Fact-checked by Grok 2 weeks ago

Neuroepithelial cell

Neuroepithelial cells are the primary neural cells that constitute the pseudostratified lining the during early embryonic development, originating from the and serving as multipotent stem cells capable of self-renewal and differentiation into all neurons and glial cells of the . These cells form through primary , where the folds and fuses to create the , establishing the foundational structure of the and . Characterized by their apical-basal polarity, neuroepithelial cells maintain tight adherens junctions at the apical surface facing the ventricular and extend processes to the , enabling coordinated behaviors such as interkinetic nuclear migration, where nuclei move along the apicobasal axis during the to facilitate at the ventricular surface. Early in development, they primarily undergo symmetric proliferative divisions to expand the progenitor pool, ensuring sufficient cells for subsequent . As development progresses, their division modes shift to asymmetric neurogenic divisions, producing one and one postmitotic , or symmetric neurogenic divisions yielding two neurons, thereby initiating the generation of diverse neuronal subtypes in a spatiotemporal manner. Neuroepithelial cells transition into radial glial cells around the onset of , retaining many of their progenitor properties while acquiring elongated radial processes that guide neuronal and provide scaffolds for cortical layering. This transformation is marked by changes in , such as upregulation of glial markers like GFAP, and is crucial for the continued production of neurons and later across regions. Disruptions in neuroepithelial cell , division, or can lead to defects, highlighting their essential role in normal CNS formation.

Definition and Characteristics

Morphology and Distribution

Neuroepithelial cells form a that constitutes the wall of the developing . These cells exhibit pronounced apical-basal polarity, with short apical processes extending to contact the ventricular surface and long basal processes reaching the pial surface, thereby spanning the full thickness of the tube wall. At their apical ends, the cells are interconnected by adherens junctions, which maintain the structural integrity and cohesion of the epithelial sheet. These cells exhibit an elongated . During the cell cycle, neuroepithelial cells undergo interkinetic nuclear migration, a process in which nuclei migrate apically toward the ventricular surface during the and , and basally during the and G1 phases of . This dynamic movement contributes to the pseudostratified appearance of the , as nuclei are positioned at varying depths across the layer. Neuroepithelial cells are initially distributed uniformly, lining the entire lumen of the neural tube from its earliest formation. In mice, this lining is established around embryonic day 8.5 (E8.5), corresponding to approximately the third week of human gestation, when the neural plate begins to fold and the tube closes. This uniform arrangement persists along the rostrocaudal axis of the neural tube during primary neurulation.

Molecular Markers and Identification

Neuroepithelial cells, as neural stem cells during early embryonic , are characterized by the expression of specific molecular markers that underscore their and multipotency. Nestin, an protein, serves as a prominent cytoskeletal marker for these cells, reflecting their undifferentiated state and capacity for self-renewal. Similarly, the Sox2 is essential for maintaining pluripotency and neural fate specification, while Pax6, a paired box , regulates and is highly expressed in neuroepithelial progenitors to control their proliferation and differentiation potential. At the apical surface, where neuroepithelial cells form junctional complexes, markers such as (also known as ), a pentaspan , localize to protrusions and indicate properties within the ventricular zone. N-cadherin, a calcium-dependent , is integral to adherens junctions in the subapical region, supporting cell-cell interactions that preserve epithelial integrity and progenitor maintenance. In contrast, basal markers like , a key component of the in the , facilitate attachment and polarity, anchoring neuroepithelial cells to the underlying substrate during formation. The , particularly through upregulation of the Notch1 receptor, plays a critical role in sustaining the neuroepithelial progenitor state by inhibiting premature differentiation and promoting symmetric cell divisions. In their undifferentiated phase, these cells lack neuronal markers such as β-III tubulin, which emerges only upon commitment to a neuronal lineage, thereby distinguishing progenitors from post-mitotic neurons. Identification of neuroepithelial cells relies on techniques like , which detects these markers through antibody staining to visualize expression patterns in sections, and single-cell RNA sequencing, which profiles transcriptomic signatures to differentiate neuroepithelial cells from adjacent ectodermal populations based on clusters.

Embryonic Development

Neural Tube Formation

Neuroepithelial cells originate during the early stages of embryonic development through a process known as , where the is specified from the . This is primarily driven by signals from the underlying , which secretes Sonic hedgehog (Shh), a key that promotes ventral neural fate and inhibits non-neural ectodermal differentiation. Concurrently, the surface ectoderm contributes by inhibiting (BMP) signaling through secreted antagonists such as Noggin and Chordin, which are expressed in the and ; this BMP inhibition is essential for the ectoderm to adopt a neural identity rather than an epidermal one. These signaling pathways act in concert during , transforming the presumptive into the , from which neuroepithelial cells emerge as the initial population of neural progenitors. The formation of the neural tube, or primary neurulation, follows neural induction and involves dynamic morphological changes orchestrated by neuroepithelial cells. The neural plate initially thickens due to the apical constriction of neuroepithelial cells, leading to the elevation of the lateral edges as neural folds. These folds then converge toward the midline, where their tips fuse to enclose the central lumen, forming the neural tube that will develop into the central nervous system. This process relies on coordinated behaviors of neuroepithelial cells, including shape changes and cytoskeletal rearrangements, to achieve tube closure without gaps. In human gestation, primary neurulation occurs between days 18 and 28 post-fertilization, with the anterior neuropore closing around day 25 and the posterior neuropore by day 28. In mice, this timeline corresponds to embryonic days 8 to 10 (E8-E10). Failure of neural tube closure disrupts the integrity of the developing and can result in severe congenital defects. For instance, incomplete fusion at the cranial end leads to , characterized by the absence of major brain structures, while caudal failures cause , where the spinal cord remains exposed. These open neural tube defects arise from disruptions in the process and highlight the critical timing of closure, as the must seal completely by embryonic day 28 in humans to prevent such malformations. Post-closure, neuroepithelial cells initiate proliferation to expand the , a process detailed in subsequent developmental stages.

Proliferation Mechanisms

Neuroepithelial cells in the early primarily undergo symmetric cell divisions, which expand the pool by producing two identical daughter cells that remain within the neuroepithelium. This mode of division predominates during initial phases of (CNS) development, enabling rapid population growth to establish the foundational layers of the . As development progresses, neuroepithelial cells transition to asymmetric divisions, where one daughter cell retains identity for self-renewal while the other commits toward differentiation, balancing pool maintenance with the onset of . Proliferation is tightly regulated by signaling pathways, including Wnt/β-catenin, which promotes cell cycle progression and progenitor expansion in the developing forebrain. Similarly, fibroblast growth factor (FGF) signaling drives mitotic activity and sustains survival in anterior neuroepithelial regions through β-catenin-dependent mechanisms. The in these early-stage neuroepithelial cells typically lasts 8-12 hours, allowing for frequent divisions that support . A key feature of neuroepithelial proliferation is interkinetic nuclear migration (INM), where nuclei oscillate along the apical-basal axis in synchrony with the : they move basally during S-phase for and return apically for . This process ensures ordered division at the ventricular surface while maintaining epithelial integrity. High expression of cyclins, such as , facilitates the G1/S transition by activating cyclin-dependent kinases, thereby accelerating progression through the and enhancing proliferative capacity. In mice, the rate of neuroepithelial progenitors peaks around embryonic day 12 (E12), coinciding with intense expansion and the generation of millions of progenitors to fuel subsequent neuronal production. During , cells exhibit apical constriction to facilitate , as noted in morphological studies. This high proliferative phase underscores the dynamic balance required for CNS scaling.

Differentiation and Transition

Neuroepithelial cells undergo a critical transition during early , shifting from symmetric proliferative divisions to asymmetric divisions that generate one and one post-mitotic . This process is regulated by the activation of proneural basic helix-loop-helix (bHLH) transcription factors, such as Neurogenin1 (Neurog1) and Neurogenin2 (Neurog2), which promote neuronal fate commitment in the differentiating daughter cell while maintaining progenitor identity in the other. These asymmetric divisions typically begin around embryonic day 11 (E11) in mice, marking the onset of neurogenic phases where cell divisions balance self-renewal and . Concomitant with these fate decisions, neuroepithelial cells morphologically and molecularly transition into radial glial cells between approximately E11 and E13 in mice, characterized by the elongation of radial processes spanning the ventricular zone to the pial surface and the upregulation of glial markers such as . This shift transforms the pseudostratified neuroepithelium into a scaffold of bipolar radial glia, which not only serve as progenitors but also provide migratory guidance for newborn neurons toward the cortical plate. The transition reflects a broader change from multipotent neuroepithelial progenitors to glial-like radial glial cells capable of generating diverse neuronal and glial lineages. A key event in this differentiation pathway is the of intermediate progenitors from the ventricular zone, where these cells retract their apical processes and migrate basally to undergo further divisions away from the ventricular surface. This , often following asymmetric divisions of radial glia, amplifies neuronal output by allowing intermediate progenitors to produce additional neurons in the . Proliferative divisions in preceding stages enable the accumulation of neuroepithelial cells necessary for this transition.

Adult Neural Stem Cells

Persistence and Locations

In the adult mammalian , a subset of neural stem cells persists with neuroepithelial-like characteristics, primarily in the ventricular-subventricular zone (V-SVZ) and the subgranular zone (SGZ) of the . These cells, often identified as quiescent type B1 cells in the V-SVZ, derive from embryonic radial glia and retain features such as apico-basal polarity and primary cilia that contact the ventricular lumen, echoing the organization of embryonic neuroepithelium. Lineage tracing studies using Nestin-Cre systems have confirmed their persistence, demonstrating that these progenitors originate from slowly dividing embryonic populations around E13.5–E15.5 and contribute to the pool without significant turnover until activation. These adult neural progenitors express markers indicative of retained neuroepithelial traits, such as and Nestin alongside astrocytic proteins. These neuroepithelial-like cells are predominantly located lining the walls of the in the V-SVZ, where type B cells form a niche interfacing with the , and in the SGZ of the , where radial glia-like cells (RGLs) reside adjacent to the layer. Despite their astrocytic morphology—characterized by GFAP expression, intermediate filaments, and processes extending to blood vessels—they harbor stem-like potential, generating transit-amplifying progenitors upon activation. In the V-SVZ, type B cells occupy positions that allow direct ventricular contact via cilia, facilitating environmental sensing, while SGZ RGLs integrate into the hippocampal circuitry. Unlike their embryonic counterparts, which proliferate rapidly with cell cycles lasting hours (typically 8–24 hours), adult neuroepithelial-like cells exhibit markedly slower division rates, often entering quiescence for weeks to months to preserve the pool. This prolonged quiescence, with asymmetric divisions occurring every 1–3 months in type B cells, contrasts sharply with the symmetric proliferative expansions of embryonic neuroepithelium. Their persistence and activity are further modulated by aging, which reduces numbers through impaired lysosomal function and diminished signaling, as well as environmental factors like vascular niche interactions.

Role in Neurogenesis

Adult neuroepithelial-like stem cells in the (SVZ) serve as the primary source of new neurons in the adult brain, generating for the through a well-defined lineage progression. These s, classified as type B cells, exhibit characteristics reminiscent of embryonic neuroepithelial cells, including the ability to undergo asymmetric division to maintain the stem cell pool while producing transit-amplifying progenitors. Type B cells give rise to type C cells, which rapidly proliferate and differentiate into migrating neuroblasts known as type A cells. These neuroblasts form chains and travel tangentially via the rostral migratory stream () to the , where they integrate as , supporting olfactory discrimination and circuit plasticity. However, the occurrence of substantial in the SVZ is controversial, with some evidence suggesting it is minimal or absent after early adulthood. The process of SVZ neurogenesis is tightly regulated by the local niche environment, which includes ependymal cells lining the lateral ventricle and vascular structures. Ependymal cells provide structural support and secrete factors such as Shh and antagonists to modulate type quiescence and activation, preventing premature exhaustion of the reservoir. Vascular endothelial cells contribute trophic signals like VEGF and EGF, promoting and while anchoring stem cells to the niche; this vascular association is crucial for asymmetric divisions that mirror embryonic mechanisms but are adapted for adult . In adult humans, the rate of new addition from the SVZ is lower than in and remains debated due to species-specific differences in niche . In the hippocampal subgranular zone (SGZ), neuroepithelial-like type 1 stem cells drive neurogenesis, essential for hippocampal-dependent learning and memory formation. These stem cells proliferate to yield intermediate s that mature into granule cells, enhancing and pattern separation. The process is potently upregulated by (BDNF), which promotes progenitor survival and dendritic arborization, and by exercise-induced factors such as IGF-1 and VEGF, which boost through enhanced vascularization and reduced . In adult humans, approximately 700 new granule cells are incorporated into the of each daily in young adults, although the extent of this is subject to ongoing debate, with some studies questioning its persistence beyond . However, hippocampal declines with age, exhibiting roughly a 50% reduction by age 50 due to diminished progenitor and increased .

Repair and Regeneration

Following injury such as or , neuroepithelial-derived neural stem cells in the adult (SVZ) exhibit upregulated proliferation, generating neuroblasts that migrate toward damaged regions like the peri-infarct to support repair. In rodent models of focal , this response markedly increases SVZ , with chains of migrating neuroblasts extending to the , where they differentiate into medium spiny neurons—the primary neuronal type lost in such injuries—contributing to partial neuronal replacement. This process amplifies baseline SVZ , which under physiological conditions maintains limited neuronal turnover. Despite this potential, regeneration remains incomplete in mammals, as the majority of SVZ-derived cells post-injury adopt non-neuronal fates, such as undifferentiated precursors or , with fewer than 2% maturing into neurons even after weeks. Reactive often predominates, forming a that limits neuronal integration; for instance, shifts neural fate toward at the expense of astrogliogenesis, yet newborn neurons display abnormal morphology, including reduced dendritic spines, hindering functional incorporation. Studies in stroke models indicate that SVZ supports modest , with newborn neurons aiding synaptic and vascular , though overall neuronal accounts for only a small fraction of lost cells. Human postmortem evidence from cases of ischemic stroke corroborates this response, revealing elevated numbers of proliferating SVZ cells and neuroblasts, particularly in aged individuals, suggesting a conserved repair mechanism despite advanced age. Enrichment of growth factors like epidermal growth factor (EGF) and vascular endothelial growth factor (VEGF) further enhances this potential; VEGF, acting via VEGFR2 receptors, boosts SVZ proliferation by up to 2.5-fold and promotes neuroblast differentiation after traumatic brain injury, while EGF synergistically supports precursor expansion. These factors not only increase newborn neuron numbers but also correlate with reduced lesion volumes and improved motor outcomes in preclinical models.

Pathological Conditions

Neoplastic Disorders

Neoplastic disorders involving neuroepithelial cells primarily manifest as gliomas and glioneuronal tumors, arising from dysregulated proliferation of these progenitors or their derivatives. These tumors range from low-grade, indolent lesions to aggressive malignancies, often driven by genetic alterations that disrupt normal pathways. Key examples include dysembryoplastic neuroepithelial tumor (DNT) and oligodendroglial tumors, with high-grade forms like multiforme (GBM) linked to neuroepithelial origins through mechanisms. Dysembryoplastic neuroepithelial tumor (DNT) is a benign glioneuronal classified as WHO grade 1, typically arising in the of children and young adults. It is histologically characterized by a mucin-rich nodules containing oligodendrocyte-like cells and floating neurons in a specific glioneuronal element. DNT accounts for approximately 1.2% of neuroepithelial tumors in patients under 20 years old and is strongly associated with intractable , often presenting as the primary symptom leading to surgical intervention. Oligodendroglial tumors represent grade II-III diffuse gliomas that originate from neuroepithelial progenitors, defined molecularly by and complete 1p/19q codeletion as a hallmark genetic feature. These mutations, particularly IDH1 R132H, occur early in gliomagenesis and promote oncogenic transformation by altering cellular metabolism and epigenetic regulation. The 1p/19q codeletion confers sensitivity to and is prognostic for improved survival compared to non-codeleted gliomas. Glioblastoma multiforme (GBM), a high-grade , can trace its origins to neuroepithelial cells via of mature or glial progenitors into stem-like states. EGFR is a frequent alteration in these high-grade tumors, occurring in up to 57% of primary GBM cases and driving aggressive growth through enhanced receptor signaling. In contrast, BRAF mutations are identified in some low-grade neuroepithelial tumors, such as certain glioneuronal lesions, and are associated with MAPK pathway activation, offering potential therapeutic targets with BRAF inhibitors.

Developmental Malformations

Developmental malformations of neuroepithelial cells arise primarily from disruptions during embryogenesis, leading to structural anomalies in the without neoplastic transformation. These include fluid-filled cysts and defects in closure, resulting from impaired , , or of neuroepithelial progenitors. Such abnormalities often stem from genetic or environmental factors affecting key signaling pathways, manifesting as congenital conditions that may remain or cause neurological deficits depending on severity and location. Neuroepithelial cysts, also known as glioependymal cysts, represent benign, congenital lesions originating from incomplete separation or displacement of neuroepithelial tissue during formation. These fluid-filled structures are typically lined by cuboidal or columnar ependymal-like resting on a glial layer, distinguishing them from other cystic lesions. They are rare and often incidental findings, though larger cysts can exert leading to symptoms like or . Classification differentiates arachnoid cysts, which are extra-axial with meningeal (mesothelial) lining and located in subarachnoid spaces, from intraventricular neuroepithelial cysts, which are intra-axial with ependymal or glial components and arise within ventricular systems. Holoprosencephaly and neural tube defects exemplify malformations due to failed neuroepithelial cell or . results from disruptions in the Sonic hedgehog (Shh) signaling pathway, causing incomplete division and midline defects, as seen in Shh-null models where ventral neuroepithelial patterning fails. defects, such as , affect about 1 in 1,000 births globally and arise from inadequate closure of the caudal , often linked to that impairs and neuroepithelial cell division. In folate-deficient models, reduced hinders the morphogenetic movements required for tube closure, underscoring the role of nutritional factors in neuroepithelial function.

Research Advances

In Vitro Modeling

In vitro modeling of neuroepithelial cells has advanced significantly through three-dimensional (3D) culture systems derived from induced pluripotent stem cells (iPSCs), enabling the recapitulation of early stages. organoids, first established in 2013, self-organize from iPSCs into structures containing neuroepithelial layers that mimic radial organization and cortical layering observed . These models differentiate iPSCs into neuroectodermal progenitors, forming ventricular-like zones with apical-basal polarity characteristic of neuroepithelial cells. Assembloids extend this approach by fusing distinct organoids, such as and spheroids, to study inter-regional interactions and circuit formation. A key technique for generating neuroepithelial cells in these models involves directed protocols using dual SMAD inhibition, which blocks and TGF-β signaling to promote neural and the formation of neuroepithelial rosettes—cylindrical structures resembling the . This method, originally described in 2009, yields high-efficiency conversion of human iPSCs into PAX6-positive neuroepithelial progenitors within 10-14 days, facilitating scalable production for assembly. However, exhibit variability in size, typically ranging from 1-5 mm in , and cellular heterogeneity due to diffusion-limited supply, which can be mitigated by incorporating vascular networks through co-culture with endothelial cells or for . Single-cell transcriptomic analyses have provided detailed maps of neuroepithelial heterogeneity in these models. A 2021 atlas of early organoids highlighted diverse neuroepithelial subpopulations, including radial glia-like cells, mirroring embryonic stages from 5-9 post-conception weeks and revealing transcriptional gradients along the ventricular surface. More recent assembloid studies in 2025 have modeled forebrain-midbrain interactions, demonstrating neuron integration and synaptic connectivity relevant to pathogenesis. These systems have been instrumental in studying infectious impacts on neuroepithelial proliferation. Since 2016, brain organoids infected with have shown reduced progenitor proliferation, increased , and disrupted cortical layering, attributing microcephaly phenotypes to direct targeting of neuroepithelial cells by the virus. Ongoing refinements, such as vascularized assembloids, continue to enhance model fidelity for developmental and disease research.

Therapeutic Potential

Neuroepithelial-derived neural progenitor cells hold promise for stem cell transplantation therapies aimed at repairing spinal cord injuries (SCI). In rodent models of thoracic SCI, transplantation of human neuroepithelial stem cells has demonstrated the ability to form relay circuits that bridge injured areas, leading to significant functional recovery; for instance, grafted cells improved Basso Mouse Scale (BMS) scores from less than 1 (no movement) to 4 (occasional stepping) in mice with severe injuries, and up to 5 (consistent stepping) in milder cases, compared to controls that remained below 3. Similar engraftment of induced pluripotent stem cell (iPSC)-derived neuroepithelial progenitors in mouse contusion models has supported histological regeneration and motor function gains, with cells surviving, differentiating into neurons and glia, and integrating into host circuitry without tumor formation. Research also explores neuroepithelial cell modulation in psychiatric disorders, particularly , where enhancing in regions like the (SVZ) and could underlie therapeutic effects. Antidepressants such as selective serotonin reuptake inhibitors (SSRIs) have been shown to increase neural in these areas; in human postmortem studies of (MDD) patients treated with SSRIs, hippocampal progenitor numbers rose dramatically to approximately 19,800 per section from 1,100 in untreated cases, representing over a 17-fold increase. Animal models corroborate this, with chronic SSRI administration boosting SVZ and hippocampal by promoting survival and differentiation of neuroepithelial-like progenitors, potentially contributing to . In oncology, neuroepithelial markers expressed on glioblastoma (GBM) cells—such as Nestin and —serve as targets for emerging therapies. Phase I clinical trials as of 2025 investigate oncolytic viruses delivered via s to exploit these markers for selective tumor ; for example, the NSC-CRAd-S-pk7 agent, using a modified HB1.F3 line loaded with tumor-selective adenovirus, is being tested in recurrent high-grade gliomas, with the first patient enrolled in 2023 and ongoing multi-center enrollment evaluating safety across up to four intracerebral doses. Neural chimeras, formed by transplanting neuroepithelial s into host brains, further aid therapeutic development by modeling integration; in studies, these chimeras reveal progenitor , with donor cells migrating and forming functional connections in the host , informing strategies for brain repair. Despite these advances, challenges persist in translating neuroepithelial cell therapies. Immune rejection remains a hurdle, even with autologous iPSC-derived cells, due to potential from reprogramming or incomplete , necessitating or gene editing for better matching. Ethical concerns also arise, particularly around iPSC sourcing from human embryos or gametes, raising issues of , in , and the of tumorigenicity from residual undifferentiated cells, which could form teratomas post-transplantation. models provide a critical testing ground for optimizing these approaches before clinical use.

References

  1. [1]
    Cell biological regulation of division fate in vertebrate ...
    The developing nervous system derives from neuroepithelial progenitor cells that divide to generate all of the mature neuronal types.
  2. [2]
    Embryology, Neural Tube - StatPearls - NCBI Bookshelf - NIH
    May 1, 2023 · Primary neurulation is a complex process of formation of the neural tube from the neural plate and eventually undergoing neuro-epithelialization ...
  3. [3]
    Neuroepithelial Cell - an overview | ScienceDirect Topics
    Neuroepithelial cells (NECs) are defined as the most proliferative neural progenitor cells in the developing mouse neocortex, characterized by apical–basal ...
  4. [4]
    Neural Progenitor Cell Terminology - Frontiers
    Dec 5, 2018 · RG cells appear through differentiation of precursor cells known as “neuroepithelial cells” that initially form the walls of the neural tube.
  5. [5]
    Role of adherens junctions and apical-basal polarity of neural stem ...
    Adherens junctions (AJs) are organized as belt-like structures at the most-apical lateral plasma membrane of the apical processes. These junctional complexes ...
  6. [6]
    Dynamic behaviour of human neuroepithelial cells in the developing ...
    Jan 31, 2017 · As part of the neural plate and the early neural tube, NE cells contribute to the structure and shape of the developing nervous system. When the ...
  7. [7]
    Adherens Junctions: Guardians of Cortical Development - PMC
    Adherens junctions (AJs) play an obligate role in the maintenance of radial glial apicobasal polarity and neuroepithelial cohesion.
  8. [8]
    Shroom family proteins regulate γ-tubulin distribution and ...
    Apr 1, 2007 · At the onset of neurulation (stages 13-14), cells of the neural plate almost doubled their height, from approximately 12 μm to approximately 22 ...
  9. [9]
    Myosin II is required for interkinetic nuclear migration of neural ...
    Interkinetic nuclear migration (INM) is a hallmark of neuroepithelial and radial glial cells, the primary stem and progenitor cells of the vertebrate CNS (1–5).
  10. [10]
    Regulation of Neurogenesis by Interkinetic Nuclear Migration ...
    Sep 19, 2008 · The nuclei of progenitors move between the apical and basal surfaces of the neuroepithelium in phase with their cell cycle, a process termed interkinetic ...
  11. [11]
    Cellular basis of neuroepithelial bending during mouse spinal ... - NIH
    Bending of the neural plate at paired dorsolateral hinge points (DLHPs) is required for neural tube closure in the spinal region of the mouse embryo.
  12. [12]
    Human ES cell-derived neural rosettes reveal a functionally distinct ...
    Immunocytochemistry in NSCsFGF2/EGF for markers of NSCs (Nestin and Sox2; L), radial glia and neuroepithelial cells (Pax6, 3CB2, and Sox1; M), and anterior fate ...Missing: III | Show results with:III
  13. [13]
    Molecular Mechanisms Governing the Stem Cell's Fate in Brain ...
    These cells are probably capable of asymmetric division with generation of more differentiated cells expressing stem cell markers—CD133, CD44 and CD15.
  14. [14]
    N-cadherin-based adherens junction regulates the maintenance ...
    N-cadherin is responsible for the integrity of adherens junctions (AJs), which develop in the sub-apical region of NPCs in the neural tube and brain cortex.
  15. [15]
    Cytokinesis of neuroepithelial cells can divide their basal process ...
    Bifurcating basal processes of M-phase NE cells visualized by two independent fluorescent markers ... laminin α1, a basal lamina marker (Figure 1Ca). Remarkably, ...
  16. [16]
    Directed differentiation of human hindbrain neuroepithelial stem ...
    In their undifferentiated state, we confirmed that hbNES cells were not committed to the neural or glial lineage, indicated by absence of βIII-tubulin and GFAP ...
  17. [17]
    Single-cell atlas of early human brain development highlights ...
    To characterize the earliest stages of human brain development, we performed single-cell RNA-sequencing across regions of the developing human brain, including ...
  18. [18]
    The Molecular Basis of Neural Induction - Neuroscience - NCBI - NIH
    Neural induction involves signals from the primitive pit and notochord, using molecules like retinoic acid, FGF, TGF, and sonic hedgehog to modulate gene ...
  19. [19]
    Embryology, Gastrulation - StatPearls - NCBI Bookshelf - NIH
    Apr 23, 2023 · The notochord secretes Sonic Hedgehog, Chordin, and Noggin in a morphogenic gradient pattern where the highest concentration is near the ...
  20. [20]
    Patterning the Vertebrate Neural Plate by Wnt Signaling - NCBI - NIH
    The neural inducers noggin and chordin act by neutralizing a BMP signal in the ectoderm, and it is believed that neural crest forms in a region where ...
  21. [21]
    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.
  22. [22]
    Mechanics of morphogenesis in neural development: In vivo, in vitro ...
    Primary neurulation is the morphogenetic process during which the neural tube (NT) forms from the two-dimensional neural plate (NP). The mechanisms of ...
  23. [23]
    Neural Tube - an overview | ScienceDirect Topics
    Adjacent to the lumen of the neural tube is the germinal layer of proliferating neuroepithelial cells. This proliferative mitotic activity will ultimately ...Introduction · Embryological Development... · Neural Tube Differentiation...
  24. [24]
    Human Neural Tube Defects: Developmental Biology, Epidemiology ...
    According to Sakai, who wrote a comprehensive review of available data in mice and rat, rodent neural tube fusion occurs between day E8 to day E10 of gestation ...Missing: timeline | Show results with:timeline
  25. [25]
    Neural tube defects – disorders of neurulation and related ... - PMC
    Neural tube defects (NTDs) are severe congenital malformations affecting 1 in every 1000 pregnancies. 'Open' NTDs result from failure of primary neurulation.
  26. [26]
    Overview of Neural Tube Defects: Gene–Environment Interactions ...
    They are a result of the failure of the morphogenetic process of the neural tube closure. Two of the most common types of NTDs are spina bifida and anencephaly.
  27. [27]
    Species-specific pace of development is associated with ... - Science
    Sep 18, 2020 · Cell cycle duration in equivalent staged neural progenitors from mouse and human was 10.8 ± 8.3 hours compared with 28.4 ± 13.9 hours ...
  28. [28]
    Timing of CNS Cell Generation: Neuron & Glial Cell Production
    In the mouse or rat cerebral cortex, for example, neurogenesis commences around embryonic day 12 (E12), peaks around E15, and finishes around birth (5, 23).
  29. [29]
    Neural bHLH Genes Control the Neuronal versus Glial Fate ...
    We have addressed the role of the proneural bHLH genes Neurogenin2 (Ngn2) and Mash1 in the selection of neuronal and glial fates by neural stem cells.Bhlh Genes Are Expressed In... · Bhlh Genes Promote Neuronal... · Ngn2 Expression And Function...
  30. [30]
    Neural Stem Cells to Cerebral Cortex: Emerging Mechanisms ...
    Nov 9, 2016 · After their early expansion through symmetric divisions, APs begin to divide asymmetrically at ∼E11 in mice, such that one daughter cell retains ...
  31. [31]
    Gfap-Positive Radial Glial Cells Are an Essential Progenitor ...
    After producing the first neurons, neuroepithelial cells transition into radial glial cells, which have been suggested to act as the predominant NSC for the ...
  32. [32]
    Cerebral cortex development: From progenitors patterning to ...
    Mar 30, 2009 · The radial glia neurogenic progenitors are neuroepithelial cells that express glial cell markers and divide asymmetrically to give rise to a new ...
  33. [33]
    Radial Glial Cells: Defined and MajorIntermediates between ...
    Here, we review the evidence for the distinct “glial” nature of radial glial cells and contrast these cells with their progenitors, the neuroepithelial cells.Missing: neurogenin | Show results with:neurogenin
  34. [34]
    Neuronal Delamination and Outer Radial Glia Generation in ...
    Feb 5, 2021 · Neurogenic cell delamination, in which these neuronally differentiating cells retract their apical processes and depart from the apical surface, is the first ...
  35. [35]
    The Transition from Radial Glial to Intermediate Progenitor Cell Is ...
    Nov 18, 2009 · These results indicate that FGFs normally repress the radial glia to progenitor cell transition during corticogenesis.Loss Of Fgf Signaling Leads... · Fgf Signaling Inhibits Rgcs... · Fgf Signaling Acts...
  36. [36]
  37. [37]
  38. [38]
  39. [39]
  40. [40]
  41. [41]
    The Adult Ventricular–Subventricular Zone (V-SVZ) and Olfactory ...
    A large population of neural stem/precursor cells (NSCs) persists in the ventricular–subventricular zone (V-SVZ) located in the walls of the lateral brain ...
  42. [42]
    Adult SVZ Stem Cells Lie in a Vascular Niche - ScienceDirect.com
    Sep 11, 2008 · Neurogenesis Is Associated with SVZ Blood Vessels. Slowly dividing type B SVZ stem cells produce rapidly dividing transit-amplifying type C ...
  43. [43]
    Exercise Influence on Hippocampal Function: Possible Involvement ...
    Feb 13, 2017 · Research involving animals and humans supports the view that physical exercise, enhancing hippocampal neurogenesis and function, improves cognition, and ...
  44. [44]
    The BDNF-Interactive Model for Sustainable Hippocampal ...
    Dec 1, 2024 · In adult humans, arguably around 700 new neurons are added in each hippocampus per day until the tenth decade of life in a process known as ...
  45. [45]
    Rat forebrain neurogenesis and striatal neuron replacement after ...
    Brains examined 10-21 days after stroke showed markedly increased SVZ neurogenesis and chains of neuroblasts extending from the SVZ to the peri-infarct striatum ...
  46. [46]
    Subventricular zone cytogenesis provides trophic support for neural ...
    Oct 10, 2023 · Here, we characterize the identity of cells arising from the SVZ after stroke and uncover a mechanism through which they facilitate neural ...
  47. [47]
    Traumatic brain injury promotes neurogenesis at the cost of ... - Nature
    Jun 18, 2024 · We show that TBI modifies NSC fate to promote neurogenesis at the cost of astrogliogenesis and identify specific cell populations as possible targets.
  48. [48]
    Increased Generation of Neuronal Progenitors after Ischemic Injury ...
    Dec 13, 2006 · We analyzed cell proliferation and expression of neuronal progenitor antigens in the SVZ of adult human brains from postmortem cases of injured ...
  49. [49]
    Vascular endothelial growth factor (VEGF) stimulates neurogenesis ...
    Comparatively little is known regarding the possible role of VEGF in adult neurogenesis, although other growth factors, including epidermal growth factor (EGF) ...
  50. [50]
    Vascular endothelial growth factor increases neurogenesis after ...
    In conclusion, VEGF significantly augments neurogenesis and angiogenesis and reduces lesion volumes after TBI. These changes are associated with significant ...
  51. [51]
    Cell of Origin for Malignant Gliomas and Its Implication in ... - PMC
    One or more regenerative cell types in the brain—neural stem cells, astrocytes, or oligodendrocyte precursor cells—may serve as cell(s) of origin for gliomas.
  52. [52]
    Dysembryoplastic Neuroepithelial Tumors - PMC - PubMed Central
    Oct 23, 2015 · Dysembryoplastic neuroepithelial tumor (DNT) is a benign glioneuronal neoplasm that most commonly occurs in children and young adults.Missing: incidence | Show results with:incidence
  53. [53]
    Dysembryoplastic neuroepithelial tumor: A rare brain tumor not to be ...
    It is included in WHO 2007 classification as neuronal and mixed neuronal-glial tumor. Incidence among all neuroepithelial tumors is 1.2% in patients under 20 ...Missing: DNT characteristics
  54. [54]
    Dysembryoplastic Neuroepithelial Tumor: A Benign but Complex ...
    Jul 28, 2022 · Clinically, DNET is highly associated with intractable epilepsy in young children and adolescents. Therefore, the burden of the tumor comprises ...Missing: incidence | Show results with:incidence
  55. [55]
    Oligodendroglioma: pathology, molecular mechanisms and markers
    Further studies have shown that IDH mutation is an early event in gliomagenesis ('driver mutation'), and likely precedes the development of the 1p/19q ...
  56. [56]
    Oligodendroglioma - StatPearls - NCBI Bookshelf - NIH
    Apr 3, 2025 · The OG is defined by its IDH mutation and 1p/19q deletion status and several imaging features that suggest the presence of an OG as opposed to ...
  57. [57]
    IDH Mutations in Human Glioma - PMC - PubMed Central - NIH
    Although both TP53 mutations and 1p19q codeletions have been associated with improved prognosis, these mutations are mutually exclusive in gliomas, providing ...Missing: codeletion | Show results with:codeletion
  58. [58]
    Glioblastoma Cancer Stem-like Cells - PubMed Central - NIH
    We now have evidence that NSCs, glial progenitor cells, and mature astrocytes by dedifferentiation, could be the cell of origin of GBM. New approaches to GBM ...
  59. [59]
    Epidermal growth factor receptor (EGFR) and EGFRvIII in ...
    Amplification of the EGFR gene occurs in 57.4% of primary GBM patients compared to 8% of secondary GBM patients and is associated with high levels of EGFR ...
  60. [60]
    Implications of BRAF V600E mutation in gliomas - PMC - NIH
    The focus of this review is to summarize the molecular landscape of BRAF across glioma subtypes and the novel therapeutic strategies for BRAF V600E mutated ...
  61. [61]
    The relationship between Sonic hedgehog signalling, cilia and ...
    Absence of Shh ligand is associated with the midline defect holoprosencephaly, while increased Shh signalling is associated with exencephaly and spina bifida.
  62. [62]
    Nonneoplastic Cystic Lesions of the Central Nervous System ...
    Oct 7, 2019 · Both epidermoid and dermoid cysts are predominantly maldevelopmental, as a result of the inclusion of ectoderm during neural tube closure. The ...Missing: malformations | Show results with:malformations
  63. [63]
    Intracranial glioependymal (neuroglial) cysts: a systematic review
    May 25, 2018 · They are congenital benign lesions with a neuroectodermal origin that share many radiological characteristics with a variety of intracranial ...Missing: malformations | Show results with:malformations
  64. [64]
    Neural tube defects and folate: case far from closed - PMC - NIH
    A major clinical achievement has been the use of periconceptional folic acid supplements, which prevents ~50–75% of cases of neural tube defects.
  65. [65]
    Human Neuroepithelial Stem Cell Spinal Cord Repair
    Aug 24, 2018 · Human neuroepithelial stem cells may provide an anatomically specific relay function for spinal cord injury recovery.
  66. [66]
    Functional Recovery by Transplantation of Human iPSC-Derived ...
    Transplantation of hiPSC-derived neural progenitor cells (NPCs) is a promising reparative strategy for spinal cord injury (SCI), but clinical translation ...
  67. [67]
    Antidepressants increase neural progenitor cells in the human ...
    We determined whether SSRIs or TCAs increase neural progenitor (NPCs) and dividing cells in the human DG in major depressive disorder (MDD).
  68. [68]
    Serotonin-Induced Increases in Adult Cell Proliferation and ... - Nature
    Sep 3, 2003 · Among molecular factors, only some trophic factors and serotonin (5-HT) can increase cell proliferation and neurogenesis both in the SVZ and SGL ...
  69. [69]
    Phase 1 study of multiple intracerebral doses of a neural stem cell ...
    May 29, 2024 · Phase 1 study of multiple intracerebral doses of a neural stem cell-based oncolytic virotherapy for treatment of recurrent high-grade gliomas.
  70. [70]
    Building brains: neural chimeras in the study of nervous system ...
    Neural chimeras have revealed a remarkable degree of plasticity in the developmental potential of neuroepithelial precursor cells.Missing: integration | Show results with:integration
  71. [71]
    The Challenges to Advancing Induced Pluripotent Stem Cell ... - PMC
    Jan 5, 2024 · iPSC therapies pose significant unique challenges with respect to safety, potency, genetic stability, immunogenicity, tumorgenicity, cell reproducibility, ...Induced Pluripotent Stem... · Autologous Vs Allogeneic... · Engraftment Challenges Of...
  72. [72]
    Research and therapy with induced pluripotent stem cells (iPSCs)
    Nov 21, 2019 · In this review, we discuss bioethical, legal, and societal concerns associated with research and therapy using iPSCs.Human Ipscs: Methods Of... · Informed Consent In Research... · Ipscs: Potential For Human...
  73. [73]
    Merits and challenges of iPSC-derived organoids for clinical ...
    ... cells can be derived with clinically relevant phenotypes, without the ethical and immune rejection concerns associated with ESCs. Human-derived iPSCs cell ...