developmental timing, especially neural tube defects, and are made from ...Neural Tube Closure In Human... · Neuronal Maturation In Human... · Glial Cell Formation In...Missing: 2020-2025 | Show results with:2020-2025