Fact-checked by Grok 2 weeks ago

Neuroblast

A neuroblast is a dividing progenitor cell in the nervous system whose progeny differentiate into neurons, playing a central role in neurogenesis during both embryonic and adult stages of development. These cells originate primarily from the neuroectoderm in the neural tube for the central nervous system (CNS) or from neural crest cells for the peripheral nervous system (PNS), where they delaminate from proliferative zones such as the ventricular zone and migrate to their final positions guided by radial glia and extracellular cues. In the CNS, progenitors in the ventricular zone undergo asymmetric divisions to generate neuroblasts that differentiate into postmitotic neurons, establishing precise laminar organization and connectivity essential for brain function. For the PNS, particularly the sympathetic lineage, neuroblasts arise from sympatho-adrenal progenitors expressing markers like PHOX2B and ASCL1, differentiating into noradrenergic neurons or chromaffin cells over weeks in human embryogenesis (7–17 weeks post-conception). Neuroblasts persist into adulthood in mammals, contributing to ongoing neurogenesis in regions like the subventricular zone and dentate gyrus of the hippocampus, where they support neural plasticity, repair after injury, and potentially therapeutic regeneration. Dysregulated proliferation of neuroblasts, often due to genetic alterations like MYCN amplification, can lead to neuroblastoma, the most common extracranial solid tumor in children, highlighting their dual role in normal development and oncogenesis.

Definition and Properties

Biological Definition

A neuroblast is defined as an immature neural progenitor cell that serves as a precursor to neurons in the developing nervous system. These cells undergo division to produce neurons, contributing to the diversity of cell types in the central and peripheral nervous systems. In vertebrates, neuroblasts emerge as committed intermediates in the neurogenic lineage, amplifying the output of upstream progenitors through limited proliferative divisions before terminal differentiation. The term "neuroblast" was coined in the late 19th century by Swiss anatomist Wilhelm His during his pioneering histological studies of human and animal embryos, where he described these cells as early precursors observed in the forming neural structures. His' work, utilizing innovative techniques like serial sectioning and paraffin embedding, laid the foundation for understanding neural tube histogenesis and distinguished neuroblasts from the surrounding neuroepithelium in early neuroembryology. Neuroblasts differ from neural stem cells in their restricted proliferative potential and lineage commitment; while neural stem cells exhibit extensive self-renewal and broad multipotency, neuroblasts represent downstream progenitors with finite divisions, primarily dedicated to generating post-mitotic neurons. This distinction underscores their role as transit-amplifying cells that expand neural populations without indefinite maintenance of stemness. Neuroblasts predominantly arise during embryonic development within the , where they populate the ventricular and mantle zones to form the foundational layers of the and , and from the , contributing to peripheral ganglia and autonomic structures. In adulthood, they persist in specialized neurogenic niches, such as the and subgranular zone of the , where they support ongoing, albeit limited, neuronal addition in mammals.

Cellular Characteristics

Neuroblasts are characterized by a small, round with a notably high -to-cytoplasm , often appearing as compact cells with minimal cytoplasmic relative to their euchromatic , which facilitates rapid and . In models, such as those in the (SVZ), neuroblasts exhibit an elongated shape during chain , extending pseudopodia-like processes to navigate through the rostral migratory stream toward the . This supports their identity, distinguishing them from more differentiated neurons, which possess extensive neurites and lower nuclear density. At the molecular level, neuroblasts express a suite of progenitor-specific markers that underscore their undifferentiated state, including protein Nestin, Sox2, and Doublecortin (DCX), while lacking mature neuronal markers such as NeuN. Nestin provides cytoskeletal support for their dynamic shape changes, Sox2 maintains self-renewal and multipotency, and DCX promotes stabilization essential for and early neurite extension. These markers are dynamically regulated, with their expression peaking during the transit-amplifying phase before downregulation upon neuronal commitment. Neuroblasts proliferate through rapid cell cycles dominated by asymmetric divisions, in which a single neuroblast divides to yield one self-renewing daughter cell that retains the progenitor fate and one committed progeny that differentiates into a neuron. This process ensures balanced expansion of the progenitor pool while generating neural diversity, with cell cycle lengths typically shorter in embryonic stages compared to adult contexts. In vertebrates, radial glia-like progenitors in the ventricular zone undergo such divisions, coupling mitosis with fate specification. Polarity establishment is crucial for asymmetric division, involving apical-basal orientation mediated by conserved proteins that segregate fate determinants. In model systems like , apical proteins such as the PAR complex (including aPKC) exclude basal determinants like and from the neuroblast cortex, ensuring localizes to the basal daughter cell to promote differentiation. acts as an , linking apical exclusion with basal transport of mRNAs and proteins like , thereby enforcing fate asymmetry. In vertebrates, analogous mechanisms in radial progenitors rely on similar polarity cues to orient the mitotic spindle along the apical-basal axis.

Embryonic Development in Vertebrates

Origin and Formation

Neuroblasts in the vertebrate central nervous system (CNS) primarily originate from the ventricular zone of the neural tube, where neuroepithelial progenitor cells initially form a pseudostratified epithelium during early embryogenesis. These neuroepithelial cells undergo interkinetic nuclear migration and symmetric divisions to expand the progenitor pool, subsequently transforming into radial glial cells that serve as neural stem cells capable of generating neuroblasts through asymmetric divisions. This process establishes the foundational layer for CNS neurogenesis, with the ventricular zone acting as the primary germinal niche. The specification of neuroectodermal fate, leading to neuroblast formation, is driven by key inductive signals including morphogens such as Sonic hedgehog (Shh) and Bone Morphogenetic Proteins (BMPs). Shh, secreted from the and floor plate, promotes ventral neural patterning and progenitor proliferation by activating Gli transcription factors, while BMP antagonists like Chordin and Noggin inhibit signaling to favor neural induction over epidermal fate in the presumptive . s, in turn, establish dorsal-ventral gradients that refine neuroblast identities, with low levels diffusing into the to subdivide progenitor domains expressing specific transcription factors such as Nkx6.1 (ventral) and (dorsal). In mammals, this formation begins around embryonic day 9-10 (E9-E10) in mice, coinciding with closure, and around gestational week 5-6 in humans, resulting in the initial pseudostratified that supports subsequent . For the peripheral nervous system (PNS), neuroblasts arise from cells, which delaminate from the dorsal through an epithelial-to-mesenchymal transition () to generate migratory progenitors, including sympathoadrenal neuroblasts that contribute to autonomic ganglia. This delamination is regulated by intermediate levels and transcription factors like and , enabling and subsequent differentiation into PNS neurons, with processes initiating around E8-E9.5 in mice during late .

Proliferation and Differentiation

Neuroblasts in vertebrate embryos initially undergo symmetric proliferative divisions to expand the pool, followed by a transition to asymmetric neurogenic divisions that generate one self-renewing neuroblast and one post-mitotic or glial precursor. This switch ensures balanced growth of the neural while initiating , with the timing of the transition varying across and brain regions; for instance, in the mouse telencephalon, symmetric divisions predominate early in corticogenesis before asymmetric divisions produce the majority of projection neurons. Asymmetric divisions are orchestrated by the unequal segregation of polarity proteins and determinants, such as Numb, which inhibit signaling in the neuronal daughter cell to promote its . Notch/Delta signaling plays a central role in regulating neuroblast proliferation and differentiation through lateral inhibition, where Delta-expressing cells activate Notch in neighboring progenitors to suppress proneural gene expression and maintain their undifferentiated state. This pathway promotes symmetric divisions in the progenitor pool by repressing neuronal differentiation, while its downregulation in selected cells allows proneural genes like Neurogenin (Neurog) to drive commitment to a neural fate. Proneural basic helix-loop-helix (bHLH) transcription factors, such as Neurog1 and Neurog2, are essential for initiating ; they activate downstream neuronal genes and inhibit glial fates, with Neurog expression preceding and inducing differentiation in vertebrate neural progenitors. Differentiation outcomes from neuroblasts include the production of inhibitory neurons, excitatory neurons, or glial cells, with lineage specification influenced by regional cues such as dorsoventral patterning signals. In the dorsal telencephalon, neuroblasts primarily generate projection neurons, whereas ventral regions yield interneurons; in the , early neuroblasts differentiate into a mix of and neurons depending on local Shh gradients. Glial fates emerge later, often through sustained activation that diverts progenitors from neuronal lineages. Cell cycle progression in neuroblasts is tightly regulated by cyclin-dependent kinases (CDKs), particularly CDK2 and CDK4/6, which drive to sustain , while inhibitors like p27Kip1 promote cell cycle exit and by binding and sequestering CDK-cyclin complexes. Elevated p27Kip1 levels, induced by proneural factors, accumulate in post-mitotic neurons to enforce G1 arrest, preventing re-entry into the and facilitating neuronal maturation. This regulation ensures timely withdrawal from , with disruptions in p27Kip1 leading to prolonged divisions and reduced neuronal output in models.

Migration and Positioning

During embryonic development in vertebrates, neuroblasts employ distinct modes of migration to reach their appropriate positions within the nervous system. Somal translocation involves the extension of a leading process that attaches to the pial surface, followed by the nucleus and centrosome translocating forward as the process shortens, a mechanism primarily used by early-generated subplate and layer VI neurons in the cortex. Glial-guided migration, the predominant mode for most cortical projection neurons, occurs along radial glial scaffolds extending from the ventricular zone to the pial surface, enabling an inside-out layering pattern as neuroblasts climb these fibers. Tangential migration, often orthogonal to radial glia, is characteristic of GABAergic interneurons originating from the ganglionic eminences, allowing them to disperse horizontally across the cortical plate without direct glial contact in many cases. Migration is precisely directed by extracellular guidance cues that interact with receptors on neuroblasts to regulate and . The chemokine stromal cell-derived factor-1 (SDF-1, also known as ) binds to its receptor on migrating , providing a chemotactic that orients tangential migration from the medial toward the cortical targets, with disruption leading to ectopic positioning. Similarly, the Slit proteins, acting through Robo receptors, mediate repulsive signals that channel neuroblasts away from barriers like the , ensuring accurate tangential trajectories during cortical dispersal. In specific brain regions, these modes contribute to organized . Cortical neuroblasts undergoing radial glial-guided sequentially populate layers II through VI, with later-born cells overtaking earlier ones to form the characteristic laminar structure essential for cortical function. Peripherally, neural crest-derived neuroblasts migrate ventrolaterally from the dorsal to coalesce into dorsal root ganglia, providing sensory neurons, or further to the , where they differentiate into chromaffin cells. Defects in neuroblast migration can result in severe congenital malformations, such as , characterized by a smooth cerebral surface and disrupted cortical layering due to impaired radial migration, often linked to mutations in genes like LIS1 that regulate cytoskeletal dynamics during glial-guided movement.

Adult Neurogenesis

Neuroblasts in the Adult Mammalian Brain

In adult mammals, neuroblasts persist in two primary neurogenic niches: the (SVZ) lining the and the subgranular zone (SGZ) of the hippocampal . These regions sustain the generation of new neurons throughout life, contrasting with the more widespread during embryonic stages. The SVZ niche, adjacent to the ependymal layer, supports the production of neuroblasts that migrate to the , while the SGZ contributes neurons to the hippocampal granule cell layer, potentially influencing learning and . Within the SVZ, neuroblasts (type A cells) originate from adult neural stem cells identified as type B cells, which are astroglia-like and express (GFAP). Type B cells undergo asymmetric division to produce transit-amplifying type C cells, which rapidly proliferate and differentiate into type A neuroblasts expressing (DCX) and polysialylated (PSA-NCAM). This lineage progression has been demonstrated through tracing and studies, where depleting type C and A cells prompts type B cells to regenerate the downstream populations. The existence of adult neuroblasts was first evidenced in the 1990s through bromodeoxyuridine (BrdU) labeling techniques that detected proliferating cells and their neuronal differentiation in brains. Subsequent confirmation came from genetic tracing in mice using transgenic models, such as Cre-lox systems targeting promoters like Nestin or GFAP, which specifically labeled and tracked adult-born neurons without embryonic contamination. While robust in rodents, where thousands of new neurons are generated daily in these niches, adult neurogenesis in humans remains debated and appears more limited. A 2025 study using single-nucleus RNA sequencing on postmortem human hippocampal tissue identified proliferating neural progenitors in the subgranular zone (SGZ), providing evidence for adult neurogenesis in the human hippocampus, though at levels more limited than in rodents. Evidence for SVZ neuroblasts in humans is sparser, though recent analyses suggest low-level persistence, potentially constrained by evolutionary tradeoffs for cognitive stability.

Generation and Integration Processes

In the (SVZ) of the adult mammalian brain, neuroblasts arise from transit-amplifying progenitor cells known as type C cells, which undergo rapid proliferation driven by (EGF) and (FGF) signaling to amplify neuroblast production. These type C cells respond to EGF stimulation, promoting their division and the generation of type A neuroblasts, while FGF signaling supports progenitor maintenance in the niche. Migrating neuroblasts in this process express polysialylated (PSA-NCAM), which facilitates chain migration and reduces to enable movement through the . Once generated, neuroblasts primarily migrate via the rostral migratory stream (RMS), a specialized pathway from the SVZ to the olfactory bulb, where they travel in chains ensheathed by astrocytes over distances of several millimeters. In the hippocampal dentate gyrus, neuroblast migration occurs over shorter distances, typically within the subgranular zone, allowing local integration into the granule cell layer without long-range streams. Upon reaching their destinations, neuroblasts differentiate into mature neurons, extending dendrites and axons to form synapses with preexisting circuitry through , which is critical for their functional incorporation. However, survival is selective; approximately 50% of newly generated neurons undergo during this maturation phase, pruning non-integrated cells to maintain network stability. These integrated neurons contribute to olfactory learning by enhancing discrimination and pattern separation in the , while in the , they support memory formation, particularly spatial and contextual tasks. The generation and integration of neuroblasts are modulated by external factors, with physical exercise increasing proliferation and survival through enhanced neurotrophic support, and enriched environments promoting via increased sensory stimulation and social interaction.

Development in Invertebrate Models

Neuroblasts in Drosophila

In Drosophila melanogaster, neuroblasts function as neural cells that generate the majority of the (CNS) through repeated asymmetric cell divisions, serving as a premier model for studying biology and . During embryogenesis, approximately 30 neuroblasts per hemisegment delaminate sequentially from the ventral between embryonic stages 9 and 11, adopting an apical-basal polarity that positions their nuclei basally. These neuroblasts undergo initial divisions in the embryo to produce early-born neurons and , accounting for about 10% of the adult CNS, before entering a quiescent state at the end of embryogenesis. Reactivation occurs shortly after hatching, with proliferation resuming during the first larval and continuing through the third , ultimately generating the remaining 90% of CNS neurons via hundreds of divisions per neuroblast. Drosophila neuroblasts are classified into two primary types based on their proliferative output and lineage , both present in the ventral nerve cord (VNC) and . Type I neuroblasts, which constitute the majority in the VNC and both anterior and posterior brain lobes, undergo asymmetric divisions to self-renew while producing one ganglion mother cell () per cycle; each then divides once to yield two postmitotic neurons or . In contrast, Type II neuroblasts, restricted to the ~8 per posterior brain lobe, amplify their output by dividing asymmetrically to produce an intermediate neural progenitor (INP); each INP undergoes 3–5 divisions to generate multiple s, enabling the production of up to 500 neural cells per Type II lineage. This distinction allows Type II neuroblasts to contribute disproportionately to , with INPs themselves exhibiting a modified asymmetric division akin to Type I neuroblasts. Asymmetric division in both neuroblast types ensures maintenance and daughter through the apical-basal of molecular determinants, orchestrated by and orientation machinery. is established in late by the apical Par complex (, Par-6, atypical protein kinase C), which excludes basal determinants from the apical , while alignment is mediated by the Inscuteable-Pins-Mud complex to orient the mitotic perpendicular to the axis. Key basal determinants include Numb (a Notch signaling inhibitor), (a promoting GMC ), and (a translational ); these localize as cortical crescents in the neuroblast and are inherited exclusively by the GMC. relies on adaptor proteins such as , which binds and tethers and to the basal via interactions, and Partner of Numb, which facilitates Numb localization; disruptions in these adaptors, such as Miranda mutants, lead to symmetric divisions and ectopic proliferation. Temporal patterning further diversifies neuronal identities by imposing a sequential program on neuroblast divisions, where determines subtype specification through a cascade of transiently expressed transcription factors. In embryonic VNC neuroblasts, this series begins with Hunchback (early-born neurons), transitions to Krüppel, then Pdm1/Pdm2, and finally , with each factor defining a temporal window of ~2–3 divisions that instructs distinct neuronal fates upon GMC inheritance. These transitions are driven by cross-repressive interactions and coupling—for instance, Hunchback repression requires a neuroblast division to activate Krüppel—ensuring irreversible progression and preventing fate reversion. Larval neuroblasts extend this patterning with additional factors like Seven-up following in thoracic VNC lineages, while brain neuroblasts, including Type II, employ parallel cascades (e.g., Dichaete → Grainyhead in INPs) to generate region-specific neurons over extended proliferative phases. This , first elucidated in seminal studies of VNC neuroblasts, underscores how intrinsic timers integrate with asymmetric segregation to produce neural diversity.

Comparative Insights from Other Invertebrates

In , neuroblast development features a limited number of that follow highly invariant lineages, producing a precisely fixed complement of 302 through stereotyped divisions. For instance, the ABp blastomere, an early embryonic , generates a consistent set of descendants, including specific classes such as those in the ABpl/rppap and ABpr/rppap lineages, via asymmetric cell divisions regulated by Wnt/β-catenin asymmetry (WβA) signaling. This pathway polarizes daughter cells by asymmetrically distributing SYS-1/β-catenin and POP-1/TCF, ensuring one daughter adopts a neuronal fate while the other differentiates differently, thereby enforcing the invariant production of fixed numbers without variability across individuals. In decapod crustaceans, such as (Procambarus clarkii), adult neurogenesis persists lifelong from clusters of neuroblasts in the brain's proliferative zones, particularly in the anterior medial cluster and olfactory lobe pathway, generating new that integrate into existing circuits. These neuroblasts divide asymmetrically to produce mother cells that migrate and differentiate, with rates modulated by environmental cues; serotonin regulates the in these clusters. Unlike embryonic phases where neuroblasts degenerate post-lineage completion, adult clusters are replenished in part by self-renewing divisions and by immigration of precursor cells derived from hemocytes of the , supporting in dynamic habitats. Across invertebrates, neuroblast asymmetric division represents a conserved mechanism for generating neuronal diversity, involving the unequal partitioning of determinants like Numb in Drosophila and β-catenin in C. elegans, which biases spindle orientation and fate specification in daughter cells. In many invertebrates, such as insects, Notch-mediated lateral inhibition selects neuroblasts from proneural clusters, where activated Notch in non-neuroblast cells represses proneural gene expression (e.g., via Enhancer of split in flies), ensuring singular neuroblast emergence per equivalence group; nematode models like C. elegans employ Notch in asymmetric fate decisions but lack proneural clusters. These processes highlight evolutionary conservation in balancing self-renewal and differentiation. Invertebrate development diverges from vertebrates primarily due to shorter organismal lifespans, which constrain the persistence and scale of adult neurogenic niches compared to the protracted mammalian ; additionally, invertebrates emphasize proliferation bursts tied to or molting cycles, as seen in crustacean post-embryonic remodeling, rather than continuous vertebrate-like integration in stable adult brains.

Pathological and Clinical Aspects

Neuroblastoma and Oncogenic Transformation

is the most common extracranial solid tumor in children, accounting for approximately 6–10% of all childhood cancers and up to 15% of pediatric cancer deaths. It arises from neural crest-derived sympathoadrenal neuroblasts, which are progenitor cells destined to form the and . These tumors typically develop in the or along the sympathetic chain in the , , or , reflecting the distribution of the sympathoadrenal lineage during embryogenesis. Key genetic alterations drive the oncogenic transformation of these neuroblasts, leading to uncontrolled proliferation and blocked differentiation. Amplification of the MYCN occurs in 20–25% of cases and promotes aggressive tumor growth by enhancing while inhibiting maturation into neurons or chromaffin cells. Activating mutations in the ALK gene are found in 10–15% of primary tumors, with variants accounting for most familial cases; these mutations constitutively activate downstream signaling pathways that sustain neuroblast survival and expansion. Deletions of chromosome 1p, present in about 25% of neuroblastomas, particularly those with segmental chromosomal aberrations, correlate with poor outcomes by disrupting tumor suppressor genes that normally regulate proliferation, resulting in differentiation arrest. Staging and risk assessment for neuroblastoma rely on the International Neuroblastoma Staging System (INSS), which categorizes disease from stage 1 (localized, fully resectable) to stage 4 (metastatic) and stage 4S (special case in infants with metastatic disease limited to skin, liver, or ). Risk stratification integrates INSS stage, patient age (typically <18 months for lower risk), and genetic features like MYCN amplification to classify tumors as low, intermediate, or high risk; low-risk cases often require minimal intervention, while high-risk tumors have 5-year event-free survival rates below 50%. Notably, up to 50% of infants with stage 4S disease experience spontaneous regression without therapy, attributed to heightened immune surveillance and neuroblast maturation capacity in early development. Post-2020 research has advanced understanding and treatment of by targeting oncogenic drivers and epigenetic dysregulation. , such as , have shown promise in clinical trials for ALK-mutant cases, achieving response rates over 30% in relapsed high-risk disease by blocking aberrant signaling. Epigenetic defects, including ATRX mutations in high-risk tumors, impair deposition and maintenance, leading to replicative stress and failure of progenitors to , as revealed by single-cell analyses. These insights support emerging therapies like HDAC inhibitors to restore , potentially improving outcomes in genetically driven cases. As of 2025, ongoing trials of reduced-intensity regimens, such as the N10 protocol, and immune-boosting approaches are exploring ways to improve outcomes for high-risk and relapsed cases.

Implications in Neurological Disorders

Dysregulation of neuroblast , , and in has been implicated in the pathogenesis of several neurological disorders, particularly those involving hippocampal and (SVZ) niches. In neurodegenerative conditions, chronic suppresses neuroblast generation, leading to reduced neuronal replacement and exacerbated cognitive deficits. For instance, proinflammatory cytokines such as TNF-α and IL-1β inhibit by activating and disrupting the neurogenic microenvironment, contributing to a vicious cycle of neuronal loss. Similarly, in psychiatric disorders like , genetic risk factors alter neuroblast maturation, resulting in aberrant hippocampal circuitry and impaired memory processing. In (AD), impaired adult hippocampal emerges as an early event, with a sharp decline in neuroblast numbers correlating with amyloid-β plaque accumulation and pathology. Amyloid-β oligomers interact with (TLR4), contributing to , memory impairments, and suppression of ; blocking TLR4 abolishes Aβ-induced memory deficits and can enhance . In (PD), aggregates impair SVZ neuroblast survival and migration toward the , worsening dopaminergic loss and motor symptoms. Overexpression of human in models significantly reduces newly generated survival, highlighting its direct toxic effect on neuroblasts. Epilepsy involves ectopic neuroblast migration, where seizure-induced aberrant in the leads to mossy fiber sprouting and hyperexcitability. In models, disrupted DISC1 signaling promotes abnormal neuroblast integration, contributing to chronic seizures and cognitive comorbidities. For , risk genes like DISC1 and NRG1 dysregulate neuroblast proliferation in the , linking reduced to prefrontal-hippocampal deficits and psychotic symptoms. In acute injuries such as and (TBI), neuroblasts exhibit enhanced migration from the SVZ toward lesion sites, offering reparative potential but often failing due to inflammatory barriers. Post-, matrix metalloproteinases facilitate neuroblast invasion into the peri-infarct zone, yet only a fraction integrate functionally, limiting recovery. In TBI models, reactive guide neuroblast chains via BDNF gradients, but or excessive hinders this process, perpetuating neurological deficits. These findings underscore neuroblasts as therapeutic targets, with strategies enhancing their migration—such as agents—showing promise in preclinical restoration of .

References

  1. [1]
    Glossary - Neuroscience - NCBI Bookshelf - NIH
    neuroblast. A dividing cell, the progeny of which develop into neurons. neurogenesis. The development of the nervous system. neuroglial cells. see glia ...
  2. [2]
    Neuronal Migration - Neuroscience - NCBI Bookshelf - NIH
    In the nervous system, migration during development brings different classes of neurons together so that they can interact appropriately.
  3. [3]
    Recent advances in the developmental origin of neuroblastoma
    Neuroblastoma (NB) is a pediatric tumor that originates from neural crest-derived cells undergoing a defective differentiation due to genomic and epigenetic ...
  4. [4]
    Neuroblasts - BrainFacts
    Sep 19, 2017 · These immature neural cells can grow into anything from neurons to glia as they populate the diverse cells that composes the nervous system.Missing: definition | Show results with:definition
  5. [5]
    Neural Progenitor Cell Terminology - PMC - PubMed Central - NIH
    Dec 6, 2018 · C cells are rapidly dividing transit amplifying cells that produce to Type A cells (red), which are migratory neuroblasts. B1 cells contact ...
  6. [6]
    Neurogenesis in the Developing and Adult Brain—Similarities and ...
    Adult neurogenesis in the mammalian brain is often viewed as a continuation of neurogenesis at earlier, developmental stages.
  7. [7]
    The revolutionary developmental biology of Wilhelm His, Sr - PMC
    Feb 22, 2022 · Swiss‐born embryologist Wilhelm His, Sr. (1831–1904) was the first scientist to study embryos using paraffin histology, serial sectioning ...
  8. [8]
    Historical perspective on neuroembryology: Wilhelm His ... - PubMed
    The article places His' work in the scientific context of 19th century embryology, concerned with reconciling the embryonic layers theory, the cell theory and ...
  9. [9]
    Neuroblast - an overview | ScienceDirect Topics
    Neuroblasts—precursors of neurones. The first population of cells produced in the neural tube are neuroblasts, the precursors of neurones.
  10. [10]
    Functional Coordination of WAVE and WASP in C. elegans ...
    We define two molecular pathways that activate the Arp2/3 complex during neuroblast migration in Caenorhabditis elegans. The transmembrane ...
  11. [11]
    Molecular Biomarkers for Embryonic and Adult Neural Stem Cell ...
    Here we gathered and generalized a series of NSC biomarkers emerging during the procedures of embryonic and adult neural stem cell.
  12. [12]
  13. [13]
  14. [14]
  15. [15]
    Principles and mechanisms of asymmetric cell division | Development
    Jun 29, 2020 · Asymmetric cell division (ACD) is an evolutionarily conserved mechanism used by prokaryotes and eukaryotes alike to control cell fate and generate cell ...Asymmetry Of The... · Spindle And Cell Size... · Asymmetric Segregation Of...<|separator|>
  16. [16]
  17. [17]
  18. [18]
    Miranda mediates asymmetric protein and RNA localization in the ...
    Miranda is required to localize prosperomRNA. prospero mRNA localizes on the apical side of neuroblasts at interphase (arrow in a) and moves to the basal cortex ...
  19. [19]
    Neurogenesis during development of the vertebrate central nervous ...
    Mar 17, 2014 · Here, we discuss the recent advances and insights into regulation of neurogenesis in the developing vertebrate central nervous system.
  20. [20]
    Current Understanding of the Neural Stem Cell Niches - PMC
    During embryogenesis, NSCs are localized in the ventricular zone of the neural tube and can give rise to all of the necessary cell types for CNS formation [101] ...
  21. [21]
    EvoD/Vo: The origins of BMP signaling in the neuroectoderm - PMC
    During the early phase of neural induction, the primary role of BMP signaling is to divert cells in epidermal regions from adopting the neural fate. This ...
  22. [22]
    Brain development in rodents and humans: Identifying benchmarks ...
    Neural tube formation occurs approximately mid-gestation in rodents, on gestational day (gd) 10.5–11 and 9–9.5 in rats and mice, respectively, with birth ...
  23. [23]
    Neurogenesis From Neural Crest Cells: Molecular Mechanisms in ...
    During the embryonic development of vertebrates, one of the main events after the gastrulation process is neurulation, which allows the formation of the neural ...
  24. [24]
    Neurogenesis during development of the vertebrate central nervous ...
    Mar 17, 2014 · As to neurogenesis, neural progenitors initially divide symmetrically to expand their pool and switch to asymmetric neurogenic divisions at the ...Mitotic Spindle Orientation · Signaling Pathways · Neural Progenitor Type...
  25. [25]
  26. [26]
    Notch in the Vertebrate Nervous System: An Old Dog with New Tricks
    Mar 10, 2011 · Notch receptor activation can inhibit neurogenesis, maintain neural progenitor character, and in some contexts promote gliogenesis and drive binary fate ...
  27. [27]
    Dynamic Notch signalling regulates neural stem cell state ...
    Nov 22, 2018 · The Notch signalling pathway regulates stem cell maintenance, proliferation and differentiation in different tissues, contributing to vertebrate ...
  28. [28]
    Identification of neurogenin, a Vertebrate Neuronal Determination ...
    In this way, the proneural genes both promote a neural fate cell-autonomously and inhibit this fate nonautonomously. Lateral inhibition mediated by vertebrate ...
  29. [29]
    Lineage origins of GABAergic versus glutamatergic neurons in the ...
    Recent findings show that neuronal diversity within the lineages of excitatory and inhibitory neurons is in part already established at the level of progenitor ...
  30. [30]
    Lineage Analysis Reveals Neurotransmitter (GABA or Glutamate ...
    Studies of cell lineage in the rat cerebral cortex have pro- vided new insights into the mechanisms of neuronal and glial determination.
  31. [31]
    Cell cycle regulation of proliferation versus differentiation in the ...
    May 25, 2014 · Cdkis of the Kip/Cip family, namely p21Cip1, p27Kip1 and p57Kip2, have recognised roles in regulating the activities of cyclin-cdk complexes ...Missing: neuroblast | Show results with:neuroblast
  32. [32]
    Neurogenesis and the Cell Cycle - Neuron - ScienceDirect.com
    In this process, they induce the expression of p27Kip1 and cause cell cycle arrest in G1 (Farah et al., 2000). External neural determination and proliferation ...
  33. [33]
  34. [34]
    Guiding Neuronal Cell Migrations - PMC - PubMed Central - NIH
    The migration of newly born neurons is a precisely regulated process that is critical for the development of brain architecture.
  35. [35]
    The Contribution of the Neural Crest to the Vertebrate Body - NCBI
    These cells will give rise to most of the PNS by coalescing to form the dorsal root ganglia (DRG) and, more ventrally, the sympathetic ganglion chains and the ...Missing: neuroblast | Show results with:neuroblast
  36. [36]
    Adult Mammalian Neural Stem Cells and Neurogenesis
    Radial glia-like NSCs generate intermediate progenitor cells (IPCs), which generate neuroblasts and these progenitor cells are closely associated with the ...
  37. [37]
    Adult Neurogenesis in the Mammalian Brain: Significant Answers ...
    The field of adult neurogenesis took off after the introduction of bromodeoxyuridine (BrdU), a nucleotide analog as a lineage tracer (Kuhn et al., 1996) and ...
  38. [38]
    Adult neural stem cells in the mammalian central nervous system
    May 12, 2009 · Though the specific niche architecture in the SVZ and SGZ is distinct, there are common features, including their cellular niche components and ...
  39. [39]
    Neurogenesis in Adult Subventricular Zone - PMC - PubMed Central
    The SVZ is largely separated from the ventricle cavity by a layer of ependymal cells (type E cells). B cells interact closely with E cells and occasionally ...
  40. [40]
    Neurogenesis in Adult Mammals: Some Progress and Problems
    Feb 1, 2002 · In the 1990s there were several developments that finally established neurogenesis in the adult rodent. ... BrdU labeling (Tanapat et al., 2001).
  41. [41]
    Transgenic mouse models for studying adult neurogenesis - PMC
    As many transgenic mouse models have been developed for labeling and tracing of different cell types in the adult neurogenic niches (Table 1, Figures 1, 2), it ...
  42. [42]
    Genetic evidence that our brains make new neurons in adulthood ...
    Jul 3, 2025 · ... adult rodents, but definitive evidence in humans has remained elusive. ... Early rodent experiments that identified neural progenitors in adult ...
  43. [43]
  44. [44]
    Reduced Adult Neurogenesis in Humans Results From a Tradeoff ...
    Jul 14, 2025 · This improvement in pattern separation is particularly noticeable in aged mice [26], suggesting that the age-related decrease in pattern ...Missing: differences | Show results with:differences
  45. [45]
    Neural stem cells and the regulation of adult neurogenesis - PMC
    Distinct neural stem cells proliferate in response to EGF and FGF in the developing mouse telencephalon. Dev Biol. 1999;208:166–188. doi: 10.1006/dbio ...
  46. [46]
    Fibroblast growth factor-2 signaling in neurogenesis and ...
    In adult neurogenesis, FGF2 has been implicated based on its expression and regulation of neural stem and progenitor cells in the neurogenic niches, the ...
  47. [47]
    Removal of Polysialic Acid Triggers Dispersion of Subventricularly ...
    The polysialylated form of neural cell adhesion molecule (PSA-NCAM) is expressed by these cells, and has been shown to both promote their migration and suppress ...
  48. [48]
    Adult Neurogenesis and the Olfactory System - PMC - PubMed Central
    Neuroblasts from the subventricular zone (SVZ) migrate along the rostral migratory stream (RMS) into the olfactory bulb, where they differentiate into ...
  49. [49]
    Neurogenesis in the Adult Hippocampus - PMC - PubMed Central
    Adult hippocampal neurogenesis is a multistep process that originates from a sequence of proliferative precursor cells and leads to the existence of a new ...
  50. [50]
    Synaptic integration and plasticity of new neurons in the adult ...
    We propose that adult neurogenesis represents not merely a replacement mechanism for lost neurons, but also an ongoing developmental process in the adult brain ...
  51. [51]
    Targeting Adult Neurogenesis for Poststroke Therapy - PMC
    Adult neurogenesis (AN) is a process that is continuously producing new neurons ... Thereafter, about 50% newborn neurons undergo apoptosis. Cells that ...
  52. [52]
    Adult Olfactory Bulb Neurogenesis - PMC - PubMed Central - NIH
    There are studies suggesting that newly generated neurons in the adult OB and hippocampus are critical for various aspects of learning, memory, and sensory ...Missing: exercise | Show results with:exercise
  53. [53]
    Functional Role of Adult Hippocampal Neurogenesis as a ...
    Adult neurogenesis, the process of generating new neurons from neural stem cells, plays significant roles in synaptic plasticity, memory, and mood regulation.
  54. [54]
    Physical Exercise-Induced Adult Neurogenesis: A Good Strategy to ...
    Physical exercise, a potent enhancer of adult hippocampal neurogenesis, has emerged as a potential preventative strategy/treatment to reduce cognitive decline.
  55. [55]
    Additive Effects of Physical Exercise and Environmental Enrichment ...
    Voluntary physical exercise (wheel running, RUN) and environmental enrichment both stimulate adult hippocampal neurogenesis but do so by different mechanisms.
  56. [56]
    Drosophila neuroblasts: a model for stem cell biology | Development
    Dec 1, 2012 · Drosophila neuroblasts, the stem cells of the developing fly brain, have emerged as a key model system for neural stem cell biology.<|control11|><|separator|>
  57. [57]
    Drosophila embryonic type II neuroblasts: origin, temporal patterning ...
    Dec 15, 2017 · Drosophila type I neuroblasts have a relatively simple cell lineage: they undergo a series of asymmetric cell divisions to produce a series of ...Missing: seminal | Show results with:seminal
  58. [58]
    Drosophila neuroblast asymmetric divisions: cell cycle regulators ...
    Jan 21, 2008 · In this paper, we highlight the growing number of molecules that are normally considered to be key regulators of cell cycle events/progression ...
  59. [59]
  60. [60]
    Invariant Cell Lineages in C. elegans Neurogenesis - NCBI - NIH
    The correlation of characteristic neuroblast divisions with patterns of neural differentiation suggests that a modular “program” for the generation of sets of ...
  61. [61]
    Adult neurogenesis in the crayfish brain: proliferation, migration and ...
    The birth of new neurons and their incorporation into functional circuits in the adult brain is a characteristic of many vertebrate and invertebrate organisms, ...Missing: cues | Show results with:cues
  62. [62]
  63. [63]
  64. [64]
    Adult Neurogenesis: A Common Strategy Across Diverse Species
    Adult neurogenesis, the generation of new neurons from adult precursor cells, occurs in the brains of a phylogenetically diverse array of animals.
  65. [65]
    Neuroblastoma—A Neural Crest Derived Embryonal Malignancy
    Jan 29, 2019 · Neuroblastoma is a neural crest derived malignancy of the peripheral nervous system and is the most common and deadliest tumor of infancy.
  66. [66]
    Neuroblastoma | Nature Reviews Disease Primers
    Nov 10, 2016 · Germline gain-of-function mutation in ALK is the main driver of most familial neuroblastomas. Somatic mutations in ALK and mutations in members ...
  67. [67]
    The International Neuroblastoma Risk Group (INRG) Staging System
    The International Neuroblastoma Risk Group (INRG) classification system was developed to establish a consensus approach for pretreatment risk stratification ...
  68. [68]
    Molecular principles underlying aggressive cancers - Nature
    Feb 17, 2025 · ALK inhibitors increase ALK expression and sensitize neuroblastoma cells to ALK.CAR-T cells. Cancer Cell 41, 2100–2116.e2110 (2023). Article ...
  69. [69]
  70. [70]
    Dysregulation of neurogenesis by neuroinflammation
    Increasing evidence suggests that impairments in adult neurogenesis are involved in major neurodegenerative disorders such as Alzheimer's disease. A hallmark ...
  71. [71]
    Adult neural stem cells and schizophrenia - PMC - PubMed Central
    This review focuses on the role of schizophrenia risk genes and related signaling pathways in adult hippocampal neurogenesis.Missing: neuroblasts | Show results with:neuroblasts
  72. [72]
    Impaired adult neurogenesis is an early event in Alzheimer's ...
    Oct 7, 2019 · Depletion of adult neurogenesis exacerbates cognitive deficits in Alzheimer's disease by compromising hippocampal inhibition. Mol ...
  73. [73]
    Adult neurogenesis in Parkinson's disease - PMC - PubMed Central
    In summary, these studies show a detrimental effect of human alpha-synuclein on the generation and the survival of newly generated neurons, depending either on ...
  74. [74]
    Disrupted in Schizophrenia 1 Regulates Ectopic Neurogenesis in ...
    Jul 1, 2022 · Studies have shown that hippocampal granule cells played a critical role in the pathophysiology of TLE (Hester and Danzer, 2014, Kelly and Beck, ...
  75. [75]
    Involvement of Matrix Metalloproteinase in Neuroblast Cell Migration ...
    Mar 29, 2006 · However, after stroke and other forms of brain injury, neuroblasts swerve away from their designated path and migrate toward damaged tissue ...<|control11|><|separator|>
  76. [76]
    Neuroblasts migration under control of reactive astrocyte-derived ...
    Jan 5, 2023 · Neuroblasts migrated along the activated astrocytic tunnel, directed by BDNF gradient between SVZ and injured cortex after TBI.
  77. [77]
    Research Advances in Neuroblast Migration in Traumatic Brain Injury
    This paper underlines the partial biological features of neuroblast migration and unravels the significance and mechanisms of the vasculature in the process.