Fact-checked by Grok 2 weeks ago

Neural stem cell

Neural stem cells are self-renewing, multipotent progenitor cells that generate , , and , thereby forming the primary cellular components of the during embryonic and contributing to limited . These cells originate from the neuroepithelium and persist in specific adult niches, such as the and hippocampal , where they maintain quiescence or proliferate in response to injury or environmental cues. Empirical evidence from models confirms their capacity for self-renewal and multilineage , though adult neural stem cells exhibit more restricted potency and slower proliferation compared to embryonic counterparts. Key research milestones include the isolation of multipotent neural stem cells from the adult mammalian brain in the early 1990s, demonstrating their potential to form neurospheres and differentiate into neural lineages. This discovery challenged prior assumptions of fixed postnatal brain cellularity and opened avenues for studying neurogenesis mechanisms. In therapeutic contexts, neural stem cells hold promise for regenerative treatments in neurodegenerative disorders like and injuries, with preclinical transplants showing neuronal replacement and functional recovery in animal models. However, clinical translation faces substantial hurdles, including poor long-term engraftment, risk of tumorigenicity from uncontrolled proliferation, and limited efficacy in human trials due to the brain's complex microenvironment and immune barriers. Controversies persist regarding source variability—embryonic versus induced pluripotent-derived cells—and ethical concerns over embryonic sourcing, though adult-derived neural stem cells mitigate some moral objections while introducing scalability issues. Despite optimism in peer-reviewed literature, causal analyses reveal that microenvironmental signaling disruptions often undermine therapeutic outcomes, underscoring the need for rigorous, mechanism-driven validation over anecdotal successes.

Definition and Fundamental Properties

Biological Definition and Characteristics

Neural stem cells (NSCs) are defined as the self-renewing, multipotent cells of the that give rise to all major (CNS) cell types, including neurons, , and . These cells originate from the early neuroepithelium during embryonic development and persist in restricted niches into adulthood in mammals. Unlike more restricted s, NSCs exhibit the functional capacity to both maintain their undifferentiated state through and generate differentiated progeny via lineage commitment. A core characteristic of NSCs is self-renewal, the process by which a single NSC divides to produce at least one identical daughter NSC, enabling long-term maintenance of the pool. This can occur via symmetric , yielding two NSCs, or asymmetric , producing one NSC and one committed ; the balance between these modes is regulated by intrinsic factors like transcription regulators and extrinsic signals from the niche microenvironment. Self-renewal ensures sustained potential without depleting the reservoir, as demonstrated in clonal assays where NSCs form self-renewing spheres . Multipotency distinguishes NSCs from lineage-restricted progenitors, allowing into the three principal CNS glial and neuronal lineages under appropriate cues. In vivo, embryonic NSCs contribute to the entire CNS, while adult NSCs primarily generate region-specific neurons and , such as granule neurons in the or olfactory . This potency is evidenced by transplantation studies where human NSCs integrate and into functional host-appropriate types, underscoring their therapeutic relevance. NSCs also display quiescence in adults, a reversible dormant state that preserves longevity by limiting division until activated by injury or demand.

Identification Markers and Assays

Neural stem cells (NSCs) are prospectively identified through expression of specific molecular markers and validated via functional assays that confirm their defining properties of self-renewal and multipotency, the capacity to generate neurons, , and . Intracellular markers such as Nestin, an associated with cytoskeletal organization in undifferentiated cells, , a regulating pluripotency and , and Musashi-1, an promoting asymmetric division, are routinely used via or RT-PCR; however, these lack specificity as they are expressed across neural progenitors and transit-amplifying cells in regions like the (SVZ). In adult SVZ NSCs, additional enriched markers include CRBP1 (cellular retinol-binding protein 1, confirmed by in Nestin-positive cells), HMGA1 (high-mobility group AT-hook 1, nuclear expression), OTX2, PRDM16, RXRα, and , identified through , RT-PCR, and analyses showing >1.5-fold SVZ-specific upregulation. For prospective isolation, particularly of human fetal or developing brain NSCs, cell-surface markers enable fluorescence-activated cell sorting (FACS) to distinguish subtypes without relying on intracellular staining. A 2023 study dissociated GW17–19 human brain tissue and used combinations of , , , , and to isolate ten NSPC classes, including ventricular radial glia (CD24⁻ THY1⁻/lo EGFR⁺), outer radial glia (CD24⁻ THY1⁻/lo EGFR⁻), and oligodendrocyte precursors (THY1⁺ EGFR⁻ PDGFRA⁺), validated by index sorting and single-cell RNA sequencing correlating phenotypes to transcriptomes.
NSPC TypeKey Cell-Surface Marker Combination
Ventricular Radial GliaCD24⁻ THY1⁻/lo EGFR⁺
Outer Radial GliaCD24⁻ THY1⁻/lo EGFR⁻
Excitatory Neuron PrecursorsCD24⁺ THY1⁻/lo EGFR⁺ CXCR4⁻
Oligodendrocyte PrecursorsTHY1⁺ EGFR⁻ PDGFRA⁺
Functional assays complement markers by directly testing NSC properties. The neurosphere assay, involving dissociation to single cells cultured in EGF/FGF-supplemented media to form clonally derived floating spheres, assesses self-renewal through serial passaging (e.g., initiation frequency of 1 in 4.5 for certain radial glia subtypes) but is critiqued for overestimating true stem cell frequency due to progenitor contamination, aggregation artifacts, and failure to capture quiescent cells. Multipotency is verified by differentiating neurospheres or sorted cells into lineage-specific markers: DCX⁺ neurons, GFAP⁺ astrocytes, and O4⁺ oligodendrocytes in vitro, or via xenotransplantation into neonatal immunodeficient mice, where engrafted cells generate all three lineages after 6 months, confirming in vivo self-renewal and differentiation without tumor formation. Alternative clonogenic assays, such as neural colony-forming cell assays, quantify large colonies from single cells to discriminate stem from progenitor activity based on size and passage potential. These assays, when combined with marker profiling, provide rigorous evidence of NSC identity, though variability in culture conditions (e.g., growth factor concentrations) can bias outcomes toward glial fates.

Anatomical Locations

Embryonic Neural Stem Cell Niches

The ventricular zone (VZ) serves as the primary niche for neural stem cells during embryonic (CNS) development, lining the lumen of the and subsequently the ventricles. This pseudostratified emerges shortly after neural tube closure, around embryonic day 9.5 in mice, and harbors progenitors that generate the full repertoire of CNS cell types, including neurons, , and . Radial glial cells (RGCs) predominate within the VZ as the key neural stem cells, exhibiting self-renewal through symmetric divisions early in development and transitioning to asymmetric divisions to produce intermediate progenitors and differentiated neurons. These cells display characteristic apical-basal polarity, with apical processes contacting the ventricular lumen via primary cilia and basal extensions reaching the pial surface, facilitating nutrient uptake and signaling integration. The VZ niche microenvironment integrates multiple components to regulate RGC behavior, including direct exposure to (CSF), which flows through the ventricular lumen and delivers soluble factors such as to promote progenitor replication and . Emerging vasculature within and adjacent to the VZ supplies endothelial-derived signals like pigment epithelium-derived factor (PEDF) and betacellulin, which modulate quiescence, , and lineage commitment of RGCs. elements, including fractones rich in collagen IV, , and proteoglycans, anchor RGCs and sequester growth factors such as fibroblast growth factor-2 (FGF-2) and bone morphogenetic protein-7 (BMP-7), thereby fine-tuning local signaling gradients. Interkinetic of RGC nuclei synchronizes with apical CSF access and basal interactions, ensuring balanced expansion of the progenitor pool. Regulation within the embryonic VZ niche relies on intrinsic and extrinsic cues to maintain multipotency while driving . Transcription factors like and sustain RGC identity and prevent premature differentiation, while microRNA-124 times the onset of neuronal fate specification. signaling from adjacent progenitors and differentiated cells inhibits excessive differentiation, preserving the reservoir through mechanisms. Regional heterogeneity arises early, with VZ subdomains specified by embryonic day 11.5 in mice—such as pallial domains for cortical progenitors and subpallial for striatal—determining future contributions to postnatal niches like the . Precursors destined for adult neural stem cells, including B1-type cells, are generated between embryonic days 13.5 and 15.5, entering quiescence until postnatal reactivation, underscoring the VZ's role as a transient yet foundational niche. This embryonic organization contrasts with adult niches by its higher proliferative rate and lack of ependymal barriers, reflecting the demands of rapid brain patterning.

Adult Neurogenic Regions

In the adult mammalian brain, constitutive neurogenesis is restricted to two primary germinal zones: the subventricular zone (SVZ), which lines the walls of the , and the subgranular zone (SGZ) of the hippocampal dentate gyrus. These niches contain quiescent neural stem cells (NSCs) that asymmetrically divide to produce transit-amplifying progenitors, which differentiate into neuroblasts and mature neurons integrated into neural circuits.00348-5) Adult in these regions declines with age, influenced by factors such as and reduced NSC proliferation, but persists at low levels throughout life in rodents and nonhuman primates. The SVZ consists of a layered where type B1 cells—radial glia-like —serve as the resident NSCs, contacting the ventricular surface and blood vessels via processes. These NSCs generate type C transit-amplifying cells, which proliferate rapidly to yield type A neuroblasts that chain-migrate rostrally through the subventricular zone-rostral migratory stream (SVZ-RMS) toward the , where they differentiate into granule or periglomerular . In , this process replenishes inhibitory circuits for olfaction, with daily production rates estimated at thousands of new neurons; in humans, SVZ-derived cells may instead contribute to striatal , though migration distances are shorter and rates lower. SVZ is regulated by neurotransmitters like and , which modulate NSC quiescence and proliferation via niche signaling. The SGZ, embedded at the interface of the granule cell layer and hilus in the dentate gyrus, features type-1 radial glia-like NSCs that extend processes to the molecular layer and generate intermediate progenitors (type-2 cells), progressing to neuroblasts that integrate as dentate granule neurons. These new neurons, born postnatally, exhibit heightened plasticity, contributing to hippocampal-dependent learning, pattern separation, and mood regulation, with integration occurring over 4-6 weeks via dendritic arborization and synaptic formation. In mice, SGZ neurogenesis yields approximately 900 new neurons daily per hippocampus in young adults, declining sharply after middle age due to NSC exhaustion and microenvironmental changes like vascular rarefaction. Human SGZ neurogenesis follows a similar trajectory but at debated rates, with immunohistochemical evidence confirming DCX+ neuroblasts up to age 59, albeit with early childhood peaks and subsequent decline. While the SVZ and SGZ represent the canonical sites, sporadic evidence suggests low-level or injury-induced in other regions like the or in , though these lack constitutive stem cell niches and robust integration, remaining controversial and non-replicable in . Such findings, often from non-specific markers or transient progenitors, do not challenge the primacy of SVZ/SGZ but highlight potential NSC reservoirs activated under or .

Developmental Origins and Regulation

In Vivo Embryonic and Fetal Development

Neural stem cells (NSCs) originate from neuroepithelial progenitors in the during early , which folds to form the , the precursor to the (CNS). In humans, neural tube formation initiates in the third gestational week (GW3), spanning embryonic days 20 to 27 (E20-E27), with anterior neuropore closure by E25 and posterior by E27. These neuroepithelial cells, lining the nascent , exhibit initial pseudostratified morphology and undergo interkinetic migration, a hallmark of their proliferative state. Transition to radial glial cells, the primary embryonic NSCs, occurs as the expands, positioning these progenitors apically in the ventricular zone (VZ) adjacent to the lumen. Early NSC proliferation features symmetric divisions to amplify the progenitor pool, predominant from E25 to E42, establishing the foundational CNS architecture by GW8. Neurogenesis onset aligns with GW5, marked by a shift to asymmetric divisions, where one daughter cell retains stem-like properties while the other differentiates into neurons or intermediate progenitors (IPs). In the developing telencephalon, IPs delaminate into the subventricular zone (SVZ), undergoing further divisions to generate diverse neuronal subtypes; this process peaks between GW8 and GW26, producing over 80% of cortical neurons in an inside-out laminar pattern via radial and tangential migration guided by radial glia scaffolds. Human-specific outer radial glia (oRG) emerge in the outer SVZ around GW8-12, driving neocortical expansion through basal process extension and oblique divisions, distinct from rodent counterparts. Fetal development sustains NSC activity, with cortical neurogenesis extending to GW24-25, followed by gliogenesis from GW13 onward, yielding astrocytes post-neurons and oligodendrocytes last in a temporally ordered sequence. Quiescent-like states may emerge in select progenitors by mid-fetation, foreshadowing adult NSC persistence, though most embryonic NSCs exhaust via terminal differentiation. Key regulators include intrinsic transcription factors (e.g., , Emx1 for regional identity) and extrinsic cues like Sonic hedgehog (Shh) for ventral patterning, Wnt for proliferation, and for maintaining stemness via . Disruptions, such as in defects, underscore the precision of these dynamics, with empirical lineage tracing confirming direct NSC-to-neuron/ trajectories without persistent multipotency in most lineages.

Adult Persistence and Quiescence Mechanisms

Adult neural stem cells (NSCs) persist from embryonic origins through two primary models: the conventional pathway, where postnatal radial glia-like cells in the (SVZ) and subgranular zone (SGZ) transition into quiescent NSCs (qNSCs) by approximately postnatal day 14, and the set-aside model, wherein a subset of embryonic progenitors enter quiescence as early as embryonic day 13.5, marked by expression of the cell cycle inhibitor p57 (Cdkn1c). This persistence is facilitated by niche-specific factors such as (VCAM1), which promotes the conversion of activated NSCs to a quiescent state, ensuring long-term maintenance of the NSC pool without premature depletion. Quiescence in adult NSCs represents a reversible G0 arrest characterized by low metabolic activity, including reduced protein synthesis, , and , alongside heightened sensitivity to local environmental cues for potential reactivation. This state preserves NSC identity and prevents exhaustion, supporting lifelong primarily in the SVZ and SGZ niches. Intrinsic mechanisms sustaining quiescence involve cell cycle regulators such as p21 (Cdkn1a), , and p57, which enforce G0 arrest and suppress symmetric proliferative divisions, with p57 particularly critical in early set-aside populations. Transcription factors like , , Klf9, and Bhlhe40 further maintain dormancy by repressing proliferative genes and promoting quiescence-associated programs, while epigenetic modifiers such as Setd1a sustain H3K4 methylation on targets like Bhlhe40 in hippocampal qNSCs, with its deficiency leading to premature activation and pool depletion in mouse models. Downregulation of UHRF1 and miR-9/ interplay also epigenetically reinforces this state. Extrinsic regulation occurs via the NSC niche, comprising , endothelial cells, and , where signaling pathways including Notch1/2/3 (via ligands like Jagged1 and Dll1), (through BMPR-IA), Wnt/β-catenin, and Eph-ephrin interactions inhibit proliferation and promote quiescence. Physiological low oxygen levels (~8%) activate Wnt/β-catenin to sustain , and choroid plexus-derived miR-204 fine-tunes niche signaling. Metabolic shifts, such as reliance on oxidation, further stabilize quiescence independently of niche inputs. These mechanisms collectively balance persistence against activation demands, with dysregulation linked to aging-related NSC decline.

In Vitro Derivation and Expansion Techniques

Neural stem cells (NSCs) are derived in vitro primarily from pluripotent stem cells or primary neural tissues. Differentiation from human embryonic stem cells (hESCs) or induced pluripotent stem cells (iPSCs) employs neural induction protocols, such as dual inhibition of SMAD signaling pathways (BMP and TGF-β) to promote rosette formation and subsequent NSC generation, yielding populations expressing markers like SOX2 and Nestin within 10-20 days. Direct isolation from embryonic or fetal brain tissue involves enzymatic dissociation (e.g., using papain or trypsin) followed by selective culturing in serum-free media supplemented with fibroblast growth factor-2 (FGF-2), achieving viable hNSCs from gestational weeks 8-12 tissues as described in protocols established by 2022. From iPSCs, feeder-free monolayer differentiation avoids embryoid body formation by sequential exposure to retinoic acid or Wnt agonists, generating primitive NSCs capable of long-term propagation while retaining multipotency, as demonstrated in methods yielding radial glia-like cells by 2019. These derivation approaches prioritize high-purity neural commitment over heterogeneous populations, though variability in donor genetics influences efficiency, with iPSC-derived NSCs showing transcriptional profiles akin to primary fetal NSCs but potential epigenetic drift over passages. Expansion of derived NSCs maintains self-renewal through mitogenic signaling in defined, serum-free conditions. Suspension neurosphere cultures, initiated by low-density plating (10^4-10^5 cells/mL) with (EGF) at 20 ng/mL and FGF-2 at 20 ng/mL, allow clonal and achieve up to 10^7-fold over extended periods while preserving multipotency, as evidenced in progenitors cultured for over one year. Adherent methods on or substrates enhance homogeneity and scalability by reducing core in larger aggregates, supporting 10-15 population doublings per passage with EGF/bFGF supplementation, outperforming neurospheres in yield for therapeutic-scale production per 2015 optimizations. (LIF) at 10 ng/mL synergizes with EGF and FGF-2 to sustain quiescence-like states and prevent premature , enabling passages beyond 20 while monitoring via BrdU incorporation or Ki67 expression. Insulin-like growth factor-1 (IGF-1) further potentiates in striatal NSCs by activating PI3K/Akt pathways, though its omission halts , underscoring dependency on multiple trophic cues for fidelity to niches. Challenges include stochastic and after 30-50 passages, mitigated by (3-5% O2) or bioreactors to improve metabolite exchange and yields up to 10-fold.

Molecular Signaling and Migration Dynamics

The plays a central role in maintaining neural stem cell (NSC) quiescence and self-renewal within adult niches like the (SVZ) and subgranular zone (SGZ), primarily through activation of transcriptional repressors such as Hes1, which inhibit proneural genes and prevent premature differentiation. Wnt/β-catenin signaling promotes NSC proliferation and biases toward neuronal differentiation, with astrocyte-derived Wnt3 in the hippocampal driving production; disruption via dominant-negative Wnt reduces in vivo. Sonic Hedgehog (Shh) signaling sustains progenitor proliferation in both SVZ and SGZ, with overexpression increasing by up to twofold in models, while loss impairs NSC maintenance. In contrast, (BMP) signaling via BMPR-IA enforces quiescence and glial fates, suppressing ; antagonism by Noggin in the SVZ enhances neuronal output. These pathways interact dynamically, with niche-derived factors like FGF and EGF modulating their balance to regulate NSC fate transitions empirically observed in conditional knockouts and overexpression studies. NSC dynamics are orchestrated by gradients and , enabling tangential chain of SVZ neuroblasts toward the or injury sites. Shh acts as a chemoattractant in this process, with pathway ablation halting neuroblast chains in SVZ models. Post-injury homing relies on the SDF-1α/ axis, where SDF-1α upregulation by and at lesions (e.g., infarcts) correlates with NSC influx (r = +0.6079, P < 0.0001 in mouse stroke models); -expressing NSCs show enhanced transmigration and chain complexity , but blockade reduces to ischemic explants by over 80% (P < 0.001). This involves downstream of p38 MAPK and paxillin for cytoskeletal remodeling. β1- signaling further facilitates perivascular along endothelial scaffolds in post-stroke brains, as demonstrated by integrin inhibition disrupting NSC tracking . These mechanisms ensure targeted dispersal, with empirical validation from lineage tracing and chemotactic assays showing radial and tangential modes distinct from developmental gliogenesis.

Physiological Functions

Self-Renewal and Multipotency

Neural stem cells exhibit self-renewal, the ability to undergo cell divisions that produce at least one identical to the in terms of potency and proliferative , thereby maintaining the stem cell pool over extended periods. This property is essential for sustaining , particularly in adult neurogenic niches like the (SVZ) and hippocampal , where NSCs often remain quiescent and activate symmetric self-renewing divisions only under specific cues to expand without immediate differentiation.30003-1) In vitro, self-renewal is evidenced by the formation of self-renewing neurospheres from single cells, capable of serial passaging while retaining proliferative potential, as demonstrated in assays isolating NSCs from embryonic or adult tissues. Mechanisms include asymmetric division, where orientated ensures one progeny inherits stemness factors, and regulatory pathways involving Bmi-1, which sustains by repressing p16^Ink4a and p19^Arf to prevent . Multipotency denotes the capacity of NSCs to generate multiple differentiated cell types within the , primarily , , and , distinguishing them from unipotent progenitors. This is rigorously demonstrated through clonal analyses , where retrovirally labeled single NSCs in the adult SVZ produce progeny across all three lineages, contributing to circuitry and confirming lineage potential at the single-cell level.00585-X.pdf) differentiation protocols further validate multipotency, with NSCs expressing markers like nestin and yielding β-III-tubulin-positive neurons, GFAP-positive astrocytes, and O4-positive oligodendrocytes upon exposure to growth factor withdrawal or lineage-specific inductors. Epigenetic controls, such as modifications and , underpin this flexibility, allowing NSCs to toggle between self-renewal and fate commitment without loss of core competence. Self-renewal and multipotency are tightly coupled yet balanced to prevent exhaustion or aberrant proliferation; for instance, REST maintains quiescence and stemness by repressing pro-differentiation genes like those in the pathway, while its absence leads to premature depletion of the NSC pool. remodelers like Lsh/HELLS promote symmetric divisions for pool expansion during development but shift toward asymmetry in adulthood to ensure multipotent output. Disruptions, such as in aging, impair these properties, reducing self-renewal efficiency and biasing toward gliogenesis over , as quantified by decreased neurosphere-forming units and lineage-restricted clones in aged murine models.30340-4)

Differentiation Pathways and Epigenetic Controls

Neural stem cells (NSCs) primarily differentiate along three major lineages: neuronal, astroglial, and oligodendroglial, with fate decisions governed by sequential activation of transcription factors and signaling cascades. In embryonic development, early NSCs favor through proneural factors like Neurogenin1 (Neurog1) and NeuroD1, which drive neuronal commitment by binding enhancers marked by active modifications such as H3K27ac. As development progresses, a gliogenic switch occurs, promoting formation via factors like nuclear factor I (NFI) proteins and signal transducer and activator of transcription 3 (), activated by cytokines such as ciliary neurotrophic factor (CNTF). Oligodendrocyte differentiation follows, involving oligodendrocyte transcription factor 2 (Olig2) and its downstream targets like , which orchestrate myelination programs in response to (PDGF) signaling. Epigenetic mechanisms impose stable, heritable controls on these pathways by modulating accessibility and without altering DNA sequence. , particularly at CpG islands in promoter regions, represses neurogenic genes like NeuroD1 in gliogenic phases by recruiting methyl-CpG-binding proteins that compact , as observed in NSC models where Ten-eleven translocation () enzymes mediate demethylation to enable . modifications provide dynamic switches: bivalent domains featuring (activating) and (repressing) poise multipotent NSCs for lineage-specific , with Polycomb repressive complex 2 (PRC2) enforcing astroglial bias in late gestation via EZH2-mediated on neuronal loci. Non-coding RNAs, including microRNAs like miR-124, further fine-tune fates by targeting mRNAs of epigenetic regulators, such as , to promote neuronal exit from quiescence. In adult NSCs, these controls maintain quiescence through elevated at proliferation genes, with requiring demethylases like JMJD3 for injury-induced . These epigenetic layers ensure fidelity in fate determination, with disruptions—such as aberrant TET2 activity—leading to skewed , as evidenced in models where Tet2 knockout impairs astrogliogenesis while enhancing oligodendrogenesis via altered Olig2 expression. Metabolic inputs intersect epigenetically, as α-ketoglutarate from fuels TET enzymes to demethylate DNA, linking nutrient availability to NSC potency. Recent human iPSC-derived NSC studies confirm conserved mechanisms, revealing an epigenetic "timer" via barriers that delays maturation until post-mitotic stages, independent of transcriptional changes.

Contributions to Brain Homeostasis and Plasticity

Neural stem cells (NSCs) in the mammalian brain, residing mainly in the along the and the subgranular zone of the in the , sustain by generating neurons and that replenish cellular populations lost to or physiological turnover, thereby preserving integrity and functional equilibrium. This ongoing , occurring at rates of approximately 700 new neurons per day in the human under basal conditions, integrates into preexisting networks to maintain excitatory-inhibitory balance and prevent circuit hyperexcitability. Evidence from mouse models demonstrates that NSC quiescence-activation cycles are tightly regulated to match homeostatic demands, with disruptions leading to premature exhaustion of the NSC pool and impaired self-renewal capacity. Beyond replacement, NSCs contribute to homeostasis through paracrine signaling via their secretome, which includes neurotrophic factors like brain-derived neurotrophic factor (BDNF) and vascular endothelial growth factor (VEGF), fostering microvascular stability and modulating inflammation to support niche integrity during aging. In the vascular niche, NSCs interact with endothelial cells to regulate blood-brain barrier permeability and homeostasis, ensuring metabolic support for neuronal activity and preventing dyshomeostasis in energy-demanding regions. For brain plasticity, adult-born neurons derived from NSCs exhibit heightened excitability and during a 4-6 week integration window, enabling enhanced (LTP) and facilitating adaptive rewiring in response to environmental stimuli. In the , these neurons are critical for pattern separation, a computational process distinguishing similar experiences, as selective reduces accuracy in tasks by up to 50% in . NSC-derived , particularly those responsive to physiological states like , transiently boost olfactory bulb circuitry to enhance sensory , demonstrating context-dependent plasticity under homeostatic modulation. This dual role in and positions NSCs as biosensors of systemic signals, such as exercise or , which upregulate to amplify circuit adaptability without destabilizing core functions. Longitudinal studies in confirm that NSC activity correlates with , where sustained buffers against age-related decline in executive function. However, the precise causal contribution remains debated, as correlative data from human postmortem analyses show variability in NSC output across individuals, influenced by genetic and lifestyle factors.

Roles in Aging and Disease

Aging profoundly impairs the regenerative capacity of adult (NSCs), with a primary manifestation being the deepening of their quiescent state, which restricts proliferation and in key niches such as the (SVZ) and hippocampal . In models, NSC quiescence increases progressively from early to mid-hood, with studies showing a shift toward dormancy-associated changes by approximately 6-12 months of age, correlating with reduced cell cycle entry and fewer activated NSCs (defined as Sox2+/Nestin+/+). This decline is evident in diminished production of neuroblasts (DCX+/Tuj1+) and mature neurons (+), contributing to overall loss of observed post-developmentally. Intrinsic molecular mechanisms reinforce this enhanced quiescence, including upregulation of cell cycle inhibitors such as p57Kip2, which maintains dormancy and prevents premature exhaustion but becomes overly restrictive in aging, and p16INK4a, whose increased expression directly suppresses progenitor proliferation. Epigenetic and metabolic shifts further entrench quiescence; for instance, age-related loss of O-GlcNAc modification in hippocampal NSCs promotes a glial rather than neuronal fate upon activation attempts. and proteostasis defects, including impaired function in quiescent NSCs from aged SVZ, lead to protein aggregate accumulation and mitochondrial dysfunction, reducing the cells' ability to exit . Regulators like Id4 and CHD7 also sustain quiescence by eliminating pro-activation factors or preventing depletion, though their overactivity in aging exacerbates functional decline. Extrinsic niche alterations amplify these intrinsic barriers, with aged environments featuring increased inflammation—such as IFNγ signaling from T cells—and stiffened , which inhibit NSC activation. Wnt pathway antagonists rise in the niche, imposing greater hurdles to quiescence exit compared to young brains. Nutrient-sensing pathways, including insulin/IGF1-FOXO signaling, and reduced choroid plexus-derived factors further modulate this process, linking systemic aging to local NSC . Recent CRISPR-Cas9 screens in aging models have identified novel regulators of quiescence-to-proliferation transitions, highlighting targets like metabolic and epigenetic modifiers that could be intervened upon to restore function. This quiescence enhancement, while initially protective against replication stress and exhaustion, becomes maladaptive, driving age-related cognitive deficits through curtailed homeostasis and . Human postmortem studies corroborate rodent findings, revealing reduced hippocampal in older individuals associated with memory impairment. Interventions enhancing or reducing niche , such as activation, have shown promise in reactivating aged NSCs in mice, suggesting causal links amenable to therapeutic targeting.

Involvement in Neurodegenerative Disorders

Neural stem cells (NSCs) contribute to neurodegenerative disorders through impaired , characterized by reduced , excessive quiescence, and defective , which fail to compensate for neuronal loss and exacerbate pathology. In (AD), (SVZ) declines early at presymptomatic stages, driven by intracellular amyloid-beta oligomers (AβOs) that inhibit NSC activation and survival, as evidenced in transgenic models where AβO accumulation correlates with diminished production. Hippocampal is similarly compromised, with human postmortem studies showing fewer doublecortin-positive immature neurons and reduced markers like Ki-67 in AD brains compared to controls. This NSC dysfunction links to cognitive deficits, as conditional ablation of hippocampal in AD models worsens memory impairment and promotes hyperphosphorylation in nascent neurons. In Parkinson's disease (PD), NSC impairment manifests as reduced neurogenesis in the hippocampal subgranular zone (SGZ), influenced by alpha-synuclein aggregates that disrupt NSC quiescence-exit mechanisms and dopaminergic signaling. Rodent models overexpressing alpha-synuclein demonstrate fewer proliferating NSCs and neuroblasts, with pathology spreading to impair NSC niches via inflammation and oxidative stress. Quiescent NSCs, which predominate in adult brains, enter deeper dormancy in PD, limiting repair; epigenetic factors like NFIX-mediated enhancer regulation fail to activate cell-cycle genes, as shown in aging NSC transcriptomes overlapping PD signatures. Amyotrophic lateral sclerosis (ALS) involves NSC exhaustion in the SVZ, where motor neuron degeneration correlates with diminished NSC-derived oligodendrocytes and astrocytes, per human iPSC-derived models revealing SOD1 mutations suppress NSC multipotency. Across disorders, vascular dysfunction and mitochondrial in NSC niches amplify quiescence; for instance, integrated response preserves NSC viability but at the cost of under proteotoxic conditions. Empirical data from lineage-tracing in mice indicate that NSC depletion accelerates neurodegeneration, underscoring their causal role in failure rather than mere bystander effects. While therapeutic NSC modulation shows promise elsewhere, endogenous NSC dysregulation directly sustains neuronal vulnerability by curtailing plasticity and repair.

Responses to Injury and Endogenous Repair

Following (CNS) injury, such as or (TBI), endogenous neural stem cells (NSCs) primarily residing in the (SVZ) and of the transition from quiescence to an activated state, initiating within hours to days post-injury. In models of focal , SVZ NSC increases markedly within the first week, peaking around days 3-7 before subsiding to baseline levels after several weeks. This response is triggered by injury-induced signals including hypoxia-inducible factors and inflammatory cytokines like IL-1β and TNF-α, which exert dual effects by promoting initial NSC expansion while potentially inhibiting long-term survival if unchecked. Proliferating NSCs generate neuroblasts that migrate toward the injury site, often traveling distances up to 2 mm in stroke models, guided by chemotactic gradients such as stromal cell-derived factor-1α (SDF-1α) binding to receptors on migrating cells. Additional mediators include (VEGF), which peaks early post-injury and supports both migration and , and (EGF), which enhances proliferation via signaling. In TBI models, similar SVZ NSC mobilization occurs, with neuroblasts directing toward the infarct boundary, though migration efficiency diminishes with lesion size and chronic inflammation. Upon reaching the lesioned area, these cells attempt , predominantly into and rather than neurons, contributing to and limited remyelination. In stroke studies, only a small fraction (~0.2% of lost striatal neurons) of migrated neuroblasts mature into functional neurons expressing markers like , with most undergoing due to hostile microenvironments characterized by and scar formation. (BDNF) and other secreted by activated NSCs promote some neuronal survival and synaptic integration, yet overall contributions to tissue replacement remain minimal. The endogenous repair process aids in reducing lesion volume and modulating inflammation through paracrine effects, such as neurotrophic factor release (e.g., NGF, GDNF), but fails to restore full functionality owing to insufficient cell numbers, poor circuit integration, and inhibitory extracellular matrix changes like chondroitin sulfate proteoglycans. In human-relevant contexts, postmortem analyses and imaging studies corroborate heightened SVZ proliferation post-injury, yet the reparative output does not scale to compensate for substantial neuronal loss, as evidenced by persistent deficits in cognition and motor function. This limited efficacy underscores the niche's evolutionary prioritization of homeostasis over robust regeneration, with epigenetic barriers like DNA methylation further constraining neuronal fate commitment.

Dysregulation in Brain Tumors

Neural stem cells (NSCs) in the adult (SVZ) serve as a primary cellular origin for glioblastomas, the most aggressive primary brain tumors, through oncogenic mutations that disrupt self-renewal and differentiation controls. These mutations, including alterations in TP53, , and PTEN, transform quiescent or proliferative NSCs into glioblastoma-initiating cells (GICs) capable of serial tumor propagation in orthotopic xenografts. Experimental evidence from genetically engineered models demonstrates that SVZ NSCs harboring glioma driver mutations, such as KrasG12D and homozygous Trp53 deletion, initiate multifocal tumors mimicking human multiforme (GBM) invasion patterns. Dysregulated signaling pathways central to NSC maintenance, including , Wnt/β-catenin, and , are hyperactivated in GICs, promoting unchecked proliferation and resistance to . For instance, aberrant signaling, which normally sustains NSC quiescence during neocortical , drives GIC self-renewal and tumor heterogeneity by inhibiting neuronal and enabling glioma progression. Similarly, Wnt pathway upregulation in SVZ-derived GICs enhances stem-like properties, as evidenced by increased β-catenin nuclear localization in patient-derived GBM spheres that retain multipotency akin to NSCs. In tumor progression, GICs exhibit NSC-like migration dynamics, invading along tracts and perivascular niches to evade therapies and fuel recurrence. Single-cell sequencing of recurrent GBM reveals enriched NSC signatures in therapy-resistant subclones originating from distant SVZ mutations, with up to 30% of relapses linked to NSC transformation rather than local clonal evolution. This dysregulation contributes to intratumoral heterogeneity, where GIC subpopulations differentiate into cells while preserving a reservoir, as shown in lineage-tracing studies where outer radial glia-like progenitors give rise to diverse GBM phenotypes. Therapeutic targeting of NSC-GIC parallels remains challenging due to shared vulnerabilities, such as reliance on hypoxia-inducible factors for niche survival, but selective inhibition of dysregulated pathways like mutant IDH1 in proneural GBM subtypes has shown promise in reducing frequency in preclinical models. However, clinical trials targeting or , such as with γ-secretase inhibitors, have yielded limited efficacy, attributed to pathway redundancy and adaptive in GICs. These findings underscore that NSC dysregulation not only initiates gliomas but sustains their through evolutionary advantages in mutagenic microenvironments.

Research Applications

Disease Modeling with Derived Cells

Derived cells from neural stem cells (NSCs), often generated via intermediate NSC stages from induced pluripotent stem cells (iPSCs), enable recapitulation of disease phenotypes in human neural lineages such as neurons, , and . These models leverage patient-derived iPSCs to preserve genetic variants associated with disorders, facilitating mechanistic studies and high-throughput drug screening that bypass interspecies differences inherent in models. Differentiation typically involves dual-SMAD inhibition to produce neural rosettes, followed by expansion as NSCs using factors like CHIR99021 and SB431542, then lineage specification with morphogens such as sonic hedgehog (SHH) and (RA). In (AD) modeling, iPSC-derived cortical neurons from familial () patients with or PSEN1 mutations exhibit elevated amyloid-beta 42 (Aβ42) secretion and Aβ42/Aβ40 ratios, alongside hyperphosphorylated and enlarged early endosomes, phenotypes observed as early as 2012 studies. Sporadic -derived neurons similarly display mitochondrial dysfunction, , and impaired , with showing reduced complexity and aberrant . For (PD), dopaminergic neurons from mutation carriers demonstrate nuclear architecture defects in NSC intermediates and reduced uptake with α-synuclein accumulation in mature neurons, as reported in 2012 analyses. These findings correlate with mitochondrial impairments and network hyperexcitability, aiding evaluation of compounds like inhibitors. Amyotrophic lateral sclerosis (ALS) models using s from or C9orf72 mutant iPSCs reveal ER stress, mitochondrial fragmentation, and hyperexcitability leading to neurite degeneration, with phenotypes emerging after 2014 optimizations in differentiation protocols. in these systems exacerbate motor neuron vulnerability via non-cell-autonomous toxicity. Despite scalability for screening—evidenced by identification of neuroprotective agents in and assays—these models face constraints from cellular immaturity, inter-line variability, and incomplete recapitulation of late-onset epigenetic changes, potentially underestimating full disease progression. Isogenic controls via editing have mitigated some genetic confounders, enhancing reliability for causal validation.

Regenerative Therapies for CNS Conditions

Neural stem cell (NSC) transplantation represents a primary approach in regenerative therapies for (CNS) conditions, aiming to replenish lost neurons, , and while modulating the injury microenvironment through neurotrophic factor secretion and anti-inflammatory effects. Preclinical studies have shown NSCs differentiating into neural lineages and promoting remyelination or axonal regrowth in models of (SCI) and , though human translation remains limited by engraftment efficiency and immune rejection risks. Clinical efforts prioritize fetal- or spinal cord-derived NSCs, often delivered intrathecally or directly into lesions, with phase I trials establishing feasibility rather than robust efficacy. In , a phase I trial involving intramedullary transplantation of spinal cord-derived NSCs (NSI-566) in 25 patients with thoracic injuries reported no treatment-related serious adverse events over 7-10 years of follow-up, with showing no ectopic growth or expansion. Four participants achieved one- to two-level improvements in sensory or motor per the International Standards for Neurological Classification of Spinal Cord Injury (ISNCSCI) scale, alongside modest gains in walking ability measured by the Walking Index for Spinal Cord Injury, though remains uncertain without controls. A separate multicenter study of 13 patients transplanted with similar NSCs confirmed at one year, with no radiographic of and preliminary voluntary motor improvements in some, but emphasized the need for phase II efficacy data. For (ALS), intracerebroventricular delivery of human NSCs has been tested in phase I , such as a 2023 study of 21 patients receiving escalating doses, which found no serious adverse events and stable clinical/laboratory parameters over 18 months. Participants exhibited slowed motor decline, with ALS Functional Rating Scale-Revised scores decreasing 30% less than predicted natural progression, and reduced atrophy on imaging, attributed to NSC-derived trophic support rather than widespread neuronal replacement. Another targeting ALS disability via intrathecal NSC injection aimed to assess measurable functional impacts but reported primarily endpoints without transformative outcomes. In progressive (PMS), a 2023 phase I (STEMS) transplanted human fetal neural progenitor cells—closely akin to NSCs—into the cerebral of 15 patients, demonstrating safety with no immunosuppression-related deaths or tumor formation after two years. Modest stabilization of disability scores occurred in some, linked to limited remyelination and , though larger trials are required to confirm therapeutic value amid variable patient responses. For , NSC therapies promote repair via and in models, with emerging human data suggesting reduced infarct volume but lacking randomized evidence of functional recovery. Parkinson's disease applications remain largely preclinical, where NSCs engineered to produce dopamine precursors have alleviated symptoms in animal models by integrating into the , but clinical translation faces hurdles in precise and long-term graft viability. Genetically modified NSCs, incorporating genes for factors like GDNF, enhance survival and efficacy in CNS injury models, with ongoing trials exploring combinatorial delivery to amplify repair. Overall, while safety profiles support advancement to phase II/III studies, empirical outcomes indicate incremental rather than curative benefits, underscoring the need for optimized sourcing, delivery, and host integration strategies.

Bioengineering Approaches and Scaffolds

Bioengineering approaches for neural stem cells (NSCs) increasingly incorporate biomaterial scaffolds to mimic the , facilitate , and guide toward neuronal lineages in (CNS) repair. These scaffolds, often hydrogels or nanofibrous structures, provide mechanical support and biochemical cues that enhance NSC survival, proliferation, and integration post-transplantation, addressing limitations like poor engraftment in ischemic or traumatic injuries. For instance, collagen-based scaffolds have demonstrated the ability to direct endogenous neural stem/progenitor cells (NSPCs) toward neuronal by optimizing the microenvironment for growth and myelination. Hydrogels, prized for their injectability and , represent a dominant scaffold type in NSC bioengineering. methacryloyl (GelMA) hydrogels, when co-cultured with NSCs and mesenchymal stem cells (BMSCs), promote regeneration in models by fostering synergistic and reducing , as shown in studies from 2024 where low-modulus variants improved axonal extension. Similarly, composite hydrogels incorporating conductive nanomaterials, such as carbon nanotubes or , stimulate NSC differentiation via electrical cues; exposure to 5–20 V potentials has yielded increased neurite outgrowth and neuronal marker expression . Myoglobin-functionalized hydrogels further bolster NSC integration by creating oxygen reservoirs, enhancing graft functionality in models as evidenced by improved behavioral outcomes in 2023 experiments. Nanofiber and 3D-printed scaffolds offer tunable to emulate native neural , promoting directed and formation. Electrospun scaffolds loaded with NSCs have supported mesenchymal stem cell-mediated repair in neural injury models, bridging cavities and reducing formation through aligned fiber orientations that guide regrowth. In human (iPSC)-derived NSC applications, composite hydrogels post-ischemic transplantation in 2023 studies preserved cell viability above 70% at day 7 and upregulated neurogenic genes like Nestin and β-III tubulin, outperforming non-scaffolded controls. Hybrid scaffolds combining hydrogels with decellularized matrices address multifactorial repair needs, as reviewed in 2023 analyses emphasizing their role in modulating while sustaining NSC quiescence-to-activation transitions. Despite advances, design must balance biodegradability with mechanical stiffness to avoid inhibiting NSC potency; overly rigid matrices, for example, shift toward glial fates, per causal observations in CNS paradigms. Ongoing refinements, including incorporation of growth factors or CRISPR-edited NSCs, aim to amplify these effects, with preclinical data from 2024–2025 underscoring potential for scalable therapies in neurodegenerative contexts.

Clinical Trials and Empirical Outcomes

In phase I clinical trials, neural stem cell (NSC) transplantation has demonstrated feasibility and safety across various neurological conditions, with limited but encouraging signals of in small cohorts. For progressive , the STEMS trial transplanted human fetal neural progenitor cells (hfNPCs) into the cervical of 12 patients across four escalating dose cohorts. Primary outcomes confirmed tolerability, with no dose-limiting toxicities, severe procedure-related adverse events, or evidence of tumor formation or rejection over 96 weeks of follow-up; secondary showed dose-dependent reductions in ( r=0.73, P=0.007) and gray matter loss (r=0.66, P=0.02), alongside cerebrospinal fluid shifts suggestive of . For chronic thoracic spinal cord injury, a single-site phase I study implanted spinal cord-derived NSCs (NSI-566) into four patients, achieving surgical feasibility without major complications. Long-term follow-up to five years revealed sustained safety, with two participants exhibiting durable neurological gains, including improved motor and sensory scores, electromyography-detectable innervation, and reduced pain, while the other two showed no functional decline but stable outcomes. In , phase I trials of intraspinal NSC injections in up to 18 patients reported procedural safety and absence of severe adverse events, though functional benefits were transient and survival prolongation unconfirmed in long-term analyses, highlighting the need for optimization. Emerging phase I data for using human embryonic stem cell-derived dopaminergic progenitors (functionally akin to NSCs) in 12 patients met safety endpoints at 12-18 months, with no graft-related tumors, hemorrhages, or dyskinesias; high-dose recipients (2.7 million cells per ) showed greater motor improvements on MDS-UPDRS scores (-23 points off-medication) and increased "good ON" time (+2.7 hours) versus low-dose (-8.6 points, +0.2 hours), corroborated by imaging of graft survival. Across these trials, empirical outcomes underscore NSC tolerability under , modest neurorestorative potential via integration or trophic effects, but underscore the predominance of early-phase data with small samples (n<20 per study), variable cell sources (fetal, induced, or spinal-derived), and inconsistent efficacy metrics, necessitating phase II/III validation for disease modification. No NSC-specific trials have advanced to phase III approval as of 2025, with ongoing challenges including graft rejection risks and limited scalability.00445-4)

Challenges, Criticisms, and Controversies

Technical and Biological Limitations

Neural stem cells (NSCs) exhibit inherent biological constraints, primarily stemming from their quiescent state in the mammalian , where the majority remain dormant with low metabolic activity and heightened to local signaling cues, restricting their and in response to physiological demands or injury. This quiescence serves to preserve the finite NSC pool over a lifetime but limits endogenous , as quiescent cells must balance entry into the with risks of depletion, often favoring asymmetric divisions that yield progenitors over symmetric self-renewal. Aging exacerbates this, with increased quiescence duration and reduced reactivation, contributing to diminished neuronal production and impaired repair capacity. Differentiation potential of NSCs is further biologically constrained, showing a preferential bias toward glial lineages (astrocytes and ) over neurons in the , partly due to microenvironmental factors and intrinsic epigenetic controls that hinder neuronal fate commitment. Self-renewal mechanisms, regulated by pathways like signaling, impose additional limits by gating entry and preventing unchecked expansion, which, while protective against exhaustion, curtails the overall regenerative output. The scarcity of NSCs in tissues—confined to niches like the and —compounds these issues, yielding insufficient cells for robust repair without exogenous supplementation. Technical challenges in NSC research and application include difficulties in and , where assays like neurospheres fail to reliably distinguish true multipotent stem cells from progenitors due to heterogeneity and variable self-renewal markers, often overestimating stemness. Expansion is inefficient, with poor scalability from limited starting numbers and risks of losing stem-like properties during passaging. In transplantation therapies, NSCs face low post-engraftment survival rates, often below 5-10% in preclinical models, attributed to hostile host environments, inadequate vascularization, and immune-mediated rejection despite . Integration into existing neural circuitry remains elusive, with transplanted cells frequently failing to form functional synapses or migrate accurately, potentially disrupting delicate brain architecture during delivery procedures like intracerebral injection. Tumorigenicity risks persist, particularly with pluripotent-derived NSCs, due to incomplete and residual proliferative potential, necessitating rigorous purity controls that current protocols struggle to achieve consistently.

Ethical Debates on Cell Sources

The primary ethical debate surrounding cell sources for neural stem cell research centers on the use of human embryonic stem cells (hESCs), which requires the destruction of early-stage embryos to derive pluripotent cells capable of into neural lineages. This process involves disaggregating blastocysts, typically 4-5 days post-fertilization, resulting in the irreversible loss of the embryo's developmental potential, a practice critics equate with the termination of nascent human life. Opponents of hESC sourcing, drawing from principles of human dignity and the sanctity of life, contend that human embryos possess inherent moral status from fertilization onward due to their unique genetic identity and organized developmental trajectory toward , rendering their sacrificial use for impermissible regardless of therapeutic promise. Proponents, however, argue for a graduated moral status, positing that pre-implantation embryos lack , individuality, or viability outside a , thus permitting their use under strict oversight when balanced against potential cures for neurodegenerative diseases like Parkinson's, where neural stem cell-derived therapies could replace lost neurons. This divide has influenced global policies, with some jurisdictions prohibiting hESC derivation while allowing on existing lines, though from neural applications shows hESCs' superior pluripotency compared to alternatives, fueling utilitarian defenses. In response to these concerns, researchers have shifted toward non-embryonic sources, including adult neural stem cells harvested from regions like the adult or , which avoid embryo destruction but raise secondary issues of limited proliferative capacity and ethical sourcing if derived from fetal neural tissue obtained via elective abortions. Induced pluripotent stem cells (iPSCs), reprogrammed from somatic cells such as fibroblasts using factors like Oct4, , , and c-Myc (reported by Yamanaka in 2006), offer an ethically preferable alternative by generating patient-specific neural progenitors without involving or fetuses, thereby sidestepping debates over intrinsic rights while enabling autologous transplants to minimize immune rejection. Despite these advances, iPSC sourcing introduces subtler ethical challenges, including for donor cells—given their potential into gametes or whole organisms—and risks of unintended applications like reproductive , though these are deemed less grave than embryonic destruction by most ethicists. neural cells, while ethically uncontroversial in , face for inefficient yields in culture, prompting hybrid approaches that prioritize iPSCs for scalable neural differentiation, as validated in studies modeling via generation. Overall, the progression from hESCs to iPSCs reflects a causal prioritization of ethical feasibility driving , with ongoing debates underscoring the between scientific and foundational respect for human origins.

Safety Risks and Overhyped Claims

Neural stem cell transplantation carries significant safety risks, primarily due to the cells' proliferative capacity, which can lead to uncontrolled growth and tumor formation. In a documented case from 2009, a pediatric patient with ataxia-telangiectasia developed a donor-derived multifocal brain tumor following intrathecal injection of neural stem cells, highlighting the potential for malignant transformation from exogenous cells. Laboratory studies have further demonstrated that long-term cultured human neural stem cells can undergo spontaneous genetic and epigenetic changes, resulting in tumorigenic phenotypes capable of forming tumors in animal models. Similarly, transplantation of human embryonic stem cell-derived neural progenitors has been associated with teratoma formation in postischemic brain environments, underscoring environment-specific risks of aberrant differentiation. Additional hazards include immune-mediated rejection, ectopic tissue formation, and off-target migration, which may exacerbate neurological deficits. Expansion of neural stem cells in culture often involves animal-derived sera, introducing risks of or viral transmission and xenogeneic immune responses upon transplantation. Clinical trials have reported adverse events such as transient , headaches, and inflammatory responses, though some phase I studies for conditions like and have observed no severe treatment-related events at short-term follow-up. However, these findings are preliminary, with long-term surveillance revealing potential for late-onset complications like or in broader contexts applicable to neural applications. Claims surrounding neural stem cell therapies have often been overhyped, promising regenerative cures for neurodegenerative diseases like Parkinson's and despite limited empirical validation beyond preclinical models. After decades of research, including over 25 years since key advancements, no neural stem cell intervention has achieved regulatory approval for widespread clinical use in disorders, with efficacy largely confined to modest symptomatic relief or animal data. and clinic promotions frequently exaggerate outcomes from early-phase trials, such as isolated cases of functional improvement, while downplaying the absence of robust, randomized controlled for modification. This disparity has fueled unproven interventions, contributing to patient harms and eroding scientific credibility, as evidenced by expert consensus on the prevalence of unsubstantiated commercial offerings.

Historical Milestones

Early Discoveries in Neurogenesis

In 1962, Joseph Altman published the initial experimental evidence challenging the dogma that no new neurons form in the adult mammalian brain, using tritiated autoradiography to label dividing cells in adult rats. He observed heavily labeled neurons and neuroblasts in structures including the , of the , and , suggesting ongoing proliferation and migration of neuronal precursors. Subsequent studies by Altman and Gopal Das in 1965 detailed postnatal in the , , , and other regions, quantifying labeled cells via and confirming their neuronal identity through morphology and persistence of labels over time. These findings indicated that adult brains retain proliferative zones capable of generating functional neurons, though methodological constraints like label dilution and lack of lineage tracing limited conclusive proof of integration. Altman's work encountered significant resistance, as the prevailing view—rooted in y Cajal's assertions and reinforced by mid-20th-century neuroanatomical studies—held that mammalian ceases after early development, with adult confined to synaptic remodeling. Critics questioned whether labeled cells represented true neurons or , and the results were often dismissed due to insufficient replication and the era's technical challenges in distinguishing cell types. Despite this, Altman's publications in journals like and Journal of Comparative Neurology provided foundational data, including time-course analyses showing label incorporation peaking 24 hours post-injection and persisting in mature neurons. Confirmation emerged in the 1970s through Michael Kaplan's electron microscopy investigations, which visualized ultrastructural details of progenitor proliferation and differentiation into granule cells in the rat dentate gyrus subgranular zone. Kaplan's 1977 studies demonstrated chain migration of newly born neurons toward the granule cell layer and their morphological maturation, including dendrite and synapse formation, offering direct morphological evidence absent in earlier autoradiographic methods. He also extended observations to the neocortex and conducted initial primate experiments, revealing sparse but detectable neurogenesis in adult rhesus monkeys, thus bridging rodent models to higher mammals. These advances substantiated Altman's claims by addressing prior interpretive ambiguities, establishing neurogenesis as a verifiable process localized to specific neurogenic niches like the hippocampus and subventricular zone.

Key Advances in Isolation and Culture (1960s–1990s)

In the 1960s, foundational evidence for adult neurogenesis emerged through autoradiographic studies using tritiated to label dividing cells in the brains of adult mammals. Joseph Altman demonstrated that new granule cells continued to form in the and hippocampal of adult rats, challenging the prevailing dogma that the mammalian brain lacked regenerative capacity post-development. These findings, initially met with skepticism due to technical limitations in tracing cell lineages, established the existence of proliferative zones in the adult (CNS), laying groundwork for later identification without achieving direct or culture. Advances in isolation and culture accelerated in the early with the development of methods to propagate multipotent neural precursors from adult tissue. In 1992, Brent Reynolds and Samuel Weiss isolated cells from the striatal of adult mouse brains, dissociating them and culturing in serum-free medium supplemented with (EGF), which induced proliferation into floating aggregates termed neurospheres. These neurospheres exhibited self-renewal upon dissociation and replating, and upon withdrawal of growth factors, differentiated into neurons, , and , confirming multipotency and fulfilling operational criteria for neural stem cells. Similar protocols applied to embryonic CNS tissue yielded comparable results, broadening the evidence for neural stem cell presence across developmental stages. The neurosphere assay, refined through clonal analyses, became a cornerstone for assessing stem cell properties, enabling serial passaging and expansion while distinguishing stem cells from progenitors based on sustained multipotency over multiple generations. By the mid-1990s, extensions of this work confirmed EGF-responsive multipotent cells in additional adult regions, such as the , using combined EGF and (bFGF) supplementation to enhance yield and viability. These techniques overcame prior barriers to culturing non-adherent CNS cells, providing empirical tools for dissecting stem cell quiescence, niche signals, and lineage commitment, though debates persisted on whether all neurosphere-initiating cells were true stem cells or included lineage-restricted progenitors.

Modern Developments and iPSC Era (2000s–Present)

The generation of induced pluripotent stem cells (iPSCs) marked a pivotal shift in neural stem cell research, enabling the reprogramming of adult somatic cells into a pluripotent state without relying on embryonic sources. In 2006, Shinya Yamanaka's team demonstrated that mouse fibroblasts could be reprogrammed into iPSCs using four transcription factors—Oct3/4, , , and c-Myc—achieving pluripotency comparable to embryonic stem cells.00377-6) This was extended to human cells in 2007, with Yamanaka and colleagues deriving iPSCs from human dermal fibroblasts via similar factors, opening avenues for patient-specific neural lineages.01471-0) These advances addressed ethical constraints of embryonic stem cells while providing renewable sources for neural stem cell (NSC) derivation, as iPSCs could be differentiated into neural progenitors expressing markers like Nestin and Sox2. Subsequent refinements focused on directing iPSCs toward neural stem cell fates, with protocols emerging for efficient generation of multipotent NSCs capable of self-renewal and into neurons, , and . By 2008, researchers reported the production of functional neurons from human iPSCs, confirming their neural potential through and marker expression. Further optimizations in the early yielded stable NSC lines from iPSCs using dual-SMAD inhibition or gradients, enhancing yield and purity while minimizing heterogeneity—key for , with differentiation efficiencies reaching over 90% in some serum-free systems. These iPSC-derived NSCs mirrored endogenous counterparts in profiles and trilineage potential, validated via single-cell sequencing, facilitating models of human inaccessible via primary adult NSCs limited by scarcity and donor variability. The iPSC era spurred integrative applications, including disease modeling and preclinical therapeutics, with NSCs generated from patient iPSCs revealing mechanisms in disorders like Parkinson's and through CRISPR-edited lines. Brain organoids derived from iPSC-NSCs, first reported in , recapitulated cortical layering and interconnectivity, advancing of neurodevelopment. By the , xeno-free and feeder-free protocols reduced contamination risks, while direct reprogramming techniques bypassed pluripotency for faster NSC induction from fibroblasts using factor cocktails like and FoxG1, yielding transplantable cells with improved integration in rodent models. These developments, though promising, underscore ongoing needs for maturation fidelity, as iPSC-NSCs often exhibit immature phenotypes compared to fetal counterparts, per transcriptomic analyses.

References

  1. [1]
    Neural stem cells: developmental mechanisms and disease modeling
    Neural stem cells (NSCs) are the stem cells of the nervous system. During development they give rise to the entire nervous system.
  2. [2]
    Neural Stem Cell - an overview | ScienceDirect Topics
    Neural stem cells (NSCs) are defined as self-renewing and multi-lineage derivatives of the early neuroepithelium that can generate neurons, astrocytes, ...
  3. [3]
    Current Understanding of the Neural Stem Cell Niches - PMC
    Neural stem cells (NSCs) are self-renewing, multipotent cells which give rise to all components of the central nervous system (CNS) during embryogenesis, ...
  4. [4]
    Neural stem cells: origin, heterogeneity and regulation in the adult ...
    Feb 18, 2019 · Summary: This Review discusses the identification, regulation and heterogeneity of neural stem cells and examines recent insights into ...Introduction · Gene expression of V-SVZ... · Maintenance of V-SVZ neural...
  5. [5]
    Neural Stem Cells | Circulation Research
    This review focuses on the nature and functional properties of stem cells of the adult mammalian central nervous system (CNS).
  6. [6]
    Neural stem cells: ready for therapeutic applications?
    Oct 15, 2014 · NSCs are immature cells present in the developing and adult Central Nervous System (CNS). Typically, NSCs are defined by three cardinal ...
  7. [7]
    Neural Stem Cells: Historical Perspective and Future Prospects
    May 26, 2011 · How a single fertilized cell generates diverse neuronal populations has been a fundamental biological problem since the 19th century.
  8. [8]
    Neural Stem Cells: Historical Perspective and Future Prospects - PMC
    Age of Rationalism: Origin of neural stem cell research. The realization that human brain development begins from the initially multipotent dividing cells ...
  9. [9]
    The therapeutic prospects and challenges of human neural stem ...
    Sep 2, 2022 · Human neural stem cells (NSCs) hold the potential to functionally replace lost neurons, reinforce impaired synaptic networks, and repair the damaged AD brain.
  10. [10]
    Neural stem cells: therapeutic potential for neurodegenerative ...
    Sep 17, 2012 · Areas of controversy: Individual laboratories may be using phenotypically diverse NSCs, since these cells have been differentiated by a ...Missing: applications | Show results with:applications
  11. [11]
    Adult human neural stem cell therapeutics - PubMed Central
    The controversies could be derived from the diverse types of stem cells, and from their unique pros and cons. Compared with other stem cell sources, aNSCs have ...
  12. [12]
    Current progress in the derivation and therapeutic application of ...
    However, there are still many issues and controversies associated with the derivation and therapeutic application of these cells.
  13. [13]
    Ethical Implications in the Use of Embryonic and Adult Neural Stem ...
    Even so, the use of embryonic stem cells presents legal and ethical limitations, such as the obvious destruction of live embryos to obtain stem cells.Missing: controversies | Show results with:controversies<|control11|><|separator|>
  14. [14]
    Prospects and Limitations of Using Endogenous Neural Stem Cells ...
    Furthermore, a fundamental ethical problem lies in the therapeutic use of ESCs, which are derived from blastocysts. Thus, there are serious problems ...
  15. [15]
    Neural Stem Cells: Generating and Regenerating the Brain
    Oct 30, 2013 · Stem cells afford the plasticity to generate, repair, and change nervous system function. Combined with reprogramming technology, human somatic cell-derived ...Main Text · Introduction · Enter Pluripotent Stem...<|separator|>
  16. [16]
    Neural stem cells in adult neurogenesis and their therapeutic ...
    The therapeutic application of neural stem cells holds significant promise for addressing neurodegenerative diseases, including Alzheimer's disease, stroke, ...
  17. [17]
    Neural stem cells: Brain building blocks and beyond - PMC
    Apr 19, 2012 · Neural stem cells are the origins of neurons and glia and generate all the differentiated neural cells of the mammalian central nervous system.
  18. [18]
    Neural Stem Cells (NSCs) and Proteomics - PMC - PubMed Central
    Neural stem cells, which are present both during development and in the adult, are most commonly defined by the ability to self-renew and the capacity to ...Abstract · Fig. 1 · Nsc Methodologies<|separator|>
  19. [19]
    Concise Review: Self-Renewal in the Central Nervous System
    A novel, immortal, and multipotent human neural stem cell line generating functional neurons and oligodendrocytes. Stem Cells. 2007;25:2312–2321. doi ...
  20. [20]
    Introduction to Neural Stem Cells | Stroke
    Feb 1, 2007 · Self-renewal, the ability of a neural stem cell to produce another neural stem cell can be symmetric—where 1 neural stem cell produces ...
  21. [21]
    Neural Stem Cell Self-renewal - PMC - PubMed Central
    This review focuses on cell-intrinsic regulators ... Batteries of transcription factors have been proposed to control neural stem cell self-renewal ( ...
  22. [22]
    In Vivo Fate Analysis Reveals the Multipotent and Self-Renewal ...
    Nov 15, 2007 · Neural stem cells (NSCs) are defined as cells that can self-renew (the capacity to proliferate to produce identical cells) and are multipotent ( ...Missing: assays | Show results with:assays
  23. [23]
    A Critical Review of Sphere-Formation as an Assay For Stem Cells
    Indeed, two markers commonly used to identify neural stem cells, nestin and Sox2, are expressed by all cell types in this region, and therefore cannot be used ...
  24. [24]
    Identification of molecular markers distinguishing adult neural stem ...
    ABSTRACT. Neural stem cells (NSCs) in the adult subventricular zone (SVZ) are regionally specified and have distinct molecular gene expression signatures.
  25. [25]
  26. [26]
  27. [27]
    The Glial Nature of Embryonic and Adult Neural Stem Cells - PMC
    Radial glia have apical-basal polarity: apically (down), RG contact the ventricle, where they project a single primary cilium; basally (up), RG contact the ...
  28. [28]
    Embryonic Origin of Postnatal Neural Stem Cells
    ### Summary of Ventricular Zone as an Embryonic Niche for Neural Stem Cells
  29. [29]
    Regulation of Adult Neurogenesis in Mammalian Brain - MDPI
    In the adult brain, neurogenesis is mainly localized in two specialized niches, the subgranular zone (SGZ) of the dentate gyrus and the subventricular zone (SVZ) ...
  30. [30]
    Adult Mammalian Neural Stem Cells and Neurogenesis
    There are two major neurogenic niches in the adult mammalian brain where endogenous NSCs reside, the subventricular zone (SVZ) lining the lateral ventricles and ...
  31. [31]
    how does adult neurogenesis decrease with age? | Inflammation ...
    Jun 18, 2020 · Adult neurogenesis occurs throughout life in restricted brain regions in mammals. However, the number of neural stem cells (NSCs) that ...
  32. [32]
    Human adult neurogenesis across the ages - NIH
    There are two major neurogenic niches in the adult human brain, the subventricular zone (SVZ) underlying the wall of the lateral ventricles and the subgranular ...
  33. [33]
    Neurotrophic Factor Control of Adult SVZ Neurogenesis - PMC
    The two brain regions that contain the majority of neural stem cells are the subgranular zone (SGZ) of the hippocampus and the subventricular zone (SVZ) of the ...Neurotrophic Factor Control... · Bdnf And Svz Neurogenesis · Trkb And Svz Neurogenesis<|separator|>
  34. [34]
    Neurotransmitters couple brain activity to subventricular zone ...
    Neurogenesis exists in two regions of the adult brain: the hippocampal subgranular zone of the dentate gyrus and the subventricular zone (SVZ) along the lateral ...
  35. [35]
    Orchestrating transcriptional control of adult neurogenesis - PMC
    This review summarizes recent progress on the molecular control of adult neurogenesis in the SGZ and SVZ, focusing on the role of specific transcription factors ...
  36. [36]
    Subventricular Zone Versus Hippocampal Neural Stem Cells - PMC
    Nov 27, 2020 · NSCs are found in two neurogenic niches: the subventricular zone (SVZ), lining the walls of the lateral ventricles, and the subgranular zone ( ...
  37. [37]
    Neurogenesis and progenitor cell distribution in the subgranular ...
    Aug 6, 2013 · Progenitor cell proliferation is ubiquitous in the subventricular zone (SVZ) and subgranular zone (SGZ) of adult mammalian brains, however, ...
  38. [38]
    Adult Hippocampal Neurogenesis: A Coming-of-Age Story
    Dec 5, 2018 · We published a paper in 1996 describing the generation of new neurons in the adult rat brain and the decline of hippocampal neurogenesis during aging.<|separator|>
  39. [39]
    Adult Neurogenesis in the Mammalian Brain: Significant Answers ...
    Active adult neurogenesis is spatially restricted under normal conditions to two specific “neurogenic” brain regions, the subgranular zone (SGZ) in the dentate ...
  40. [40]
    Beyond the Hippocampus and the SVZ: Adult Neurogenesis ...
    Adult neurogenesis has been best described in the hippocampus and the subventricular zone (SVZ), in which a series of distinct stages of neuronal development ...
  41. [41]
    Adult neurogenesis and aging mechanisms: a collection of insights
    Oct 23, 2023 · Adult neurogenesis is also present in the mouse hypothalamus, although at lower levels compared to the V-SVZ and the SGZ. Engel et al ...
  42. [42]
    Mechanisms and Functional Implications of Adult Neurogenesis
    Feb 22, 2008 · Although NSCs can be isolated from many areas of the adult nervous system, adult neurogenesis has only been consistently found in the SVZ and ...
  43. [43]
    The Basics of Brain Development - PMC - PubMed Central
    Human brain development is a protracted process that begins in the third gestational week (GW) with the differentiation of the neural progenitor cells and ...
  44. [44]
    Physical biology of human brain development - PMC
    Gestational week 5 marks the onset of neurogenesis. Around gestational week 5, the progenitor cells in the ventricular zone, the radial glial cells, begin to ...
  45. [45]
    The Challenging Pathway of Treatment for Neurogenesis ...
    May 11, 2022 · Cortical neurogenesis starts at GW9 and is completed at GW24-25. Cerebellar and hippocampal neurogenesis continue beyond birth. Cerebellar ...
  46. [46]
    Quiescent Adult Neural Stem Cells: Developmental Origin and ...
    Apr 24, 2024 · In this review, we mainly focus on the recent progress of the developmental origin and regulatory mechanisms that maintain qNSCs under normal conditions.
  47. [47]
  48. [48]
    Waking up quiescent neural stem cells: Molecular mechanisms and ...
    Apr 23, 2020 · Most NSCs in mammalian adult brains are quiescent, but in response to extrinsic stimuli, they can exit from quiescence and become reactivated to give rise to ...
  49. [49]
    Epigenetic maintenance of adult neural stem cell quiescence in the ...
    Jul 6, 2024 · Quiescence, a hallmark of adult neural stem cells (NSCs), is required for maintaining the NSC pool to support life-long continuous ...
  50. [50]
    Pluripotent stem cell-derived neural stem cells - PubMed Central - NIH
    Stemness, an essential characteristic of a stem cell, involves properties of self-renewal and the potential to differentiate into functional somatic cells.Pluripotent Stem... · Neural Stem Sphere As An In... · Neural Stem Sphere-Derived...
  51. [51]
    Derivation and culturing of neural stem cells from human embryonic ...
    Aug 18, 2022 · Here, we present a protocol to safely and successfully derive and culture hNSCs in vitro from human embryonic brain tissue.
  52. [52]
    Derivation of Primitive Neural Stem Cells From Human-Induced ...
    Dec 15, 2019 · This study aimed to establish human primitive neural stem cells (pNSCs) from exogene-free hiPSCs without EB formation.
  53. [53]
    Human iPSC-derived neural stem cells displaying radial glia ...
    Nov 1, 2024 · In this study, we demonstrate that hiPSC-NSCs adopt a radial glia-associated signature, sharing key epigenetic and transcriptional ...
  54. [54]
    In vitro expansion of a multipotent population of human neural ...
    Human neural progenitor cells can be expanded in vitro using bFGF, LIF, and EGF, achieving a 10(7)-fold increase and remaining multipotent for at least 1 year.
  55. [55]
    Optimization of Matrigel-based culture for expansion of neural stem ...
    Neurosphere culture is most frequently used to expand NSCs in vitro. However, the neurosphere culture method has several limitations. One is that neurospheres ...
  56. [56]
    Comparison of different culture modes for long-term expansion of ...
    Neural stem cells can be cultured in two modes of suspension and monolayer in vitro. Reynolds and most other groups cultured NSCs as neurospheres in suspension ...
  57. [57]
    Insulin-Like Growth Factor-I Is Necessary for Neural Stem Cell ...
    Sep 15, 2001 · Murine embryonic striatal NSCs can be isolated in vitroby inducing their proliferation with either EGF (Reynolds et al., 1992) or FGF-2 (Qian et ...
  58. [58]
    Next-Generation Stem Cell Expansion Technologies - Insights.bio
    Recent studies demonstrate the benefit of using perfusion of culture medium as a way to improve stem cell expansion [20,21,53,55]. Perfusion consists in the ...
  59. [59]
    Essential Roles of Notch Signaling in Maintenance of Neural Stem ...
    Mar 3, 2010 · Activation of Notch signaling induces the expression of transcriptional repressor genes such as Hes1, leading to repression of proneural ...
  60. [60]
    Signaling mechanisms regulating adult neural stem cells and ...
    Hedgehog signaling in the subventricular zone is required for both the maintenance of stem cells and the migration of newborn neurons. J. Neurosci. 2007;27 ...
  61. [61]
    Directed migration of neural stem cells to sites of CNS injury ... - PNAS
    CXCR4 expression within germinal zones suggests that NSC homing after injury and migration during development may invoke similar mechanisms. Sign up for ...
  62. [62]
    Migration and Differentiation of Neural Stem Cells Diverted From the ...
    β1 integrin signaling promotes neuronal migration along vascular scaffolds in the post-stroke brain. EBioMedicine 16, 195–203. 10.1016/j.ebiom.2017.01.005 ...
  63. [63]
    Introduction to Neural Stem Cells
    Regardless of the methods by which neural stem cells are induced to divide in vitro, they must possess the key proper- ties of self-renewal and multipotency.
  64. [64]
    Bmi-1 cooperates with Foxg1 to maintain neural stem cell self ...
    Thus, NSCs extracted from the neurogenic niche and placed in culture can maintain multipotency and self-renewal when the level of Bmi-1 is increased. We found ...
  65. [65]
    Multipotent CNS Stem Cells Are Present in the Adult Mammalian ...
    Dec 1, 1996 · The spinal cord and the entire ventricular neuroaxis of the adult mammalian CNS contain multipotent stem cells, present at variable frequency and with unique ...
  66. [66]
    Querkopf is a key marker of self-renewal and multipotency of adult ...
    Querkopf is a key marker of self-renewal and multipotency of adult neural stem cells. Bilal N. Sheikh1,2, Mathew P. Dixon1, Tim Thomas1,2,*,` and Anne K ...
  67. [67]
    REST regulation of gene networks in adult neural stem cells - Nature
    Nov 7, 2016 · The mechanisms that control adult neural stem cell self-renewal are still largely unknown. Conditional knockout of REST (repressor element 1 ...
  68. [68]
    Lsh/HELLS regulates self-renewal/proliferation of neural stem ...
    Apr 25, 2017 · A neural stem cell is capable of self-renewal (by symmetric division) for extended periods of time. Asymmetric cell division allows neural stem ...
  69. [69]
    Epigenetic Regulations in Neural Stem Cells and Neurological ...
    In this review, we discussed recent advances of DNA modifications on the regulative mechanisms of neural stem cells. Among these epigenetic modifications, DNA 5 ...
  70. [70]
    Epigenetic regulation in adult neural stem cells - Frontiers
    Jan 30, 2024 · This minireview is primarily focused on epigenetic regulations of adult NSCs during adult neurogenesis, in conjunction with transcriptional regulation in these ...Introduction · DNA methylation · Histone post-translational... · Perspectives
  71. [71]
    An epigenetic barrier sets the timing of human neuronal maturation
    Jan 31, 2024 · Our findings reveal that the rate at which human neurons mature is set well before neurogenesis through the establishment of an epigenetic barrier in ...
  72. [72]
    Epigenetic regulation of adult neural stem cells: implications for ...
    Jun 25, 2014 · Epigenetic mechanisms are likely key players within these signaling networks, as DNA methylation, chromatin remodeling and small non-coding RNAs ...
  73. [73]
    Tet2-mediated epigenetic drive for astrocyte differentiation from ...
    Apr 29, 2020 · Our research not only revealed a brand-new function of Tet2 to promote NSC differentiation into astrocytes, but also a novel mechanism for Olig2 to inhibit ...
  74. [74]
    Impact of Metabolic Pathways and Epigenetics on Neural Stem Cells
    In this review, we summarize the major metabolic pathways and epigenetic modifications that play a role in NSC. We then focus on the notion that nutrients ...
  75. [75]
    Signaling in adult neurogenesis - PubMed
    Brain plasticity, achieved at the level of cell genesis, has an essential role in the maintenance of brain homeostasis.
  76. [76]
    Review Adult Mammalian Neural Stem Cells and Neurogenesis
    Oct 1, 2015 · It is now established that neural progenitor/stem cells (NSCs) reside in the adult mammalian brain and contribute to brain plasticity throughout life.
  77. [77]
    Neural stem cell pools in the vertebrate adult brain: Homeostasis ...
    Dec 9, 2020 · These results suggest that neural stem cell assemblies in the vertebrate brain behave as self-organized systems, such that the stem cells ...
  78. [78]
    Neural stem cell secretome: a secret key to unlocking the power of ...
    Jul 5, 2025 · In this review, we explore the role of adult NSCs and their secretome in sustaining brain function throughout adulthood and aging.
  79. [79]
    Brain Endothelial Cells Maintain Lactate Homeostasis and Control ...
    Nov 21, 2019 · Increased understanding of the functions of lactate has suggested a close relationship between lactate homeostasis and normal brain activity ...<|control11|><|separator|>
  80. [80]
    Aging of Neural Stem Cells and Vascular Dysfunction
    Oct 10, 2025 · Neural stem cells (NSCs) and cerebral vasculature are both critically involved in maintaining homeostasis, and their age-related dysfunction ...
  81. [81]
    Neural plasticity and adult neurogenesis: the deep biology perspective
    Adult neurogenesis and neural stem cells of the central nervous system in mammals. J Neurosci Res. 2002;69:745–749. doi: 10.1002/jnr.10378. [DOI] [PubMed] ...
  82. [82]
    Does the plasticity of neural stem cells and neurogenesis ... - Frontiers
    Sep 7, 2022 · Postnatal and adult neurogenesis takes place in the dentate gyrus of the hippocampus in the vast majority of mammals due to the persistence ...Abstract · Plasticity and hippocampal... · Neural stem cells as biosensors
  83. [83]
    α2-Chimaerin is essential for neural stem cell homeostasis in mouse ...
    Jun 17, 2019 · Adult neurogenesis in the mammalian hippocampus involves the lifelong generation of neurons, which integrate into preexisting neuronal network ( ...
  84. [84]
    Pregnancy-responsive pools of adult neural stem cells for transient ...
    Nov 23, 2023 · ... adult neurogenesis. Pregnancy-related interneurons are transient, integrating into layers in which fewer neurons are added under homeostasis ...<|separator|>
  85. [85]
    Sustaining Brain Youth by Neural Stem Cells - PubMed Central - NIH
    The recent findings showing that adult neurogenesis has a significant role in brain plasticity, learning and memory, and emotional behavior.
  86. [86]
    Neural stem cell heterogeneity in adult hippocampus
    Mar 7, 2025 · Adult neurogenesis is a unique cellular process of the ongoing generation of new neurons throughout life, which primarily occurs in the ...
  87. [87]
    Aging of the subventricular zone neural stem cell niche - PubMed
    Jan 26, 2011 · These findings demonstrate that aNSCs begin undergoing quiescence-associated changes between early and mid-adulthood in the mouse SVZ.
  88. [88]
    Mechanisms of enhanced quiescence in neural stem cell aging
    Aug 8, 2020 · This review highlights the mechanisms that regulate the NSC pool throughout adulthood and discusses how dysregulation of these processes may ...
  89. [89]
  90. [90]
    p57 controls adult neural stem cell quiescence and ... - PubMed
    Apr 3, 2013 · Adult NSCs constitute a relatively quiescent cell population but can be activated by extrinsic neurogenic stimuli.
  91. [91]
  92. [92]
    Age-related loss of neural stem cell O-GlcNAc promotes a glial fate ...
    Increased neural stem cell (NSC) quiescence is a major determinant of age-related regenerative decline in the adult hippocampus.
  93. [93]
  94. [94]
    CHD7 maintains neural stem cell quiescence and prevents ...
    NSCs are maintained in a state of reversible quiescence and the failure to maintain the quiescent state can result in the premature depletion of the stem cell ...
  95. [95]
    Stem Cell Aging? Blame It on the Niche - PubMed
    Mar 7, 2019 · Recently in Cell, Kalamakis et al. (2019) show that aged neural stem cells face greater barriers to exiting quiescence, imposed by the niche ...
  96. [96]
    CRISPR-Cas9 screens reveal regulators of ageing in neural stem cells
    Oct 2, 2024 · Ageing impairs the ability of neural stem cells (NSCs) to transition from quiescence to proliferation in the adult mammalian brain.
  97. [97]
    Age-related cognitive decline: Can neural stem cells help us?
    In this review we will discuss the systemic, cellular, and molecular changes induced by aging and affecting the neurogenic niche at the level of neural stem ...
  98. [98]
    Impaired adult neurogenesis is an early event in Alzheimer's ...
    Oct 7, 2019 · Our results demonstrate that SVZ neurogenesis is impaired already at a presymptomatic stage of AD and is caused by endogenously generated intracellular AβOs in ...
  99. [99]
    The relationship between adult hippocampal neurogenesis and ...
    Aug 21, 2024 · In this review we outline the regulation and function of AHN in healthy individuals, and highlight the relationship between AHN dysfunction and cognitive ...HUMAN ADULT... · AHN IS IMPAIRED IN AD... · PROPOSED MECHANISMS...
  100. [100]
    Depletion of adult neurogenesis exacerbates cognitive deficits in ...
    Sep 8, 2017 · This study suggests that deficits in adult neurogenesis may contribute to cognitive impairments, tau hyperphosphorylation in new neurons and compromised ...
  101. [101]
    Impact of alpha-synuclein pathology on adult neurogenesis:... - LWW
    Moreover, impairments in adult neurogenesis have been associated with many brain function disorders and diseases including PD and DLB.<|separator|>
  102. [102]
    Epigenomic enhancer annotation reveals a key role for NFIX in ...
    The majority of neural stem cells (NSCs) in the adult brain are quiescent, and this fraction increases with aging. Although signaling pathways that promote NSC ...
  103. [103]
    Neural Cells for Neurodegenerative Diseases in Clinical Trials - PMC
    Jul 24, 2023 · In this review, we present the list of cellular therapies for 7 neurodegenerative diseases including 33 trials that utilized neural stem ...Parkinson's Disease · Amyotrophic Lateral... · Multiple Sclerosis
  104. [104]
    Mechanisms of abnormal adult hippocampal neurogenesis in ...
    Feb 14, 2023 · Evidence suggests that AHN disorders caused by lipid metabolism disorders make patients more susceptible to AD. Monoacylglycerol lipase (Mgll), ...
  105. [105]
    Mechanisms of enhanced quiescence in neural stem cell aging - PMC
    Quiescent NSCs become activated in response to intrinsic or extrinsic cues and enter the cell cycle, which is the first functional step in neurogenesis.
  106. [106]
    ADULT NEURAL STEM CELLS: RESPONSE TO STROKE INJURY ...
    Both endogenous and grafted neural stem/progenitor cells are known to have the ability to migrate long distances to lesioned sites after brain injury and ...
  107. [107]
    Brain injury and neural stem cells - PMC - PubMed Central - NIH
    Stem cells can aid in decreasing lesion size and improving function following brain injury. Keywords: brain injury, brain trauma, infarction, ischemia, neural ...Ischemia/infarction · Regenerative Changes... · Activation, Migration, And...<|separator|>
  108. [108]
    Endogenous Repair Signaling after Brain Injury and Complementary ...
    As such, this review outlines the many factors of the injury microenvironment that mediate endogenous neural regeneration after TBI and the corresponding ...
  109. [109]
    The roles, mechanism, and mobilization strategy of endogenous ...
    The roles of eNSCs in the repair of brain injury include but are not limited to ameliorating cognitive function, improving learning and memory function, and ...
  110. [110]
    Neural Stem Cells as Potential Glioblastoma Cells of Origin - PMC
    Mar 29, 2023 · Recent data indicate that neural stem cells (NSCs) in the subventricular zone (SVZ) are the cells of origin of GBM.
  111. [111]
    Neural Stem Cells of the Subventricular Zone as the ... - Frontiers
    Direct in vivo evidence for tumor propagation by glioblastoma cancer stem cells. PLoS ONE. (2011) 6:e24807. doi: 10.1371/journal.pone.0024807. PubMed ...Abstract · Background · Frequent Gene Mutations in... · Therapeutic Implications
  112. [112]
    Sequential fate-switches in stem-like cells drive the tumorigenic ...
    Jan 4, 2021 · The most likely cell(s) of origin for GBM are neural stem ... Outer radial glia-like cancer stem cells contribute to heterogeneity of glioblastoma ...
  113. [113]
    The Role of Neurodevelopmental Pathways in Brain Tumors - PMC
    Disruptions to developmental cell signaling pathways and transcriptional cascades have been implicated in tumor initiation, maintenance and progression.
  114. [114]
    Notch, Neural Stem Cells, and Brain Tumors
    During neocortical development, Notch signaling inhibits neuronal differentiation and maintains the neural stem/progenitor cell pool to permit successive waves ...
  115. [115]
    Parallels Between Neural Stem Cells and Glioblastoma-Initiating Cells
    Jan 7, 2021 · This review highlights the similarities and differences between GSCs and SVZ NSCs in terms of their gene expression, regulatory molecular pathways, niche ...Abstract · Introduction · Subventricular Zone Neural... · Intrinsic Deregulations of...
  116. [116]
    Distant origin of glioblastoma recurrence: neural stem cells in the ...
    Mar 4, 2025 · Recent research has reported that neural stem cells (NSCs) in the subventricular zone (SVZ) harboring cancer-driving mutations serve as the ...
  117. [117]
    The adaptive transition of glioblastoma stem cells and its ... - Nature
    Mar 23, 2021 · Glioblastoma stem cells are considered to be responsible for tumor resistance in treating glioblastoma. Previous studies reported two subtypes, ...
  118. [118]
    Modeling neurological diseases using iPSC-derived neural cells
    These iPSC-derived neural cells provide valuable tools for studying neurological disease mechanisms, developing potential therapies, and deepening our ...
  119. [119]
    Modeling Alzheimer's disease with iPSC-derived brain cells - Nature
    Aug 7, 2019 · In addition to elevated Aβ levels and tau phosphorylation, iPSC-derived neurons from SAD patients can exhibit enlarged endosomes, mitochondrial ...
  120. [120]
    Limitations of the human iPSC-derived neuron model for early-onset ...
    We provide evidence that iPSC-derived neuron model systems, likely due to the loss of EOAD-associated epigenetic signatures arising from iPSC reprogramming, ...
  121. [121]
    The roles and applications of neural stem cells in spinal cord injury ...
    Aug 29, 2022 · Stem cell-based therapies are considered promising in attempts to repair injured spinal cords; such options include neural stem cells (NSCs).
  122. [122]
    The roles of neural stem cells in myelin regeneration and repair ...
    Jul 8, 2024 · Studies have shown their positive role in promoting tissue repair and functional recovery of the injured spinal cord [110, 111]. However, there ...
  123. [123]
    Long-term clinical and safety outcomes from a single-site phase 1 ...
    Dec 17, 2024 · We report the long-term results for a phase 1 study of neural stem cell transplantation for chronic spinal cord injury.
  124. [124]
    Long-term clinical and safety outcomes from a single-site phase 1 ...
    Dec 17, 2024 · The primary outcome of the trial was to test the feasibility and safety of human spinal cord-derived neural stem cell (NSI-566) transplantation ...
  125. [125]
    Clinical Outcomes from a Multi-Center Study of Human Neural Stem ...
    Mar 19, 2019 · At 1 year post-transplantation, there was no evidence of additional spinal cord damage, new lesions, or syrinx formation on magnetic resonance ( ...
  126. [126]
    Phase I clinical trial of intracerebroventricular transplantation of ...
    Nov 27, 2023 · No treatment-related deaths nor serious adverse events (AEs) were observed. All participants displayed stability of clinical and laboratory ...
  127. [127]
    Safety and efficacy evaluation of intracerebroventricular human ...
    May 9, 2025 · ICV transplantation of hNSCs emerges as a safe and promising strategy for ALS, demonstrating potential to delay motor decline and reduce spinal cord ...
  128. [128]
    Study Details | NCT01640067 | Human Neural Stem Cell ...
    Study Overview · assessment of the impact of human neural stem cells transplantation on the disability of ALS in a clinically significant and measurable way ...
  129. [129]
    Neural stem cell transplantation in patients with progressive multiple ...
    Jan 9, 2023 · Here we report the results of STEMS, the first phase 1 clinical trial evaluating the safety of hfNPC transplantation in patients with PMS.
  130. [130]
    How neural stem cell therapy promotes brain repair after stroke
    Jun 10, 2025 · This review explores the latest advancements in neural stem cell therapy for stroke, highlighting research insights in brain repair mechanisms.
  131. [131]
    Neural stem cells for Parkinson's disease management
    Jul 26, 2023 · Core tip: Neural stem cell (NSC) transplantation is a groundbreaking therapy with therapeutic effects for many neurodegenerative diseases.
  132. [132]
    Advances in genetically modified neural stem cell therapy for central ...
    Dec 18, 2024 · In this review, we will provide an overview of recent advances in the roles and mechanisms of NSCs genetically modified with various therapeutic genes.
  133. [133]
    Stem cell therapies for neurological disorders: current progress ...
    Jul 25, 2024 · Stem cell-based therapies have emerged as a promising approach for treating various neurological disorders by harnessing the regenerative potential of stem ...
  134. [134]
    Collagen-based biomaterials in neural injury repair
    Aug 10, 2025 · Collagen scaffolds provide an optimal microenvironment to direct endogenous neural stem/progenitor cell (NSPC) fate toward neuronal lineage, a ...<|separator|>
  135. [135]
    Co-culturing neural and bone mesenchymal stem cells in ...
    Dec 16, 2024 · Therefore, in this study, we propose co-culturing NSCs and BMSCs within low-modulus GelMA hydrogel scaffolds to promote regeneration following ...
  136. [136]
    Advances in smart hydrogels for nerve repair: A review focusing on ...
    Neural stem cells (NSCs) within such hydrogels, when exposed to 5–20 V electrical potential, show increased neuronal differentiation and neurite outgrowth [38].
  137. [137]
    Hydrogel oxygen reservoirs increase functional integration of neural ...
    Jan 28, 2023 · Altogether these results suggest that Mb functionalisation of the hydrogel resulted in enhanced integration and differentiation of neural stem ...
  138. [138]
    Electrospun Bio-Scaffolds for Mesenchymal Stem Cell-Mediated ...
    Sep 29, 2025 · Therefore, integrating stem cell therapy with advanced biomaterials offers a promising route to bridge the gap between neural injury and ...Missing: bioengineering | Show results with:bioengineering
  139. [139]
    A composite hydrogel improves the survival and differentiation of ...
    Jun 15, 2023 · A composite hydrogel improves the survival and differentiation of human iPSC-derived neural stem cells after ischemic stroke. Author links ...
  140. [140]
    Toward a New Generation of Bio-Scaffolds for Neural Tissue ... - MDPI
    This comprehensive review delves into the primary requisites, limitations, and prospective avenues for scaffold design in brain tissue engineering.
  141. [141]
    Enhancing Stem Cell-Based Therapeutic Potential by Combining ...
    Here, we summarized recent studies involving various types of stem cell-based tissue-engineering strategies for different degenerative diseases.
  142. [142]
    Conductive Nanocomposite Hydrogels for Neural Tissue Engineering
    Sep 8, 2025 · When embedded into hydrogels, these materials facilitate cellular interactions and have been shown to promote the differentiation of neural stem ...
  143. [143]
    Results from Phase I Clinical Trial with Intraspinal Injection of Neural ...
    May 18, 2019 · In this manuscript we present data from a phase I clinical trial on 18 ALS patients treated with human neural stem cells. Treatment has been ...
  144. [144]
    Phase I trial of hES cell-derived dopaminergic neurons for ... - Nature
    Apr 16, 2025 · The trial achieved its primary objectives of safety and tolerability one year after transplantation, with no adverse events related to the cell ...
  145. [145]
    Quiescence of Adult Mammalian Neural Stem Cells: A Highly ...
    Dec 4, 2019 · Quiescent neural stem cells present a low rate of metabolic activity and a high sensitivity to their local signaling environment, and they can ...
  146. [146]
    The Critical Balance Between Quiescence and Reactivation ... - MDPI
    During adulthood, most NSCs exist in a quiescent state which can be reactivated in response to metabolic and signalling changes, allowing for long-term ...2. Nsc Quiescent States... · 6. Hippo Signalling... · 8. Sumoylation And Nsc...
  147. [147]
    Increasing Neural Stem Cell Division Asymmetry and Quiescence ...
    Dec 18, 2018 · More frequent asymmetric stem cell divisions and longer quiescence are predicted. Altered stem cell behavior contributes to the age-related decline in ...
  148. [148]
    Notch3 signaling gates cell cycle entry and limits neural stem cell ...
    Aug 15, 2013 · We have previously shown that Notch signaling limits neural stem cell (NSC) proliferation in the adult zebrafish pallium. Combining ...
  149. [149]
    Strengths and limitations of the neurosphere culture system - PubMed
    This article discusses the pros and cons of the neurosphere culture system regarding its three major applications: as an assay for neural stem cells, as a model ...
  150. [150]
    Neural stem cells and neurospheres--re-evaluating the relationship
    Like all technologies, it is not without its limitations. In this article we will highlight several shortcomings of the assay related to its application and ...
  151. [151]
    Current progress in the derivation and therapeutic application of ...
    Oct 12, 2017 · A rosette-type, self-renewing human ES cell-derived neural stem cell with potential for in vitro instruction and synaptic integration. Proc ...<|separator|>
  152. [152]
    Insight Into the Mechanisms and the Challenges on Stem Cell ...
    Several concerns must be addressed in using the intracerebral or intraventricular method, including poor cell viability, invasiveness, immune rejection, and an ...
  153. [153]
    Successes and Hurdles in Stem Cells Application and Production for ...
    This review discusses the positive mechanisms triggered by cell transplantation into the brain, the limitations of adult brain plasticity that might interfere ...
  154. [154]
    Advancements and challenges in stem cell transplantation for ...
    Aug 30, 2024 · Pluripotent stem cells (PSCs) technology faces several challenges. There's a potential risk of tumorigenicity after transplantation, and ...
  155. [155]
    The benefits and risks of stem cell technology - PMC - PubMed Central
    Second, ES cells can potentially transform into cancerous tissue. Third, after transplantation, immunological mismatch can occur resulting in host rejection.
  156. [156]
    Ethical Issues in Stem Cell Research - PMC - NIH
    Apr 14, 2009 · Several ethical concerns come into play when a frozen embryo is donated, including informed consent from the woman or couple donating the embryo ...
  157. [157]
    Ethical considerations on the moral status of the embryo and embryo ...
    Sep 30, 2024 · The moral status attributed to a human embryo determines our ethical obligations towards it, sets boundaries on our actions involving embryos, ...
  158. [158]
    The moral status of human embryo‐like structures: potentiality ...
    Jul 27, 2020 · On this understanding, some countries attribute moral status to in vitro embryos on the basis of their advanced passive potentiality given that ...
  159. [159]
    Ethical questions in embryonic stem cell research - EuroGCT
    Ethical questions in embryonic stem cell research · 1. The embryo has full moral status from fertilisation onwards · 2. There is a cut-off point at 14 days after ...Missing: neural | Show results with:neural
  160. [160]
    Grounding Ethical Breakthroughs in Science: Embryonic Stem Cell ...
    In this paper, I argue that embryonic stem cell research is both ethically permissible on rational grounds and ethically essential due to its scientific, ...
  161. [161]
    From embryonic stem cells to iPS – an ethical perspective - PMC
    Ethical debate on development of stem cell research focused from the outset on issues of methods of acquiring these cells.
  162. [162]
    Stem Cell Use in Research and Health Care: A Bioethical Perspective
    May 24, 2023 · What makes iPSCs different from embryonic or donor-sourced stem cells? Primarily, iPSCs sidestep ethical issues of the fetus' right to life and ...
  163. [163]
    [PDF] Human Embryonic Stem Cell Research vs ... - Carroll Collected
    iPSCs have avoided the ethical issues raised by embryonic stem cell work. Concerns relevant to the majority of the research being conducted now on iPSCs ...
  164. [164]
    How can ethics relate to science? The case of stem cell research
    Nov 14, 2012 · Pluripotent stem cell research: embryo destruction versus non-embryonic sources. From the start embryonic stem cell research was marred by ...
  165. [165]
    Donor-Derived Brain Tumor Following Neural Stem Cell ...
    Feb 17, 2009 · Gideon Rechavi and colleagues describe the case of a boy with ataxia telangiectasia who developed a brain tumor after neural stem cell therapy.
  166. [166]
    Long-term Cultured Human Neural Stem Cells Undergo ...
    These results suggest that continuous expansion of neural stem cells in culture may lead to malignant spontaneous transformation. This phenomenon may be ...
  167. [167]
    The Postischemic Environment Differentially Impacts Teratoma or ...
    The Postischemic Environment Differentially Impacts Teratoma or Tumor Formation After Transplantation of Human Embryonic Stem Cell-Derived Neural Progenitors.
  168. [168]
    Risk factors in the development of stem cell therapy
    Mar 22, 2011 · Expansion of stem cells in medium supplemented with FBS has a potential risk of transmitting viral and prion diseases and causing immunological ...
  169. [169]
    Adverse events of cell therapy clinical trials in human chronic spinal ...
    The most common adverse events were transient backache and meningism (90%) and cord malacia (80%). The cell therapy method in which the treatment was associated ...
  170. [170]
    After 25 years of hype, embryonic stem cells still don't cure for anything
    Aug 9, 2023 · After 25 years of hype, embryonic stem cells are still waiting for their moment. Research roadblocks and political debates have delayed progress.
  171. [171]
    Stem cells: Don't believe the hype - Nature
    Apr 11, 2012 · Stem cells: Don't believe the hype ... Researchers are still a long way from using stem cells to halt the decline caused by multiple sclerosis and ...Missing: overhyped | Show results with:overhyped
  172. [172]
    Charlatans threaten stem cell research with unproven cures, say ...
    Oct 4, 2017 · The credibility of stem cell research is at risk because of charlatans and dodgy clinics peddling unproven cures for diseases, according to a group of eminent ...
  173. [173]
    Are New Neurons Formed in the Brains of Adult Mammals? - Science
    Are New Neurons Formed in the Brains of Adult Mammals? Joseph AltmanAuthors Info & Affiliations. Science. 30 Mar 1962. Vol 135, Issue 3509. pp. 1127-1128. DOI ...
  174. [174]
    Are new neurons formed in the brains of adult mammals? - PubMed
    Some neurons and neuroblasts showed labeling, suggesting the possibility of proliferation of neurons in adult rats.
  175. [175]
    the Groundbreaking Research of Joseph Altman and Gopal Das
    Postnatal neurogenesis has been documented in the cerebellum, olfactory bulb, hippocampus, striatum, substantia nigra, hypothalamus, and amygdala.
  176. [176]
    ADULT NEUROGENESIS IN HUMANS: A Review of Basic Concepts ...
    The authors studied 23 individuals aged 0.2 to 59 years and observed a marked decline in neurogenesis in SVZ and SGZ in early childhood and then after three ...
  177. [177]
    Joseph Altman and Adult Neurogenesis: The Dogma of
    There were three developments that led to a vindication of Altman's pioneering work and to general acceptance that new neurons are added to the adult mammalian ...
  178. [178]
    Neurogenesis: An Overview - BrainFacts
    Jul 21, 2016 · Pioneering research by Joseph Altman in 1962 suggested otherwise. Altman injected adult rats with a radioactive molecule that incorporates into ...
  179. [179]
    The History of Discovery of Adult Neurogenesis - PubMed
    During the 1970s, Kaplan confirmed neurogenesis in the adult mammalian neocortex and also performed some of the first neurogenesis studies on primates.Missing: early | Show results with:early
  180. [180]
    The History of Discovery of Adult Neurogenesis - Wiley Online Library
    Aug 5, 2019 · During the 1970s, Kaplan confirmed neurogenesis in the adult mammalian neocortex and also performed some of the first neurogenesis studies on primates.
  181. [181]
    Generation of Neurons and Astrocytes from Isolated Cells ... - Science
    In this study, cells isolated from the striatum of the adult mouse brain were induced to proliferate in vitro by epidermal growth factor.
  182. [182]
    Generation of neurons and astrocytes from isolated cells of the adult ...
    In this study, cells isolated from the striatum of the adult mouse brain were induced to proliferate in vitro by epidermal growth factor.
  183. [183]
    Neural stem cell isolation and characterization - PubMed
    ... Reynolds and Weiss demonstrated the presence of neural stem cells in both the adult (Reynolds and Weiss, 1992) and embryonic mouse brain (Reynolds et al., 1992) ...
  184. [184]
    Isolation of Neural Stem Cells from Whole Brain Tissues of Adult Mice
    Two neurosphere assays (Reynolds & Weiss, 1992; Reynolds & Weiss, 1996), developed in the early 1990s, have remained the gold standards for in vitro neural stem ...
  185. [185]
    Induced pluripotent stem cell technology: a decade of progress - PMC
    Human iPSCs have been widely used for disease modeling, drug discovery, and cell therapy development. Novel pathological mechanisms have been elucidated, new ...
  186. [186]
    Neural Stem Cells and Methods for Their Generation From Induced ...
    Neural stem cells is the term commonly applied to multipotent undifferentiated cells, capable of generating all types of neurons, oligodendrocytes, and ...Missing: definition | Show results with:definition