Fact-checked by Grok 2 weeks ago

Progenitor cell

Progenitor cells are biological cells capable of undergoing and differentiating into one or more specialized types within a particular or , acting as committed precursors that bridge the gap between multipotent stem cells and fully differentiated mature cells. Unlike stem cells, which exhibit extensive self-renewal and broad differentiation potential, progenitor cells typically possess limited proliferative capacity and more restricted potential, often being unipotent or oligopotent, meaning they can give rise to a limited number of cell types. This distinction arises from their more advanced state of commitment during , where they respond to specific signals to expand populations rapidly for maintenance or repair. Progenitor cells are integral to embryonic development, postnatal growth, and adult tissue homeostasis across various organ systems, including the hematopoietic system, skeletal tissues, and vascular endothelium. In hematopoiesis, for instance, they proliferate to generate mature blood cells such as erythrocytes and leukocytes, ensuring continuous replenishment of the blood supply. Similarly, in skeletal biology, progenitor cells located in niches like the bone marrow or periosteum contribute to bone formation, remodeling, and fracture healing through processes like chondrocyte-to-osteoblast transdifferentiation. Their activity is tightly regulated by environmental cues, including growth factors and redox signals, which influence proliferation, survival, and differentiation to prevent dysregulation that could lead to diseases such as leukemia or impaired wound healing. The identification and study of progenitor cells have advanced through techniques like lineage tracing, transplantation assays, and marker expression analysis (e.g., , , or PDGFRα), revealing their heterogeneity and context-specific functions. These cells hold significant therapeutic potential in , where harnessing their differentiation capacity could aid in treating conditions like , neurodegeneration, and tissue injury, though challenges remain in isolating pure populations and ensuring safe integration.

Definition and Properties

Definition

Progenitor cells represent a class of biological intermediates in cellular hierarchies, capable of undergoing and into specific, often specialized cell types within a defined lineage, while exhibiting limited self-renewal capacity compared to stem cells. These cells bridge the gap between more versatile stem cells and fully differentiated cells, serving as transient populations that commit to narrower developmental paths under appropriate conditions. The term "progenitor cell" traces its etymological roots to Latin words meaning "to beget forth," and it emerged in biological literature during early 20th-century embryological investigations into and fate determination. In modern usage, particularly since the mid-20th century advancements in —such as the identification of colony-forming units by Till and McCulloch in —the concept has evolved to emphasize oligopotency, defined as the potential to generate a limited set of related types rather than broad lineages. This refinement distinguishes progenitors from earlier, more generalized notions of precursor cells in . Progenitor cells are categorized by their potency, with unipotent progenitors restricted to producing a single mature cell type and oligopotent progenitors able to form a few closely related types within a or , in contrast to the multipotent capabilities of upstream cells. The differentiation trajectory of progenitor cells is profoundly shaped by their cellular niche—the localized microenvironment comprising supportive cells, , and signaling molecules—which provides biochemical and biophysical cues that modulate potency and direct lineage commitment. These environmental influences, including growth factors and adhesion interactions, can either maintain proliferative states or trigger irreversible , ensuring precise regulation of .

Key Properties

Progenitor cells exhibit a limited proliferative capacity, undergoing a limited number of divisions before entering , a primarily driven by progressive telomere shortening due to the absence or low levels of activity. Unlike pluripotent cells, which maintain telomere length through robust expression, progenitor cells lack sufficient to counteract replicative stress, leading to eventual chromosomal instability and arrest. This finite division potential ensures controlled expansion within specific lineages while preventing indefinite that could promote tumorigenesis. A defining feature of progenitor cells is their oligopotent differentiation potential, characterized by early commitment to a specific , restricting their ability to generate diverse types beyond a few related subtypes. This commitment often occurs through , where a progenitor divides to produce one that remains a progenitor and another that progresses toward , thereby amplifying the while preserving the progenitor pool. Such divisions are regulated by lineage-specific transcription factors and signaling pathways that reinforce the oligopotent state, ensuring precise without reverting to multipotency. Certain progenitor cells, particularly circulating types such as endothelial or hematopoietic variants, demonstrate notable mobility and migration capabilities, guided by responses to gradients that direct them to sites of or demand. These cells express receptors like , which bind ligands such as SDF-1/, facilitating and homing to hypoxic or inflamed environments where they contribute to repair processes. This migratory behavior is tightly controlled to avoid ectopic accumulation, with gradients established by damaged s providing spatiotemporal cues for precise localization. Identification of progenitor cells relies heavily on the expression of specific surface markers, which vary by lineage but commonly include proteins like for hematopoietic and endothelial progenitors. These markers serve as diagnostic tools in and isolation protocols, enabling enrichment of progenitor populations from heterogeneous tissues based on their adhesive and signaling properties. Lineage-specific variations, such as in neural contexts or PDGFRα in mesenchymal ones, further refine identification, reflecting the cells' committed developmental stage. Progenitor cell function is also modulated by , with sensitivity to playing a key role in toggling between quiescence and activation states. Low levels of (ROS) maintain quiescence by suppressing metabolic activity and preserving long-term viability, while moderate ROS elevation—triggered by environmental cues—promotes activation, proliferation, and differentiation through -sensitive pathways like Nrf2 signaling. Excessive , however, induces or , underscoring the delicate balance that protects progenitor integrity during demands.

Versus Stem Cells

Progenitor cells and occupy distinct positions within cellular hierarchies, with stem cells serving as the more versatile upstream regulators capable of long-term tissue maintenance. A primary distinction lies in their self-renewal capacities: stem cells possess the ability for indefinite or long-term self-renewal through symmetric cell divisions that produce identical daughter cells, allowing sustained population expansion, whereas progenitor cells exhibit limited self-renewal, typically undergoing a finite number of asymmetric divisions that generate one self-renewing daughter and one differentiating cell, ultimately leading to proliferative exhaustion after several cycles. In terms of potency, stem cells demonstrate broad developmental potential, ranging from totipotency in early embryonic stages—capable of forming all types including extra-embryonic s—to multipotency in adult contexts, where they can differentiate into multiple s within a or system. cells, by contrast, are post-commitment intermediates with restricted potency, classified as oligopotent (able to form a few closely related s within a specific ) or unipotent (limited to a single ), reflecting their role in amplifying committed pathways rather than initiating broad decisions. Progenitor cells also display greater dependency on niche-derived signals compared to stem cells, lacking the latter's relative ; they require instructive cues from stem cell niches or associated stromal elements, such as chemokine signaling via or lineage-specific factors, to maintain and direct , without which they rapidly exhaust or default to terminal fates. Both cell types share evolutionary conserved regulatory networks involving transcription factors like Oct4 and , which orchestrate early pluripotency in stem cells, but progenitors downregulate these pluripotency genes shortly after , shifting expression toward tissue-specific programs that limit reversibility. Experimentally, these differences are delineated through functional assays: stem cells are identified by their capacity for long-term repopulation in transplantation models, such as serial engraftment in immunodeficient mice where they sustain multilineage hematopoiesis for months, demonstrating enduring self-renewal. Progenitor cells, however, are characterized by short-term assays, including cultures that reveal transient proliferative output and restricted lineage commitment over days to weeks, without sustained reconstitution.

Versus Differentiated Cells

Progenitor cells occupy a transitional state, retaining partial plasticity that enables them to undergo further lineage-specific maturation in response to environmental cues, in contrast to terminally differentiated cells, which exhibit fixed profiles and a permanent exit from the . This plasticity allows progenitors to respond adaptively during or , whereas differentiated cells, such as mature neurons or erythrocytes, are post-mitotic and committed to specialized functions without the capacity for or fate reversal under normal conditions. In terms of , progenitor cells display intermediate levels of lineage-specific transcription factors, reflecting their oligopotent to restricted fates, while fully differentiated cells show complete or repression of these factors to support . For instance, in erythroid , early erythroid progenitors (BFU-E) express low levels of alongside , which increases to high levels in late progenitors (CFU-E and proerythroblasts) to drive synthesis, before declining sharply in maturing erythroblasts and becoming minimal in anucleate mature erythrocytes. This graded expression in progenitors facilitates progressive , unlike the stable, fully committed profiles in differentiated cells that lock in effector functions such as oxygen transport in erythrocytes. Functionally, progenitor cells serve as lineage amplifiers, undergoing limited divisions to generate multiple differentiated daughter cells that expand tissue output during development or repair, whereas differentiated cells execute specialized tasks without further proliferation. Although rare, progenitors can undergo dedifferentiation to a more stem-like state under stress conditions, such as injury or inflammation, enabling tissue regeneration; this reversibility is largely absent in terminally differentiated cells, which lack the molecular machinery for fate reversion. For example, in the airway epithelium, committed epithelial cells (such as club and ciliated cells) can dedifferentiate into basal stem cells following damage to basal stem cells, restoring proliferative capacity. Aging disproportionately affects cells, which accumulate more rapidly due to their proliferative history and exposure to cumulative divisions, thereby accelerating dysfunction compared to long-lived, post-mitotic differentiated cells. In progressive , senescent cells secrete , inhibiting their own maturation and contributing to remyelination failure, while differentiated remain functional but limited by upstream exhaustion. This -specific drives broader age-related decline in regenerative potential across s.

Biological Functions

In Embryonic Development

During embryonic development, progenitor cells play a crucial role in the expansion phase by amplifying the output of pluripotent stem cells through rapid proliferative divisions, thereby populating the three primary germ layers—, , and . In the , for instance, naïve ectodermal cells transition into neural progenitors following inhibition of () signaling, enabling the formation of that gives rise to the . For , progenitors expand to form structures like the somites, which differentiate into muscle, , and cells under signals such as Wnt and . In the , progenitors contribute to the gut tube and associated organs, responding to nodal signaling for anterior-posterior patterning. This proliferative amplification occurs in the epiblast during , where progenitors derive from pluripotent cells and expand to form the foundational layers of the embryo, ensuring sufficient cell numbers for subsequent . Neuromesodermal progenitors (NMPs), co-expressing neural () and mesodermal (T/Bra) markers, further contribute to this process by proliferating in the caudal under 8 (FGF8) influence before differentiating. Progenitor cells also facilitate spatial patterning in the embryo, organizing tissues along axes such as dorsal-ventral through responses to morphogen . Sonic hedgehog (Shh) establishes a ventral-to-dorsal that instructs progenitors to adopt ventral identities, while Wnt signaling antagonizes Shh to restrict progenitors to dorsal regions, thereby defining boundaries in the developing . These provide positional information, allowing progenitors to interpret signaling strength and differentiate accordingly, as seen in the precise allocation of progenitor domains along the dorsal-ventral axis during neural development. The temporal progression of progenitor activity occurs in sequential waves, generating diverse cell types in a timed manner to support structured embryogenesis. Early waves of progenitors, such as radial glia in the , initiate by producing initial neuronal populations, followed by later waves that yield intermediate progenitors and glial cells, ensuring orderly layering of tissues. This progression is regulated by intrinsic timers and extrinsic cues, prolonging or accelerating cell cycles as needed to match developmental stages. Evolutionary conservation of progenitor mechanisms is evident across species, with neuroblasts serving as a model for understanding human neural progenitor dynamics. In flies, neuroblasts undergo asymmetric divisions to self-renew and produce progenitors, mirroring mammalian processes and providing insights into the timing of human cortical neurogenesis, where similar sequential progenitor waves build structures. Disruptions in progenitor regulation, particularly mutations in the , can lead to severe congenital defects such as , where failure to properly pattern the results in midline malformations. interacts with ligands like Delta-like 1 (Dll1) to maintain progenitor pools, and its dysregulation impairs the balance between and during early neural development. Similarly, loss-of-function mutations in -linked genes like PRDM15 disrupt patterning by altering interactions with Wnt/planar signaling, contributing to phenotypes observed in affected individuals.

In Adult Tissue Maintenance and Repair

In adult tissues, progenitor cells contribute to homeostatic turnover by undergoing low-level activation to replenish short-lived differentiated cells, ensuring steady-state tissue integrity. For instance, in the skin epidermis, progenitor cells in the basal layer proliferate and differentiate to replace the outer layer, which renews approximately every 28 days to maintain . This process involves balanced self-renewal and differentiation, preventing excessive while compensating for constant cell loss due to . Upon tissue injury, progenitor cells are recruited to the damage site and exhibit enhanced to facilitate repair and restore structural . This response is mediated by pro-inflammatory cytokines such as tumor factor-α (TNF-α), which activates signaling pathways in progenitors like alveolar epithelial type 2 cells, promoting their expansion without compromising differentiation potential. In models of and cardiac injury, TNF-α-driven recruitment helps bridge the gap between inflammation and regeneration, limiting scar formation. Adult progenitor cells reside in specialized niches that regulate their quiescence to preserve long-term tissue maintenance. In the stroma, transforming growth factor-β (TGF-β) signaling from niche components induces quiescence in hematopoietic progenitors by inhibiting progression and formation necessary for . This niche-mediated control ensures progenitors remain dormant under steady-state conditions, activating only as needed to avoid exhaustion. With advancing age, progenitor cell populations decline in number and functionality, exacerbating conditions like through impaired . Studies indicate that osteoprogenitors significantly decrease in aged mice, with approximately one third retained, accumulating senescence markers such as p21 and DNA damage, which hinder osteogenic differentiation. Recent research links this to age-related bone loss, highlighting elevated (SASP) factors that further disrupt niche and contribute to fragility fractures. Progenitor cells engage in bidirectional with immune cells during repair, where they actively modulate to promote and avert . Mesenchymal progenitors, for example, secrete anti-inflammatory factors that dampen macrophage-driven responses, shifting from pro-inflammatory to pro-resolving states and preserving balance. This interaction, evident in models, prevents excessive deposition by coordinating immune clearance with progenitor .

Examples in Specific Tissues

Hematopoietic Progenitor Cells

Hematopoietic progenitor cells (HPCs) form a pivotal intermediate stage in the production , bridging long-term repopulating hematopoietic stem cells (HSCs) and fully committed precursors. HSCs, phenotypically defined as Lin⁻⁻CD90⁺CD45RA⁻, possess multipotent self-renewal capacity and differentiate into short-term HSCs and multipotent progenitors (MPPs), which lose self-renewal but retain broad lineage potential. MPPs subsequently branch into common myeloid progenitors (CMPs) and common lymphoid progenitors (CLPs), both marked by ⁺ expression and exhibiting oligopotent properties that restrict them to myeloid or lymphoid lineages, respectively. Differentiation from CMPs proceeds along balanced pathways to either megakaryocyte-erythroid progenitors (MEPs) or granulocyte-macrophage progenitors (GMPs), orchestrated by key transcription factors such as PU.1 (encoded by Sfpi1) and C/EBPα (encoded by Cebpa). Intermediate PU.1 levels favor MEP commitment toward platelet and red blood cell lineages, whereas high PU.1 expression, often in concert with C/EBPα, drives GMPs toward granulocytes and monocytes, ensuring balanced myeloid output. These factors act as molecular switches, with PU.1 dosage determining lineage bias and C/EBPα promoting granulocytic maturation by activating downstream targets like Gfi1. Within the bone marrow, HPCs are maintained in distinct niches that regulate their and . The endosteal niche, adjacent to bone-lining osteoblasts, fosters quiescence and homing through low-oxygen environments and gradients, while the perivascular niche, enriched around sinusoidal and leptin receptor-expressing stromal cells, promotes active . Essential cytokines include (SCF), secreted by perivascular mesenchymal cells to sustain CMP and GMP expansion via KIT receptor signaling, and thrombopoietin (TPO), which binds MPL on MEPs and GMPs to enhance and megakaryopoiesis while indirectly supporting HSC quiescence in the niche. Clinically, HPC activity is assessed through (CFU) assays, where isolated CD34⁺ cells are cultured in methylcellulose-based media supplemented with lineage-specific cytokines to enumerate multilineage colonies, quantifying proliferative potential and lineage bias as a functional readout of engraftment capacity. In , a DNA repair disorder caused by mutations in FA pathway genes like FANCA, HPC defects manifest as reduced clonogenic efficiency and hypersensitivity to DNA damage, triggering /p21-mediated arrest that impairs progenitor expansion and leads to progressive failure. As of 2025, single-cell RNA sequencing has unveiled lung-resident HPC populations, including CMP-like cells within alveolar and perivascular compartments, that seed local hematopoiesis and contribute to systemic immune surveillance by generating myeloid effectors in response to respiratory challenges.

Neural Progenitor Cells

Neural progenitor cells (NPCs) are specialized precursors critical for the and plasticity of the , primarily generating neurons and during embryogenesis and adulthood. In embryonic stages, radial glial cells serve as primary NPCs, characterized by their bipolar morphology with processes extending from the ventricular surface to the pial surface; these cells transition into intermediate progenitor cells (IPCs) that populate the (SVZ) and undergo further divisions to amplify neuronal output. In the adult brain, NPCs persist in the SVZ lining the , where type B cells act as quiescent stem-like progenitors that generate transit-amplifying type C cells, ultimately producing neuroblasts (type A cells) that migrate to the to form new . Identification of NPCs relies on specific molecular markers and functional assays. Nestin and are key intermediate filament and markers, respectively, expressed in both embryonic radial and adult SVZ progenitors to maintain their undifferentiated state and self-renewal capacity. is tracked using bromodeoxyuridine (BrdU) incorporation, which labels dividing cells and allows monitoring of their progression toward . Signaling pathways tightly regulate NPC fate: the / pathway promotes self-renewal by inhibiting in adjacent cells through , while bone morphogenetic protein (BMP) signaling, via receptors like BMPR-IA, drives exit from quiescence and promotes into neurons or by activating downstream pathways such as ERK. Adult neurogenesis, mediated by these NPCs, is restricted to the SVZ and hippocampal subgranular zone (SGZ), contributing to brain and repair. In humans, estimates suggest this process yields approximately 700 new neurons daily in the young adult , though the extent of remains a topic of scientific debate with varying rates reported across studies, with SVZ progenitors similarly supporting olfactory bulb renewal, though rates decline with age. In pathological contexts like (AD), NPC pools exhibit exhaustion, marked by reduced proliferation and Nestin expression, impairing and exacerbating cognitive decline. Recent research highlights epigenetic rejuvenation strategies, such as targeting quiescence regulators via screens, to restore NPC function in aging brains. In contexts, similar approaches show promise in countering progenitor exhaustion.

Endothelial and Mesenchymal Progenitor Cells

Endothelial progenitor cells (EPCs) are bone marrow-derived cells characterized by the expression of and vascular endothelial growth factor receptor 2 (VEGFR2), which enable their identification and isolation from peripheral or bone marrow mononuclear cells. These progenitors possess the capacity to differentiate into mature endothelial cells that form the lining of vessels, contributing to and vascular repair. In response to tissue injury or ischemia, EPCs are mobilized from the bone marrow niche into the systemic circulation primarily through stimulation by (VEGF), which promotes their homing to sites of vascular damage. Mesenchymal progenitor cells (MPCs), also known as multipotent mesenchymal stromal cells, are found in the stroma and , exhibiting self-renewal and multilineage differentiation potential. These cells can differentiate into osteoblasts, chondrocytes, and adipocytes, key components of the skeletal and connective tissues, under the regulation of transcription factors such as for osteogenic pathways and for chondrogenic commitment. This differentiation is influenced by environmental cues like growth factors and components, supporting tissue and regeneration in mesenchymal lineages. Isolation of EPCs typically involves culturing mononuclear cells on Matrigel-coated surfaces, where they form tube-like structures indicative of endothelial , allowing for enrichment without extensive . In contrast, MPCs are commonly isolated through their inherent adherence to plastic culture flasks, followed by expansion in serum-supplemented media, which selects for their fibroblastoid and multipotency. These methods facilitate the procurement of viable populations for both and therapeutic applications. Recent advances as of 2025 include the development of animal-free culture systems for placenta-derived EPCs, which enable efficient expansion and without xenogeneic components, offering a scalable source for vascular therapies. Similarly, genetic modifications to render MPCs senescence-resistant, such as overexpression of , have demonstrated the ability to slow aging processes in nonhuman primates by improving multi-organ function and reducing age-related decline. EPCs interact closely with , which often derive from MPCs, to ensure vessel stability during ; pericytes provide structural support and modulate endothelial barrier function through cell-cell contacts and . This crosstalk, involving factors like VEGF and , is essential for maturing nascent vessels and preventing leakage in regenerative contexts.

Cerebral Cortex Development

Progenitor Roles in Neurogenesis

In the developing , progenitor cells in the ventricular zone (VZ) primarily consist of radial glia, which serve as the main neurogenic cells responsible for generating neurons. These cells extend a long basal process that contacts the pial surface and an apical process that anchors to the ventricular surface, functioning both as s and scaffolds for neuronal migration. Radial glia undergo asymmetric divisions to self-renew and produce neuronal precursors, with their nuclei exhibiting interkinetic nuclear migration (INM) during the : S-phase occurs in the basal VZ, and at the apical surface, ensuring coordinated and maintaining epithelial integrity. Complementing radial glia, intermediate progenitors—also known as basal progenitors—emerge from the VZ and migrate to the (SVZ), where they amplify output through transit-amplifying divisions. These progenitors, marked by expression of Tbr2 (Eomesodermin), undergo one or more symmetric divisions to increase the pool of neuronal precursors before differentiating, thereby enhancing the efficiency of cortical production without depleting the reservoir. This amplification is crucial for generating the diverse neuronal populations required for cortical layering. Progenitor activity follows a temporal sequence that establishes the six-layered , with deep-layer neurons (layers V and VI) generated first from E11.5 to E15.5 in mice, followed by superficial-layer neurons (layers II-IV) from E15.5 onward. This inside-out pattern arises from progressive changes in division modes and fate decisions, where early divisions favor deep-layer neurons, and later ones produce upper-layer and pyramidal cells, culminating in the mature laminar organization. In humans, phases are markedly prolonged compared to , extending over months to support the larger and . This is facilitated by the expansion of the outer (oSVZ), which harbors abundant basal progenitors and contributes to increased numbers and cortical folding through enhanced gliogenesis and progenitor diversity. Recent studies (as of 2025) highlight the role of outer SVZ basal progenitors in driving cortical folding through increased neuron production and migration patterns. Key regulatory genes orchestrate these processes: Pax6 promotes and maintains radial glia identity in the VZ, while Tbr2 drives the transition to intermediate fate in the SVZ, ensuring sequential differentiation. Mutations in these genes, such as Pax6 loss, disrupt and , highlighting their essential roles.

Stages and Mechanisms

In human development, the embryonic timeline of activity marks a critical transition around embryonic day 40 (E40), approximately 6 weeks post-fertilization. Prior to E40, early neural primarily undergo symmetric divisions to expand the pool, contributing to foundational telencephalic structures before specialized cortical begins. Post-E40, shift toward producing neurons that organize into radial cortical columns, initiating the expansion of the six-layered through symmetric and asymmetric divisions in the ventricular zone. Cell cycle dynamics in cortical progenitors are finely tuned to support rapid neurogenesis, with a notably shortened G1 phase enabling quicker progression through the cell cycle compared to later stages. This shortening, typically reducing G1 duration by up to 25% during peak proliferation, promotes the amplification of progenitor pools and timely neuronal output. The process is primarily regulated by cyclin D1 (CCND1), which complexes with CDK4/6 to drive G1/S transition; overexpression of cyclin D1 further accelerates this phase, delaying differentiation while expanding basal progenitors. Apoptosis plays a pivotal role in refining cortical layers by eliminating excess progenitors, ensuring balanced neurogenesis without overproliferation. Programmed cell death in these progenitors is mediated through caspase-3 activation, triggered by the apoptosome complex involving cytochrome c release and Apaf-1, which cleaves downstream substrates to execute cell dismantling. This regulated apoptosis, peaking during mid-gestation, helps sculpt layer-specific neuronal populations by removing approximately 30-50% of generated cells, preventing hyperplasia. Species differences in progenitor behavior underscore evolutionary adaptations for brain size, with human cortical progenitors exhibiting prolonged persistence compared to rodents, where neurogenesis largely concludes by birth. In humans, progenitors remain active through the second trimester and into early postnatal periods, facilitating the production of over 20 billion neurons and enabling a threefold larger cortex relative to body size. This extended activity supports gyrification, the folding of the cortex into gyri and sulci; recent studies indicate that gyrification initiates as early as gestational week 20, driven by tangential expansion of progenitors and differential neuronal adhesion. Disruptions to progenitor pools can severely impair cortical development, as exemplified by (ZIKV) infection, which preferentially targets radial glial s in the ventricular zone. ZIKV entry via receptors induces premature differentiation and , depleting the progenitor pool by up to 40% and leading to reduced cortical thickness characteristic of . This depletion mimics congenital Zika syndrome, where infected progenitors show attenuated growth and disrupted progression, resulting in fewer upper-layer neurons.

Research and Applications

Current Research Advances

Recent advances in single-cell omics technologies have elucidated the heterogeneity of progenitor cells, particularly through single-cell RNA sequencing (scRNA-seq) that reveals distinct transcriptional profiles. In the adult human lung, scRNA-seq analysis of CD34+ hematopoietic stem and progenitor cells (HSPCs) has identified unique gene signatures characterized by elevated expression of genes associated with immune activation, megakaryocyte/platelet differentiation, and inflammatory responses, such as CEBPB, SOD2, and PLCG2, compared to bone marrow counterparts. These findings, reported in a 2025 study from the American Society of Hematology, highlight how pulmonary HSPCs maintain a poised state for rapid immune and lineage-specific responses, expanding understanding of extramedullary hematopoiesis. Investigations into trained immunity have positioned hematopoietic progenitors as key reservoirs for epigenetic , enabling enhanced responses to subsequent challenges. A 2025 eLife review synthesizes evidence that HSPCs encode innate immune through durable modifications like and H3K27ac, as well as changes, persisting for months to years after initial stimuli such as BCG vaccination. This "central trained immunity" involves selective expansion of HSPC subpopulations, metabolic reprogramming, and altered lineage biases, influencing and transplant outcomes in both murine models and studies. Aging research on progenitor cells emphasizes the roles of imbalance and pathways, independent of attrition in some contexts. Increased during aging impairs (HSC) regenerative capacity and promotes myeloid skewing, as detailed in a 2025 Blood commentary on countering damage to rejuvenate hematopoiesis. -independent mechanisms, driven by persistent DNA damage from , activate pathways like and SASP in progenitors, contributing to exhaustion without requiring critical telomere shortening. Organoid models have advanced from early 2010s protocols to sophisticated 3D cultures that derive and study tissue-specific progenitors for disease modeling. These self-organizing structures, generated from pluripotent stem cells, recapitulate progenitor niches and enable high-throughput analysis of developmental disorders and cancers, as reviewed in a 2025 Cellular and Molecular Life Sciences article on organoid applications in regenerative medicine. For instance, neural organoids produce ventricular zone-like progenitors to model gliogenesis defects in neurodegenerative diseases, providing insights into spatial and temporal dynamics previously inaccessible in 2D systems. Links between progenitor biology and cancer have intensified focus on progenitor-like cancer stem cells (CSCs) in gliomas, where Notch signaling sustains their self-renewal and therapy resistance. A 2025 Nature Communications study demonstrates that classical glioma stem cells, exhibiting astrocyte-like and neural progenitor features, rely on the MEOX2-NOTCH axis for proliferation, with Notch inhibitors like RG-4733 effectively disrupting tumor progression when combined with NF-κB targeting. This approach highlights the potential of pathway-specific interventions to eliminate heterogeneous CSC populations driving glioma recurrence.

Therapeutic Potential

Progenitor cells hold significant promise in , particularly through targeted infusions to repair damaged tissues. Endothelial progenitor cells (EPCs) have been investigated for treating ischemia, such as post-myocardial (MI) vascular repair, where their infusion promotes and improves endothelial function in clinical trials for . Similarly, mesenchymal progenitor cells (MPCs), often derived from , demonstrate efficacy when administered via intra-articular injection for , reducing pain and enhancing joint function by supporting cartilage regeneration without forming teratomas. In neurological applications, neural progenitor cells derived from induced pluripotent stem cells (iPSCs) are advancing therapies. Recent 2025 clinical trials have shown that transplanting these cells promotes brain repair, , and long-term functional by modulating pathways and reducing in the subacute post-stroke. Hematopoietic cells also offer immunomodulatory potential, serving as reservoirs for trained immunity that enhance innate immune responses against infections or mitigate ; for instance, their epigenetic supports efficacy and anti-inflammatory effects in autoimmune conditions. Despite these advances, progenitor cell therapies face key challenges, including the of tumorigenicity from uncontrolled and immune rejection in allogeneic settings, which can trigger graft failure or host-versus-graft responses. Solutions like CRISPR-Cas9 editing address these by enhancing safety, such as creating senescence-resistant MPCs that withstand aging-related stress and reduce rejection without , as demonstrated in 2025 primate studies showing slowed aging phenotypes. Regulatory progress supports clinical translation, with the FDA having approved several cord blood-derived hematopoietic progenitor cell products, such as ALLOCORD and Ducord, for hematologic malignancies and transplantation since 2011, establishing them as standard therapies. Emerging applications, like cardiac progenitor cell patches for heart repair, are entering Phase I trials as of 2025, with innovations such as foldable stem cell patches from poised for future human evaluation to regenerate damaged myocardium.

References

  1. [1]
    Progenitor Cell - an overview | ScienceDirect Topics
    Progenitor cells are defined as dividing cells with the capacity to differentiate, including putative stem cells for which self-renewal has not yet been ...
  2. [2]
    Redox Regulation of Stem and Progenitor Cells - PMC - NIH
    First, stem cells are required to differentiate into all the specialized cells that compose a specific tissue or organ, including progenitor cells. Second, stem ...
  3. [3]
    A glossary for stem-cell biology - Nature
    Jun 28, 2006 · Progenitor cell Generic term for any dividing cell with the capacity to differentiate. Includes putative stem cells in which self-renewal has ...
  4. [4]
    Stem and progenitor cells in skeletal development - PubMed Central
    Over the last decade, the definition of skeletal stem and progenitor cells has evolved from cells simply defined by their in vitro behaviors to cells fully ...
  5. [5]
    Blood cell progenitors: Insights into the properties of stem cells - Yoder
    Dec 23, 2003 · Hematopoiesis is a dynamic process in which eight lineages of mature blood cells are derived from a common stem cell.
  6. [6]
    Progenitor Cell - an overview | ScienceDirect Topics
    Progenitor cells are defined as a transient population of cells that retain the capacity for multipotency but are incapable of unlimited self-renewal. AI ...Molecular and Cellular... · Role of Neural Progenitor...
  7. [7]
    Stem cell: what's in a name? - Nature
    Jun 25, 2009 · In other instances, stem cells are referred to as progenitors when the use of the stem cell appellation appears to leave a doubt about their ...
  8. [8]
    the development of mammalian hematopoietic stem cells - PMC
    The origin of the adult mammalian blood system remains a topic of lively discussion and intense research. Interest is also focused on developmental signals that ...Missing: hematology seminal papers
  9. [9]
    Multipotent/pluripotent stem cell populations in stromal tissues ... - NIH
    Oligopotent and unipotent stem cells, often referred to as progenitor cells, have limited differentiation potential [4]. The classification of stem cells based ...
  10. [10]
    Designing stem cell niches for differentiation and self-renewal - PMC
    Niches provide structural and functional cues that are both biochemical and biophysical, stem cells integrate this complex array of signals with intrinsic ...
  11. [11]
    Review The emergence of the stem cell niche - ScienceDirect.com
    The niches not only promote regeneration but also regulate the maturation and functional potential of the progenitors or stem cells they support. Niches should ...
  12. [12]
    Telomeres and Telomerase in the Control of Stem Cells - MDPI
    Incomplete DNA replication and the ensuing telomere shortening cause a loss in the proliferative potential of stem cells. They fail to maintain ...Telomeres And Telomerase In... · 4.2. Germline Stem Cells · 4.6. Tissue-Specific...
  13. [13]
    Asymmetric Division and Lineage Commitment at the Level of ...
    Treatment with certain cytokines increases the chance that CD34−KSL cells will divide in an asymmetric rather than a symmetric manner, with one daughter cell ...
  14. [14]
    Chemokines Regulate the Migration of Neural Progenitors to Sites of ...
    Many studies have shown that transplanted or endogenous neural progenitor cells will migrate toward damaged areas of the brain.
  15. [15]
    Not just a marker: CD34 on human hematopoietic stem/progenitor ...
    Dec 26, 2017 · CD34 is routinely used to identify and isolate human hematopoietic stem/progenitor cells (HSPCs) for use clinically in bone marrow transplantation.
  16. [16]
    The hematopoietic stem cell and its niche: a comparative view
    Stem cells expand their numbers as they self-renew by symmetric division. They can also maintain their numbers and produce rapidly dividing progenitors by ...
  17. [17]
    Molecular mechanisms of stem-cell identity and fate - Nature
    Example: embryonic stem cells. Multipotent. Can form multiple lineages that constitute an entire tissue or tissues. Example: haematopoietic stem cells.
  18. [18]
    Hematopoietic Stem Cell Niches Produce Lineage-Instructive ...
    Dec 20, 2016 · We show that MPPs resided in HSC niches, where they encountered lineage-instructive differentiation signals.Cxcr4 Controls Clp... · Cxcr4 Guides Hematopoietic... · Il-7 Cells Are A Component...
  19. [19]
    The hematopoietic stem cell niche in homeostasis and disease | Blood
    CXCL12 signaling through CXC chemokine receptor 4 (CXCR4) provides a key retention signal for hematopoietic stem and progenitor cells (HSPCs) in the bone marrow ...
  20. [20]
    Sox2, a key factor in the regulation of pluripotency and neural ...
    As such, the physical interaction between Sox2 and Oct4 is likely to be critical in induction and maintenance of pluripotency. SOX2 IN NEURAL DIFFERENTIATION ...
  21. [21]
    Flk-2 is a marker in hematopoietic stem cell differentiation - PNAS
    These findings indicate that long-term repopulating HSCs are Flk-2−, whereas Flk-2+ HSCs are transiently repopulating stem cells with short-term repopulating ...
  22. [22]
    Resolving the hematopoietic stem cell state by linking functional and ...
    Aug 10, 2023 · In this review, we will give a historical overview of the functional and molecular assays in the field, identify new tools that combine molecular and ...
  23. [23]
    Assay systems for hematopoietic stem and progenitor cells
    In vitro assays include colony-forming units and long-term culture. In vivo assays use animal models like fetal sheep and immunodeficient mice. These assays ...
  24. [24]
  25. [25]
  26. [26]
    Epidermal homeostasis: a balancing act of stem cells in the skin - NIH
    Stem cells in the epidermis have a crucial role in maintaining tissue homeostasis by providing new cells to replace those that are constantly lost during tissue ...Skin Development In The... · Asymmetric Cell Division In... · Wnt Signalling At The Root...
  27. [27]
    Gradual differentiation uncoupled from cell cycle exit generates ...
    Nov 10, 2022 · In the mammalian skin epidermis, proliferative basal layer cells differentiate and move upwards to replace barrier-forming cells that are ...
  28. [28]
    IL-1 and TNFα Contribute to the Inflammatory Niche to Enhance ...
    Apr 9, 2019 · We find that interleukin-1 (IL-1) and tumor necrosis factor α (TNFα) enhance the proliferation of AEC2s while maintaining their differentiation capacity.
  29. [29]
    Anti-inflammatory Prowess of endothelial progenitor cells in ... - Nature
    Sep 30, 2024 · The released sTNF-R1 functioned as an anti-inflammatory cytokine by neutralizing TNF-α, ultimately restoring cardiac function after MI, implying ...
  30. [30]
    Dynamic regulation of hematopoietic stem cells by bone marrow ...
    Sep 1, 2019 · It has been proposed that active TGFβ promotes HSC quiescence by inhibiting the formation of cytokine-mediated lipid raft clustering, which is ...
  31. [31]
    Haematopoietic ESL-1 enables stem cell proliferation in the ... - Nature
    Jan 8, 2016 · TGF-beta as a candidate bone marrow niche signal to induce hematopoietic stem cell hibernation. Blood 113, 1250–1256 (2009). Article CAS ...
  32. [32]
    Bone Aging, Cellular Senescence, and Osteoporosis | JBMR Plus
    In studies of mouse osteoprogenitors, it was found that their numbers decline more than 50% with aging and are positive for markers of DNA damage, increased p21 ...<|separator|>
  33. [33]
    Identification and validation of the cellular senescence-associated ...
    Sep 2, 2025 · Cell senescence plays an important role in mediating age-related bone loss and targeting these cells could prevent age-related bone loss [14, 16] ...
  34. [34]
    Cellular senescence and other age-related mechanisms in skeletal ...
    Jul 7, 2025 · In this review, we outline the cellular senescence and other age-related mechanisms in developing skeletal diseases, including osteoporosis, ...
  35. [35]
    The Cross-Talk between Mesenchymal Stem Cells and Immune ...
    Mar 1, 2021 · In this review article, we summarized current knowledge about molecular mechanisms that regulate the crosstalk between MSCs and immune cells in tissue repair ...
  36. [36]
    exploring stem cell roles from injury detection to tissue repair
    Oct 6, 2025 · This activation and proliferation are critical early steps in tissue repair and regeneration, ensuring a sufficient number of cells to replace ...
  37. [37]
    Simultaneous measurement of human hematopoietic stem and ...
    Dec 13, 2015 · In human subjects, HSCs are identified by the cell surface marker profile Lineage- CD34+ CD38- CD90+ CD45RA- 1, 2. The first differentiated cell ...
  38. [38]
    Dynamics of human hematopoietic stem and progenitor cell ...
    Multiple immunophenotypic strategies exist for analysis of hematopoietic stem and progenitor cells (HSPCs) and erythroid cells. •. Flow cytometry and single ...
  39. [39]
    Human CD34+ hematopoietic stem cell hierarchy: how far are ... - NIH
    Indeed, it is currently well recognized that the CD34+CD38+ population represents hematopoietic progenitor cells that encompass different lymphoid, myeloid, ...
  40. [40]
    Article Enhancer and Transcription Factor Dynamics during Myeloid ...
    May 29, 2018 · Transcription factors PU.1 and CEBPA are required for the proper coordination of enhancer activity during granulocytic-monocytic (GM) ...
  41. [41]
    C/EBPα determines hematopoietic cell fate in multipotential ... - NIH
    C/EBPα is an essential transcription factor required for myeloid differentiation. While C/EBPα can act as a cell fate switch to promote granulocyte ...
  42. [42]
    Distinct myeloid progenitor differentiation pathways identified ...
    Oct 4, 2016 · Two myeloid pathways were identified: a Gata1-expressing pathway for mast cells, eosinophils, megakaryocytes, and erythroid cells, and a Gata1- ...
  43. [43]
    Niches that regulate stem cells and hematopoiesis in adult bone ...
    Jul 12, 2021 · Restricted hematopoietic progenitors and erythropoiesis require SCF from leptin receptor+ niche cells in the bone marrow. Cell Stem Cell, 24 ...Endothelial Cells And... · Bone Marrow Stromal Cells... · The Bone Marrow Hsc Niche...
  44. [44]
    Thrombopoietin and hematopoietic stem cells - PMC - NIH
    Thrombopoietin (TPO) supports hematopoietic stem cell (HSC) quiescence and may facilitate HSC expansion and self-renewal, vital for maintaining the HSC pool.
  45. [45]
    Article Assay optimization for the objective quantification of human ...
    Colony-forming unit (CFU) assays can be used to assess the lineage potential of hematopoietic stem and progenitor cells (HSPCs) in vitro. CFU assays are ...
  46. [46]
    Bone marrow failure in Fanconi anemia is triggered by an ...
    Bone marrow failure in Fanconi anemia is triggered by an exacerbated p53/p21 DNA damage response that impairs hematopoietic stem and progenitor cells.
  47. [47]
    The bone marrow of mouse-rat chimeras contains progenitors ... - NIH
    Apr 17, 2024 · To identify respiratory progenitors in donor BM, we performed single-cell RNA sequencing (scRNAseq). ... lung-resident hematopoietic progenitors ...Results · Figure 2 · Figure 5
  48. [48]
    Radial Glial Cells: Defined and Major Intermediates between ...
    Both basal progenitors in the early SVZ and radial glial cells arise from the earlier neuroepithelial cells. (Haubensak et al., 2004; Figure 1). At ...
  49. [49]
    Radial Glial Lineage Progression and Differential Intermediate ...
    Jul 12, 2018 · At 96 hr, Ascl1+ cells exclusively localized to SVZ and were scored as bIPs. ... progenitors located in the SVZ without apical endfeet (Figure 3K) ...
  50. [50]
    For the Long Run Maintaining Germinal Niches in the Adult Brain
    The adult mammalian brain retains neural stem cells that continually generate new neurons within two restricted regions: the subventricular zone (SVZ) of ...
  51. [51]
    An essential role of SVZ progenitors in cortical folding in ... - Nature
    Jul 12, 2016 · Neural progenitors in the cerebral cortex are organized in two germinal layers: the ventricular zone (VZ) and the subventricular zone (SVZ). The ...
  52. [52]
    SOX2 Functions to Maintain Neural Progenitor Identity: Neuron
    We show here that constitutive expression of SOX2 inhibits neuronal differentiation and results in the maintenance of progenitor characteristics.
  53. [53]
    Neurogenesis from Sox2 expressing cells in the adult cerebellar cortex
    Jul 21, 2017 · Triple labelling revealed that 61.4% of Sox2+ Blbp− cells express the stem cell marker Nestin, (Fig. 2a–c; quantification on n = 4 animals; at ...
  54. [54]
    Enhanced differentiation of neural progenitor cells in Alzheimer's ...
    In the brains of patients with AD, reduced expression of proliferation markers such as bromodeoxyuridine (BrdU) and nestin, and increased expression of ...
  55. [55]
    Notch in the Vertebrate Nervous System: An Old Dog with New Tricks
    The Notch pathway regulates neural progenitor maintenance and differentiation, and is involved in cell fate specification, and is studied in both embryonic and ...
  56. [56]
    Bone morphogenetic protein 4 stimulates neuronal differentiation of ...
    Feb 28, 2009 · BMP4 activates ERK pathway and induces differentiation via up-regulation of Ras in NSCs. The Ras-ERK pathway is a major signaling route ...
  57. [57]
    Signaling through BMPR-IA Regulates Quiescence and Long-Term ...
    Jul 2, 2010 · BMPR-IA signaling regulates the balance between quiescence and proliferation of neural stem cells in the hippocampus, and is required for ...
  58. [58]
    Endoplasmic reticulum stress contributes to the decline in ... - Nature
    Jan 19, 2022 · ... hippocampal subgranular zone (SGZ) and subventricular zone (SVZ). A total of 700 new neurons are generated daily in the hippocampus, but ...
  59. [59]
    CRISPR–Cas9 screens reveal regulators of ageing in neural stem ...
    Oct 2, 2024 · Ageing impairs the ability of neural stem cells (NSCs) to transition from quiescence to proliferation in the adult mammalian brain.
  60. [60]
    Brain aging and rejuvenation at single-cell resolution
    Jan 8, 2025 · Finally, we pro- pose new directions, including models of brain aging and neural stem cells as a focal point for rejuvenation. INTRODUCTION.
  61. [61]
    Endothelial Progenitor Cells: An Appraisal of Relevant Data from ...
    Nov 28, 2021 · The mobilization of endothelial progenitor cells (EPCs) into circulation from bone marrow is well known to be present in several clinical ...
  62. [62]
    Circulating endothelial progenitor cells - PMC - NIH
    This increased circulating VEGF promotes the mobilisation of EPCs from the bone marrow (Asahara et al, 1999; Hattori et al, 2001). In contrast, Ang-1 inhibited ...
  63. [63]
    Progenitors in motion: mechanisms of mobilization of endothelial ...
    VEGF contributes to postnatal neovascularization by mobilizing bone marrow-derived endothelial progenitor cells. EMBO J. 1999;18:3964–72. doi: 10.1093/emboj ...
  64. [64]
    Mesenchymal progenitor cells derived from traumatized human ...
    Finally, the MPCs readily exhibited evidence of differentiation into osteoblasts, adipocytes and chondrocytes under the appropriate induction conditions, but ...
  65. [65]
    Determinants of stem cell lineage differentiation toward ... - NIH
    The role of BMP-7 in chondrogenic and osteogenic differentiation of human bone marrow multipotent mesenchymal stromal cells in vitro. J Cell Biochem. 2010 ...
  66. [66]
  67. [67]
    Isolation and characterization of endothelial progenitor cells from ...
    When cultured in Matrigel, the embryonic cells assume the characteristic endothelial cobblestone morphology and form tubes. Injection into chicken embryos ...Missing: methods | Show results with:methods
  68. [68]
    Isolation of Functional Human Endothelial Cells from Small Volumes ...
    Endothelial cells have been successfully isolated from umbilical veins, microvessels, umbilical cord blood, and peripheral blood, but umbilical cord blood is an ...Matrigel Assay · Hucb-Ec Colony Yield · Ec Isolation And...<|separator|>
  69. [69]
    Isolation, differentiation, and characterization of mesenchymal stem ...
    The most important property of BM-MSCs, which is used in isolation and purification process, is their physical adherence to the plastic cell culture plate (15).
  70. [70]
    An improved protocol for isolation and culture of mesenchymal stem ...
    BM-MSCs are usually isolated and purified through their physical adherence to the plastic cell culture plate [16]. Several techniques have been used to purify ...
  71. [71]
    Human placenta-derived endothelial progenitor cells: an animal-free ...
    Jul 2, 2025 · This study developed an animal-free system for isolating, induction, and expanding EPCs from the human placenta, evaluating their potential for ...
  72. [72]
    Senescence-resistant human mesenchymal progenitor cells counter ...
    Emerging research implicates the decline of stem cell populations and a diminishing regenerative potential as pivotal to the aging process.
  73. [73]
    FOXO3-enhanced mesenchymal progenitor cells impede aging in ...
    Jul 7, 2025 · Lei et al. now demonstrate that injection of senescence-resistant mesenchymal progenitor cells reduces molecular and functional features of primate aging.
  74. [74]
    The role of endothelial cell–pericyte interactions in vascularization ...
    Intercellular communication between ECs and PCs is vital for the formation, stability, and function of blood vessels. Various signaling pathways, such as the ...
  75. [75]
    Pericyte-Endothelial Cross-Talk: Implications and Opportunities for ...
    Differentiation of mesenchymal cells to pericytes when co-cultured with endothelial cells is associated with increased VEGF production by mesenchymal cells and ...
  76. [76]
    Mesenchymal Stem Cells: Roles and Relationships in Vascularization
    This review aims to provide a summary of relevant relationships between MSCs, vascularization, and other relevant cell types, along with an overview discussing ...
  77. [77]
    Dividing Precursor Cells of the Embryonic Cortical Ventricular Zone ...
    Apr 15, 2002 · Taken together, these data show that radial glia, in addition to being scaffolds for neuronal migration, also serve as neuronal progenitors. It ...
  78. [78]
    How neural stem cells contribute to neocortex development
    Aug 16, 2021 · Green arrows indicate the interkinetic nuclear migration during the cell cycle. (B) NPCs in the developing neocortex. Apical radial glia (aRG) ( ...
  79. [79]
    Pax6, Tbr2, and Tbr1 Are Expressed Sequentially by Radial Glia ...
    Jan 5, 2005 · The sequential expression of Pax6, Tbr2, and Tbr1 suggests that each TF regulates discrete steps in projection neuron differentiation.
  80. [80]
    Intermediate progenitors and Tbr2 in cortical development - Hevner
    Jan 24, 2019 · In developing cerebral cortex, intermediate progenitors (IPs) are transit amplifying cells that specifically express Tbr2 (gene: Eomes), a T-box ...
  81. [81]
    Brn-1 and Brn-2 share crucial roles in the production and positioning ...
    Lineage analyses and birthdating studies suggest that common cortical precursor cells first produce neurons of layer VI and then layer V (at E11.5–E15.5) and, ...<|control11|><|separator|>
  82. [82]
    Novel Subtype-Specific Genes Identify Distinct Subpopulations of ...
    Sep 30, 2009 · CPN of the deep layers are born during early corticogenesis (E11.5–E13.5 in the mouse) from progenitors located in the ventricular zone (VZ) ...
  83. [83]
    Gliogenesis in the outer subventricular zone promotes enlargement ...
    Mar 20, 2019 · Our data indicate that oSVZ gliogenesis, rather than neurogenesis, correlates with rapid enlargement of the cerebrum and development of convolutions.
  84. [84]
    The Outer Subventricular Zone and Primate-Specific Cortical ...
    Feb 18, 2015 · Evolutionary expansion and complexification of the primate cerebral cortex are largely linked to the emergence of the outer subventricular zone (OSVZ).Main Text · Osvz And Cortical Expansion · Diversity Of Basal Precursor...<|control11|><|separator|>
  85. [85]
    Intermediate progenitors and Tbr2 in cortical development - PMC
    In developing cerebral cortex, intermediate progenitors (IPs) are transit amplifying cells that specifically express Tbr2 (gene: Eomes), a T‐box transcription ...
  86. [86]
    Cortical progenitor cells in the developing human telencephalon
    Proliferating RG (4A4+) cells in the ventricular zone (VZ) showed clear caudorostral and ventrodorsal gradients, spreading from the spinal cord to the ventral ...Missing: timeline pre- E40 post- mesenchymal daughters column formation
  87. [87]
    Molecular signatures of cortical expansion in the human foetal brain
    Nov 8, 2024 · Our findings demonstrate a spatial coupling between areal differences in the timing of neurogenesis and rates of neocortical expansion during gestation.
  88. [88]
    Forced G1-phase reduction alters mode of division, neuron number ...
    Dec 22, 2009 · Here, we induced a 25% reduction in TG1 in mouse cortical precursors via forced expression of cyclin D1 and cyclin E1. We found that in utero ...
  89. [89]
    Cdk4/CyclinD1 Overexpression in Neural Stem Cells Shortens G1 ...
    Sep 4, 2009 · Overexpression of cdk4/cyclinD1 inhibited neurogenesis while increasing the generation and expansion of basal (intermediate) progenitors.
  90. [90]
    CYCLIN-B1/2 and -D1 act in opposition to coordinate cortical ...
    Jun 9, 2020 · For example, by regulating the length of the G1 phase in the cortex, CYCLIN-D1 has been implicated in controlling the onset of neurogenesis by ...
  91. [91]
    Caspases in the Developing Central Nervous System: Apoptosis ...
    A multiprotein complex called the apoptosome, comprising cytochrome c, Apaf-1 and caspase-9 activates caspase-3, leading to cell death. The IAPs prevent caspase ...
  92. [92]
    Caspase-3 Mediated Cell Death in the Normal Development ... - MDPI
    Caspases are endoproteases and mediate some forms of neuronal programmed cell death (PCD), such as apoptosis and pyroptosis, which are dependent on caspase-3 ...Caspase-3 Mediated Cell... · 3. Caspase-3 In Cerebellar... · Apoptosis<|separator|>
  93. [93]
    From stem and progenitor cells to neurons in the developing ...
    Feb 27, 2021 · Humans have a threefold larger cerebral cortex than chimpanzees [[5]], and it is estimated to contain twice as many neurons [[6]] (Figure 1). It ...
  94. [94]
    Cortex folding by combined progenitor expansion and adhesion ...
    Aug 28, 2025 · Folding of the mammalian cerebral cortex into sulcal fissures and gyral peaks is the result of complex processes that are incompletely ...
  95. [95]
    Potential mechanisms of Zika‐linked microcephaly - PMC
    ... depletes the pool of neural progenitors in the developing brain. ... Zika virus infects human cortical neural progenitors and attenuates their growth.
  96. [96]
    Recent Zika Virus Isolates Induce Premature Differentiation of ...
    Mar 2, 2017 · NPC depletion leads to cortical thinning resembling ZIKV-associated microcephaly. Summary. The recent Zika virus (ZIKV) epidemic is associated ...
  97. [97]
    Decoding functional hematopoietic progenitor cells in the adult ...
    May 1, 2025 · The gene signature of pulmonary and medullary CD34+ hematopoietic progenitors indicates greater baseline activation of immune-, megakaryocyte/ ...
  98. [98]
    Hematopoietic stem and progenitor cells as a reservoir for trained ...
    Sep 4, 2025 · Long-lived hematopoietic stem and progenitor cells (HSPCs) serve as a reservoir for innate immune memory, sustaining trained immunity.
  99. [99]
    Make hematopoiesis great again: countering oxidative stress! | Blood
    Mar 13, 2025 · During aging, increased oxidative stress reduces HSC regenerative capacity and skews lineage differentiation toward myelopoiesis. Similarly, ...
  100. [100]
    Hallmarks and mechanisms of cellular senescence in aging ... - Nature
    Aug 4, 2025 · DNA damage, oxidative stress, and telomere shortening are the primary triggers of cellular senescence, endowing senescent cells with deregulated ...
  101. [101]
    Organoids in disease modeling and regenerative medicine
    Apr 21, 2025 · Organoid technology has the potential to revolutionize biomedical research by providing more physiologically relevant models for studying human development.<|separator|>
  102. [102]
    Neural Organoids: Disease Modeling & Regenerative Medicine
    May 24, 2025 · Neural organoids hold great potential for disease modeling. Organoids generated from patient-derived stem cells can model the pathological ...
  103. [103]
    Combined targeting of glioblastoma stem cells of different cellular ...
    Mar 26, 2025 · Calculation of achievable Notch and NF-κB pathway inhibitors concentrations. Our studies used 200 mg/kg RG-4733 (Notch signaling inhibitor) ...
  104. [104]
    Stem cell therapy for myocardial infarction and atherosclerosis
    Oct 9, 2025 · Clinical trials of EPC infusion in peripheral artery disease and coronary artery disease have demonstrated safety and improved endothelial ...<|control11|><|separator|>
  105. [105]
    Treatment of knee osteoarthritis with intra-articular injection of ...
    May 21, 2019 · Human adipose-derived mesenchymal progenitor cells (haMPCs) are stem cells with multiple differentiation potential and immunomodulatory function ...
  106. [106]
    Neural xenografts contribute to long-term recovery in stroke via ...
    Sep 16, 2025 · Here, we show that local transplantation of iPSC-derived neural progenitor cells (NPCs) improves brain repair and long-term functional recovery ...
  107. [107]
    Hematopoietic stem and progenitor cells as a reservoir for trained ...
    Sep 4, 2025 · In this review, we summarize the evidence that innate immune memory is epigenetically encoded in HSPCs, and we consider whether HSPC ...
  108. [108]
    Trained Immunity Contribution to Autoimmune and Inflammatory ...
    Trained immunity can induce a beneficial response both in AIF and autoimmune conditions by inducing anti-inflammatory responses.
  109. [109]
    Effects of histocompatibility and host immune responses on the ... - NIH
    Among the major concerns for the clinical application of stem cell-derived grafts are the risks of immune rejection and tumor formation. Pluripotency and ...
  110. [110]
    Immunological considerations and challenges for regenerative ...
    Jun 25, 2021 · Here we describe the role of the host innate and adaptive immune responses as triggers of allogeneic graft rejection.
  111. [111]
    Applications of CRISPR-Cas9 in mitigating cellular senescence and ...
    Jul 8, 2025 · This paper explores the potential of CRISPR/Cas9 as a gene-editing tool to target these mechanisms and mitigate age-related conditions.
  112. [112]
    Attenuation of primate aging via systemic infusion of senescence ...
    Jun 27, 2025 · Infusion of FOXO3-enhanced SRCs slowed aging in primates, improving cognitive function, bone integrity, and immune function, reversing the  ...
  113. [113]
    Approved Cellular and Gene Therapy Products - FDA
    Aug 15, 2025 · Approved Products ; CLEVECORD (HPC Cord Blood), Cleveland Cord Blood Center ; Ducord, HPC Cord Blood, Duke University School of Medicine ; ELEVIDYS ...HPC, Cord Blood · ALLOCORD (HPC Cord Blood) · DUCORD (HPC Cord Blood)Missing: hematology cardiac
  114. [114]