Fact-checked by Grok 2 weeks ago

CFU-GEMM

The CFU-GEMM (colony-forming unit–granulocyte, erythrocyte, , , ) is a multipotent derived from long-term hematopoietic stem cells (LT-HSCs) in the , capable of differentiating into multiple myeloid lineages including , erythrocytes, monocytes/, and ( to platelets). These progenitors form multilineage colonies when cultured in semisolid media supplemented with specific cytokines such as (GM-CSF), (EPO), and (SCF), serving as a key intermediate stage in between pluripotent stem cells and more lineage-committed progenitors like burst-forming unit-erythroid (BFU-E) and colony-forming unit-granulocyte/ (CFU-GM). Unlike true stem cells, CFU-GEMM cells exhibit limited self-renewal capacity and high proliferative potential, with their activity regulated by a stromal microenvironment and hematopoietic growth factors.

Definition and Overview

Role in Hematopoiesis

The colony-forming unit-granulocyte, erythrocyte, , megakaryocyte (CFU-GEMM) represents an oligopotent common myeloid capable of differentiating into multiple myeloid lineages, distinguishing it from more primitive hematopoietic cells with broader potential. This progenitor is defined by its ability to form multilineage colonies , encompassing cells from granulocytic, erythroid, monocytic, and megakaryocytic compartments, as initially characterized through culture assays of human samples. In the hematopoietic hierarchy, CFU-GEMM resides downstream of long-term hematopoietic stem cells (LT-HSCs), which possess extensive self-renewal capacity, and upstream of lineage-committed progenitors such as the granulocyte-macrophage progenitor (CFU-GM) or the erythroid progenitor (CFU-E). This positioning enables CFU-GEMM to serve as a critical intermediate stage, amplifying myeloid output while restricting pluripotency to myeloid fates. CFU-GEMM plays an essential role in producing key myeloid cell types, including granulocytes (such as neutrophils, eosinophils, and basophils), erythrocytes (red blood cells), monocytes (which differentiate into macrophages), and megakaryocytes (precursors to platelets). These contributions support steady-state hematopoiesis, maintaining daily blood cell turnover, as well as emergency responses to stressors like infections—where granulocytes and monocytes combat pathogens—or blood loss, which demands rapid replenishment of erythrocytes and platelets. A point of debate concerns the lineage's origin, with classical views assigning it to CFU-GEMM-derived , while recent lineage-tracing studies propose that may emerge from a distinct erythro-myeloid pathway separate from development.

Historical Background

The foundational work on hematopoietic colony-forming units began in the with James Till and Ernest McCulloch, who developed the and spleen colony assay to identify primitive hematopoietic cells (HSCs) capable of multilineage and self-renewal in irradiated mice. Their experiments demonstrated that single cells could generate macroscopic colonies in the , comprising multiple types, laying the groundwork for recognizing multipotent progenitors downstream of HSCs. This approach extended in the early to distinguish committed progenitors from more primitive cells, highlighting the in hematopoiesis where multipotent cells give rise to lineage-restricted ones. In the mid-1970s, the specific identification of multipotent myeloid progenitors, later termed CFU-GEMM (colony-forming unit-granulocyte, erythrocyte, /), emerged through culture assays using semisolid media like methylcellulose. Axel A. Fauser and Hans A. Messner first described these cells in 1978, observing mixed granuloerythropoietic colonies from human , peripheral blood, and that contained granulocytes and erythrocytes, distinguishing them from unipotent progenitors such as CFU-E (erythroid-only) or CFU-GM (granulocyte-macrophage). By 1979, they formalized the term CFU-GEMM to denote these oligopotent cells with multilineage potential limited to myeloid lineages, contrasting them with the broader pluripotency of HSCs. The brought key advancements in confirming CFU-GEMM's multilineage potential through refined semisolid systems supplemented with conditioned media and early cytokines, enabling clearer visualization of megakaryocytic within colonies. These techniques solidified CFU-GEMM as an intermediate with limited self-renewal, unlike self-renewing HSCs, evolving the view from a vaguely "primitive" to a defined oligopotent entity in the hematopoietic . A notable clinical milestone came in with studies by G. David Roodman and colleagues, who linked CFU-GEMM numbers in autologous grafts to faster and platelet recovery post-high-dose transplantation, bridging experimental assays to hematologic recovery in patients.

Cellular Properties

Morphology and Location

CFU-GEMM cells exhibit a blast-like morphology, appearing as small, round structures typically measuring 8-12 μm in diameter, with a high nucleus-to-cytoplasm ratio, scant basophilic cytoplasm, and immature, finely dispersed chromatin. These characteristics reflect their progenitor status, distinguishing them from more mature myeloid cells while sharing features with early hematopoietic blasts. Compared to more primitive hematopoietic stem cells (HSCs), which are similarly small (7-10 μm) with an even higher nucleus-to-cytoplasm ratio and minimal cytoplasm, CFU-GEMM display slightly more differentiated traits, such as subtle cytoplasmic basophilia indicative of emerging multilineage potential, yet they retain overall blast-like immaturity. These progenitors are primarily localized within the , residing in specialized niches including the endosteal region near osteoblasts and the perivascular areas around sinusoidal vessels, where interactions with stromal cells provide essential support for maintenance and quiescence. CFU-GEMM constitute a low-abundance population, representing approximately 0.001-0.01% (1-10 per 10^5) of nucleated cells in healthy adults, underscoring their rarity and the efficiency of the hematopoietic hierarchy. Under normal conditions, CFU-GEMM are confined to the , but they possess limited migration potential, occasionally entering peripheral blood circulation during stress responses such as or , though at exceedingly low frequencies. This mobilization supports emergency but is tightly regulated to prevent ectopic differentiation.

Surface Markers

CFU-GEMM cells exhibit a distinct immunophenotypic profile characterized by the expression of key surface markers that distinguish them as multipotent myeloid progenitors. The primary marker is , a sialomucin associated with hematopoietic stem and progenitor cells, which is uniformly expressed on CFU-GEMM to facilitate their identification and isolation. They also positively express , a sialic acid-binding immunoglobulin-like specific to myeloid lineage cells, and , a major histocompatibility complex class II molecule that supports on early progenitors. Additionally, CD117 (c-Kit), the for , is present at moderate to high levels, promoting survival and proliferation signals. , an ectoenzyme involved in and signaling, is expressed at positive levels (CD38+), marking commitment to myeloid lineages. In contrast, CFU-GEMM display low or absent expression of mature lineage commitment markers, rendering them lineage-negative (Lin-). This includes the lack of CD3 (T-cell marker), (B-cell marker), and CD235a (, a mature erythroid marker), which ensures their multipotency across , erythrocyte, , and lineages. The typical profile is thus ++CD45RA-Lin-, often refined further by low IL-3Rα (CD123) to exclude more differentiated subsets like granulocyte-macrophage progenitors. These negative markers are crucial for excluding contaminated mature cells during purification. This immunophenotype enables precise enrichment of CFU-GEMM via sorting, allowing for downstream functional assays such as assays to validate their multipotent potential. Heterogeneity exists within CFU-GEMM populations, with subpopulations showing varying expression levels of CD117 and low or absent (Thy-1), a marker more prominent in long-term hematopoietic stem cells, reflecting subtle differences in developmental priming. Recent advances in single-cell sequencing have confirmed these marker combinations, enabling the isolation of highly pure CFU-GEMM fractions and revealing transcriptional signatures that correlate with their colony-forming efficiency and differentiation bias; emerging markers like CD49f and refined /CD45RA panels as of 2024 further improve specificity.

Development and Regulation

Origin and Progenitor Hierarchy

The CFU-GEMM arises within the myeloid branch of the hematopoietic , deriving from multipotent hematopoietic cells (HSCs) through an myeloid (CMP) . HSCs, residing primarily in the niche, first generate multipotent progenitors (MPPs), which then commit to the myeloid lineage as CMPs; these CMPs possess multilineage potential and serve as the direct upstream source for CFU-GEMM cells, enabling the formation of colonies with , erythrocyte, , and lineages in functional assays. This progression reflects a stepwise restriction of developmental potential, with CMPs identified by surface markers such as Lin⁻ ⁺ in humans, confirming their clonogenic capacity to produce CFU-GEMM. Upstream commitment from long-term HSCs (LT-HSCs) to this myeloid pathway is regulated by niche-derived signals, including Wnt and pathways, which balance self-renewal and differentiation decisions in the microenvironment. Wnt signaling, in particular, acts in a dosage-dependent manner to promote HSC maintenance while facilitating progression toward myeloid progenitors like CMPs, whereas signaling supports early HSC survival and influences lineage priming toward myeloid fates. These extrinsic cues integrate with intrinsic factors to drive LT-HSC quiescence exit and initial myeloid specification, ensuring controlled expansion of the progenitor pool without exhaustion. Downstream of CFU-GEMM, the branches into more lineage-restricted progenitors, primarily megakaryocyte-erythroid progenitors (MEPs) and granulocyte-macrophage progenitors (GMPs), marking the toward erythro-megakaryocytic versus granulocytic-monocytic outputs. This limits CFU-GEMM's multipotency, as it transitions from generating mixed colonies to supporting unilineage commitment in MEPs (e.g., toward erythrocytes and megakaryocytes) and GMPs (e.g., toward neutrophils and monocytes). Unlike HSCs, which possess indefinite self-renewal potential, CFU-GEMM exhibits limited self-renewal capacity but high proliferative potential, forming large colonies while being unable to sustain long-term repopulation . This restriction underscores their progenitor status, preventing indefinite expansion and ensuring rapid turnover in response to hematopoietic demands. Key transcription factors, such as PU.1 and C/EBPα, drive this commitment at balanced expression levels; moderate PU.1 dosage promotes multilineage potential in CFU-GEMM, while C/EBPα cooperates to enforce myeloid fate decisions from CMPs, integrating with other regulators to halt self-renewal and initiate .

Growth Factors and Cytokines

The growth and differentiation of CFU-GEMM cells are primarily regulated by a suite of cytokines and growth factors that support survival, proliferation, and multilineage commitment. (SCF), also known as kit ligand, plays a crucial role in promoting the survival of CFU-GEMM progenitors by binding to the c-Kit receptor, preventing in early hematopoietic stages. (IL-3) acts as a potent multilineage inducer, stimulating the proliferation and differentiation of CFU-GEMM into multiple myeloid lineages, including , erythrocytes, monocytes, and megakaryocytes, through activation of the IL-3 receptor alpha chain. (GM-CSF) provides a toward myeloid differentiation, enhancing the formation of CFU-GEMM-derived colonies with a predominance of and progenitors when combined with other factors. For lineage-specific commitment, (EPO) is essential in directing CFU-GEMM toward the erythroid pathway, promoting erythrocyte maturation by signaling through the EPO receptor to induce synthesis and cell survival. Similarly, thrombopoietin (TPO) drives lineage development from CFU-GEMM, supporting megakaryopoiesis and platelet production via the c-Mpl receptor, with effects amplified in multilineage contexts. These factors often exert synergistic effects, where combinations markedly enhance CFU-GEMM activity beyond individual contributions; for instance, SCF combined with IL-3 and EPO can increase multipotential colony formation up to 15-fold in cultures, typically at concentrations of 10-100 ng/mL. Such synergies are evident in serum-free assays, where SCF + IL-3 + EPO yield mixed colonies reflecting the full differentiation potential of CFU-GEMM. Negative regulation is provided by transforming growth factor-beta (TGF-β), which inhibits CFU-GEMM proliferation to maintain hematopoietic and prevent excessive myeloid output; at concentrations around 400 pmol/L, TGF-β suppresses colony formation in the presence of IL-3 or GM-CSF by up to 98%. Recent advances since 2015 have highlighted the small molecule UM171, a pyrimidoindole derivative, in expansion protocols for enhancing CFU-GEMM and related progenitors, promoting robust hematopoietic reconstitution in transplant settings by preserving quiescence and multilineage potential.

Functional Characteristics

Colony Formation

The colony-forming unit-granulocyte, erythrocyte, , (CFU-GEMM) assay is a standard method to detect and quantify these multipotent hematopoietic progenitors by their ability to form mixed-lineage colonies in semisolid culture media. In the classic methylcellulose-based protocol, such as using MethoCult™ media, mononuclear cells from or peripheral blood are plated at densities of 1 × 10^4 to 5 × 10^4 cells per 35-mm dish in a mixture containing 1.1% methylcellulose, Iscove's modified Dulbecco's medium, , and a cocktail of recombinant growth factors including , interleukin-3, , , and . Cultures are incubated at 37°C in a humidified atmosphere with 5% CO2 for 14 days, after which CFU-GEMM colonies—defined as aggregates exceeding 500 cells exhibiting multiple lineages (granulocytes, erythrocytes, , and )—are identifiable under an by their heterogeneous morphology, including erythroid bursts with surrounding myeloid cells and occasional . To assess the potential and self-renewal capacity of CFU-GEMM, replating is evaluated by dissociating primary colonies and re-plating cells or colony fragments into secondary cultures under similar conditions. This measures the proportion of primary CFU-GEMM that generate secondary mixed colonies, serving as an indicator of hierarchy. blood-derived CFU-GEMM demonstrate notably higher replating compared to adult counterparts, with up to 80-90% of primary colonies yielding secondary CFU-GEMM when enhanced by plasma, reflecting greater proliferative potential in neonatal sources. Colony formation can be augmented by cytokines such as interleukin-1 (IL-1), which indirectly stimulates CFU-GEMM growth by inducing fibroblasts to secrete supportive factors like colony-stimulating activities. In s supplemented with recombinant IL-1 at concentrations of 10-100 ng/mL, the number of CFU-GEMM colonies increases in a dose-dependent manner, with maximal enhancement observed around 140 ng/mL when combined with standard growth factors. Modern variants of the incorporate alternative matrices to better mimic the niche, such as fibrin clot cultures where cells are embedded in thrombin-polymerized fibrinogen gels (0.5-1 mg/mL) supplemented with growth factors, or 3D systems like heparin-functionalized DNA networks loaded with , which promote higher CFU-GEMM yields and primitive phenotypes compared to traditional 2D methylcellulose. Quantification involves manual or automated scoring of colonies based on size, morphology, and lineage composition, typically using inverted or imaging software like STEMvision™ for . In normal human , CFU-GEMM frequency ranges from approximately 1 in 10^4 to 10^5 mononuclear cells, though this varies with donor age and health status. Despite its utility, the CFU-GEMM has limitations, including incomplete recapitulation of hematopoiesis due to the absence of vascular and stromal interactions, leading to potential underestimation of function. Additionally, subjective scoring can introduce variability, with non-clonal aggregates (clusters of 20-50 cells) sometimes mistaken for true colonies, and inter-laboratory remains challenging without .

Differentiation Potential

The CFU-GEMM (colony-forming unit-granulocyte, erythrocyte, , ) represents a multipotent capable of differentiating into multiple myeloid lineages, including granulocytes, erythrocytes, , and , thereby contributing to the diversity of production. This multilineage potential is demonstrated through colony-forming assays where single CFU-GEMM cells generate mixed colonies containing cells from all four lineages, confirming their oligopotency as early myeloid . Differentiation into specific lineages is guided by key growth factors and cytokines. commitment occurs primarily via (G-CSF), which promotes proliferation and maturation of precursors from CFU-GEMM-derived progenitors. differentiation is driven by (EPO), inducing synthesis and enucleation in maturing precursors. development relies on (M-CSF, also known as CSF-1), supporting expansion and phagocytic function. For megakaryocytes, thrombopoietin (TPO) is essential, facilitating polyploidization and platelet release. These pathways involve both symmetric cell divisions, which expand progenitor pools, and asymmetric divisions, which produce one progenitor and one more differentiated daughter cell, allowing balanced self-renewal and output. In culture, CFU-GEMM typically unfolds over a 14- to 16-day time course, with initial proliferation yielding committed progenitors by day 7 and mature cells appearing by days 14 to 21, as observed in methylcellulose-based assays. differentiation is marked by lineage-specific surface antigens, such as (CD235a) on erythrocytes, indicating late-stage erythroid maturation, and CD41 ( αIIb) on megakaryocytes, signifying proplatelet formation competence. CFU-GEMM exhibits plasticity in response to environmental cues, skewing output toward erythroid lineages under hypoxic conditions through upregulated EPO signaling, while inflammatory signals promote myeloid ( and ) bias via enhanced G-CSF and M-CSF pathways. This adaptability ensures adaptive hematopoiesis but can contribute to pathological imbalances if dysregulated. In vivo validation of CFU-GEMM potential comes from lineage-tracing studies in models, which demonstrate multilineage reconstitution paralleling observations, with common myeloid progenitors giving rise to , erythroid, , and compartments in competitive repopulation assays. These findings underscore the conserved hierarchy across species, supporting the relevance of murine data to hematopoiesis.

Clinical and Research Significance

Involvement in Hematological Disorders

In (AML), dysfunction of CFU-GEMM progenitors manifests as clonal expansion due to blocked at early myeloid stages, leading to accumulation of immature blasts that impair normal multilineage output. This blockage often involves fusion proteins like CBFβ-SMMHC, which disrupt hematopoietic and promote leukemic transformation by altering progenitor self-renewal. In contrast, features a marked reduction in CFU-GEMM numbers, contributing to through depleted pluripotent progenitors and impaired repopulation. Myelodysplastic syndromes (MDS) exhibit impaired CFU-GEMM function, resulting in ineffective hematopoiesis characterized by dysplastic changes and cytopenias, particularly and . CFU-GEMM colony formation is significantly reduced in MDS patients compared to healthy controls, with frequencies as low as 6.6 ± 0.9 per 10^5 mononuclear cells versus 18.5 ± 2.1, reflecting multilineage progenitor defects that exacerbate peripheral blood deficiencies. This impairment stems from intrinsic abnormalities and microenvironmental factors, leading to apoptotic loss and maturation arrest in erythroid, myeloid, and megakaryocytic lineages. Post-chemotherapy, delayed regeneration of CFU-GEMM progenitors prolongs neutropenia by depleting bone marrow reserves and slowing multilineage recovery, often requiring growth factor support to restore granulocyte output. In acute lymphoblastic leukemia patients at diagnosis, CFU-GEMM numbers are reduced to 12.1 per ml bone marrow (95% CI: 2.1-70), with intensified chemotherapy further affecting reconstitution and contributing to infection risks during nadir periods. Genetic mutations in transcription factors such as RUNX1 and disrupt CFU-GEMM multilineage potential, predisposing to disorders like familial platelet disorder with AML propensity or -associated megakaryoblastic leukemia. RUNX1 mutations enhance self-renewal while blocking granulocytic differentiation in hematopoietic progenitors, altering target in stem and progenitor compartments. Similarly, truncating mutations inhibit CFU-GEMM colony formation, particularly in models of , by downregulating megakaryocytic and erythrocytic pathways while sparing some myeloid output. Low CFU-GEMM frequency serves as a diagnostic and prognostic marker in hematological disorders, predicting poor outcomes in autologous marrow transplantation. In patients receiving high-dose , CFU-GEMM counts correlated directly with and platelet recovery times, with reduced frequencies indicating delayed engraftment and higher complication risks.

Therapeutic Applications and Studies

CFU-GEMM cells play a critical role in transplantation by serving as multipotent progenitors that contribute to rapid engraftment and multilineage reconstitution. Enrichment of CFU-GEMM populations in grafts has been shown to enhance engraftment efficiency, particularly in transplants where progenitor numbers are limited. For instance, expansion using the small molecule UM171 significantly increases CFU-GEMM output while preserving long-term repopulating potential, leading to improved hematopoietic recovery in preclinical models. In for inherited blood disorders, of CFU-GEMM progenitors enables stable correction of genetic defects and sustained production of functional blood cells. This approach has been applied to β-thalassemia, where lentiviral vectors CFU-GEMM to express normal β-globin, restoring production in erythroid lineages without impairing multipotency. Early studies in models confirmed efficient in CFU-GEMM, supporting their use in autologous transplants to achieve therapeutic engraftment. Key foundational studies have highlighted the potential of CFU-GEMM modulation for therapeutic expansion. Zucali et al. demonstrated that interleukin-1 (IL-1) enhances CFU-GEMM colony formation by stimulating stromal cells to produce colony-stimulating factors, providing a basis for cytokine-based protocols in transplant settings. Additionally, cord blood-derived CFU-GEMM exhibit superior replating capacity compared to counterparts, allowing greater expansion while maintaining multilineage differentiation, which underscores their advantage in pediatric transplants. Recent advances from 2015 to 2025 have expanded CFU-GEMM applications in targeted therapies. CRISPR-Cas9 editing of CFU-GEMM has been explored to confer resistance to leukemic therapies, such as ablating to protect healthy progenitors during antibody-drug conjugate treatments for (AML), enabling safer allogeneic transplants. A 2024 study revealed lung-resident CFU-GEMM-like progenitors in adult human pulmonary tissue, capable of multilineage hematopoiesis, suggesting novel sites for extramedullary progenitor harvesting or manipulation in respiratory-related disorders. Despite these progresses, challenges persist in therapeutic manipulation of CFU-GEMM, including limited expansion without loss of multipotency, which risks differentiation bias and reduced engraftment. Ongoing clinical trials are addressing these by investigating progenitor-enriched grafts for AML relapse prevention, focusing on mutation-bearing subpopulations like GMP-like cells that drive recurrence post-remission.

References

  1. [1]
    Hematopoiesis - Holland-Frei Cancer Medicine - NCBI Bookshelf - NIH
    Although the CFU-GEMM is a cell that is multipotent by virtue of its ability to give rise to multiple cell lineages, it has very limited capacity for ...
  2. [2]
    [PDF] Human Colony-Forming Unit (CFU) Assays Using MethoCult™
    CFU-GEMM: Colony-forming unit-granulocyte, erythrocyte, macrophage, megakaryocyte. A multi-potential progenitor that produces a colony containing erythroblasts ...Missing: name | Show results with:name
  3. [3]
    Granuloerythropoietic colonies in human bone marrow, peripheral ...
    Dec 1, 1978 · Colonies that contain granulopoietic and erythropoietic cells can be grown in specimens of human bone marrow, peripheral blood, and cord ...Missing: Granular | Show results with:Granular
  4. [4]
    Granuloerythropoietic colonies in human bone marrow ... - PubMed
    Colonies that contain granulopoietic and erythropoietic cells can be grown in specimens of human bone marrow, peripheral blood, and cord blood.Missing: Granular pdf
  5. [5]
    The role of CFU-GEMM in human hemopoiesis | Annals of Hematology
    Fauser AA, Messner HA (1978) Granuloerythropoietic colonies in human bone ... About this article. Cite this article. Messner, H.A. The role of CFU-GEMM in human ...
  6. [6]
    Colony-forming unit cell (CFU-C) assays at diagnosis - NIH
    In vitro colony-forming unit cell (CFU-C) assays are usually-used to detect the quantitative and qualitative features of haematopoietic stem cells (HSCs).
  7. [7]
    CFU-GEMM - an overview | ScienceDirect Topics
    The CFU-GEMM is the most primitive progenitor cell which forms multilineage colonies of granulocytes, erythrocytes, monocytes, and macrophages on semisolid ...
  8. [8]
    Hematopoietic colony-forming cells derived from human embryonic ...
    Cells that form hematopoietic colonies (so-called CFUs or CFCs) represent a stage of hematopoietic differentiation between HSCs and more terminally ...
  9. [9]
    Colony Forming Unit GEMM - an overview | ScienceDirect Topics
    The CFU-S represents what we now refer to as committed myeloid progenitors or colony-forming unit–granulocyte, erythrocyte, monocyte, and megakaryocyte (CFU- ...
  10. [10]
  11. [11]
    Proliferative state of human pluripotent hemopoietic progenitors ...
    Nov 1, 1979 · The proliferative state of human pluripotent hemopoietic progenitors (CFU-GEMM) was assessed in normal bone marrow transplant donors and in ...Missing: original | Show results with:original
  12. [12]
  13. [13]
    CFU-GEMM correlate with neutrophil and platelet recovery in ...
    CFU-GEMM correlate with neutrophil and platelet recovery in patients receiving autologous marrow transplantation after high-dose melphalan chemotherapy · Authors.
  14. [14]
    The White Blood Cell and Differential Count - Clinical Methods - NCBI
    In most cases blasts will be found. These are large, immature-looking cells with a high nucleus to cytoplasm ratio. Myeloblasts have a thin, lacy chromatin ...
  15. [15]
    Isolation of Myeloid Progenitor Cells From Peripheral Blood of ...
    These cells were large (approximately 15μ), basophilic, and had a high nuclear:cytoplasmic ratio, multiple nucleoli, and few or no cytoplasmic granules.Missing: nucleus | Show results with:nucleus
  16. [16]
    Characteristics of hematopoietic stem cells of umbilical cord blood
    The morphology of cord blood hematopoietic cells. The morphology of human HSC is similar in appearance to a small cell with a narrow hem cytoplasm, in which ...
  17. [17]
    The physical microenvironment of hematopoietic stem cells and its ...
    Nov 19, 2019 · In this review, we focus on emerging evidence that hematopoietic stem cell fate is altered in response to a variety of microenvironmental physical cues.
  18. [18]
    Bone Niches, Hematopoietic Stem Cells, and Vessel Formation - PMC
    Jan 13, 2017 · HSCs are localized in both the endosteum, in the so-called endosteal niche, and close to thin-walled and fenestrated sinusoidal vessel in the center of BM, in ...
  19. [19]
    Prospective isolation of human clonogenic common myeloid ... - PNAS
    The hierarchical development from hematopoietic stem cells to mature cells ... (CFU-GEMM), burst-forming units erythroid, CFU-megakaryocyte, CFU ...Materials And Methods · In Vitro Assays To Determine... · Results
  20. [20]
    Hematopoietic stem and progenitor cells are differentially mobilized ...
    ... (CFU-GM) assay is specifically designed to quantify the frequency of HPC. CFU-GM were cultured by resuspending 5×10 peripheral blood cells, 5×10 bone marrow ...
  21. [21]
    Social Stress Mobilizes Hematopoietic Stem Cells to Establish ...
    Nov 27, 2018 · We show that social stress in mice mobilizes hematopoietic stem progenitor cells (HSPCs) from the bone marrow that enter circulation, engraft into the spleen,
  22. [22]
    Severe Trauma and Chronic Stress Activates Extramedullary ... - NIH
    On day seven, the addition of CS to LCHS increased spleen weight by 22%. LCHS/CS increased splenic growth of CFU-GEMM, BFU-E and CFU-E colonies by 28–39% vs.
  23. [23]
    Page not found | Nature
    Insufficient relevant content.
  24. [24]
    A comprehensive single cell transcriptional landscape of human ...
    Jun 3, 2019 · Using single-cell RNA-Seq, we show that existing approaches to stratify bone marrow CD34+ cells reveal a hierarchically-structured ...
  25. [25]
    [PDF] CD34-POSITIVE CELLS - Haematologica
    CFU-GEMM contain prec u rs or cl on ogen i c cells of both myel oid and eryt h roid lineage s and ex press CD34, HLA-DR, CD38, CD117 and CD45RA. Th ey also ex ...
  26. [26]
    Prospective isolation of human clonogenic common myeloid ... - NIH
    ... CFU-GEMM), GMPs exclusively gave rise to granulocyte/macrophage colonies ... common myeloid progenitor. GMP. granulocyte/macrophage progenitor. MEP.
  27. [27]
    Notch signaling in mammalian hematopoietic stem cells - PMC - NIH
    This review will focus on the role of Notch signaling in HSCs. Although it is known that Notch is required to establish the earliest embryonic HSCs.
  28. [28]
    Myeloid Lineage Commitment from the Hematopoietic Stem Cell
    Jun 22, 2007 · ... Akashi et al., 2000). The CMP differentiates into the GMP and the MEP. The CMP can generate all types of myeloid colonies, whereas the GMP ...
  29. [29]
    C/EBPα binds and activates the PU.1 distal enhancer to induce ...
    These results suggest that induction of PU.1 by C/EBPα plays a central role in the CMP to GMP transition and in subsequent commitment to the monocyte lineage.
  30. [30]
    Stem Cell Factor Directly Stimulates the Development of Enriched ...
    SCF can directly act on highly enriched committed progenitor cells in serum-deprived conditions to promote survival, proliferation, and development.Missing: GEMM | Show results with:GEMM
  31. [31]
    Human Granulocyte-Macrophage Colony-Stimulating Factor (GM ...
    IL-3 and GM-CSF as single factors were equally active in stimulating CFU-GEMM, but the combination of both factors produced additive stimulative effects upon ...
  32. [32]
    Evolving insights into the synergy between erythropoietin and ... - NIH
    ... EPO, G-CSF, GM-CSF and TPO). These investigators also confirmed the EPO/TPO synergy that we had shown on more mature progenitor cells, and using single cell ...
  33. [33]
    Thrombopoietin promotes mixed lineage and megakaryocytic colony ...
    The role of thrombopoietin (Tpo) in promoting hematopoiesis has been extensively studied in late fetal, neonatal, and adult mice.Materials And Methods · Results · Tpo And C-Mpl Are Expressed...<|separator|>
  34. [34]
    Growth characteristics and expansion of human umbilical cord blood ...
    SLF plus erythropoietin (Epo) plus interleukin 3 (IL-3) enhanced detection of cord blood multipotential (CFU-GEMM) progenitors 15-fold compared to stimulation ...
  35. [35]
    Transforming growth factor beta selectively inhibits normal ... - PubMed
    In the presence of GM-CSF or IL-3, optimal concentrations of TGF-beta (400 pmol/L) inhibited colony formation by erythroid (BFU-E), multipotential (CFU-GEMM), ...
  36. [36]
    Ex vivo HSC expansion challenges the paradigm of unidirectional ...
    Jun 14, 2019 · Ex vivo expansion of UCB-CD34+ cells with UM171. UM171, a pyrimidoindole derivative, is another potent small molecule that triggers a robust ex ...Missing: CFU- post-
  37. [37]
    Regulation of early human hematopoietic (BFU-E and CFU-GEMM ...
    Both human monocyte conditioned medium (MCM) and human recombinant IL-1 (hrIL-1) induced fibroblasts to produce stimulators of BFU-E and CFU-GEMM in a dose- ...
  38. [38]
    Recent advances in engineering hydrogels for niche biomimicking ...
    A CFU assay showed SCF-loaded HGDN hydrogels had more primitive CFU-GEMM (granulocyte, erythrocyte, monocyte, megakaryocyte) phenotype compared to the cells ...
  39. [39]
    Megakaryocytic Progenitors Can Be Generated Ex Vivo and Safely ...
    In a fibrin-clot assay, 1 to 3 × 105 nucleated cells or 5 to 20 × 103 purified CD34+ cells in IMDM were resuspended in a solution of 10% L-asparagine, 10 ...
  40. [40]
    Pluripotent hemopoietic progenitors (CFU‐GEMM) in chronic ...
    The frequency ranged from 1-36 per 2 × 105 mononuclear cells of density less than 1.077 gm/ml. The requirements for PHA-LCM and EPO varied for patients with ...<|separator|>
  41. [41]
    Current practices and Prospects for Standardization of the ...
    The CFU assay takes advantage of a hematopoietic progenitor's ability to proliferate and differentiate to form a colony of cells committed to specific blood ...Missing: capacity | Show results with:capacity
  42. [42]
    Frequently Asked Questions on Performing the CFU Assay
    Yes, the scoring of CFU assays can be subjective. Some of the most common and particularly challenging phenomena encountered when scoring CFU colonies is the ...
  43. [43]
    ROLE OF HAEMATOPOIETIC GROWTH FACTORS IN MEDICINE
    Jun 26, 2017 · G-CSF stimulates precursor cells and shortens the time required for maturation and release into the circulation. It decreases the incidence of ...Granulocyte/macrophage... · Granulocyte Colony... · Colony Stimulating Factor-1...Missing: TPO | Show results with:TPO
  44. [44]
    Role of Erythropoietin and Other Growth Factors in Ex Vivo ...
    Oct 2, 2014 · Erythropoietin (EPO) is a key regulator in most ex vivo protocols along with other growth factors such as SCF, IL-3, IGF-1, and Flt-3.<|separator|>
  45. [45]
    CFU-GEMM - an overview | ScienceDirect Topics
    CFU-GEMM refers to a common progenitor cell involved in the proliferation and differentiation of megakaryocytes, which are responsible for platelet production.
  46. [46]
    Protein Segregation Between Dividing Hematopoietic Progenitor ...
    Mar 28, 2012 · In the present study, we investigated how the symmetry/asymmetry of cell division in mitotic CD34+ cells can be evaluated by determining the ...
  47. [47]
    A common bipotent progenitor generates the erythroid and ...
    Aug 20, 2009 · (A) CD41 and CD42 flow cytometry analysis. Expression of CD41 and CD42 MK differentiation markers, shown, respectively on the y-axis and x-axis ...
  48. [48]
    A system-level model reveals that transcriptional stochasticity is ...
    Dec 5, 2024 · Perhaps hypoxia is a control factor to keep the plasticity of HSC differentiation limited, in order to provide physiological stability to the ...
  49. [49]
    Inflamm-Aging of Hematopoiesis, Hematopoietic Stem Cells, and the ...
    (B) Inflammation enhances myeloid lineage production, including myeloid progenitors and mature myeloid cells, leading to myeloid bias in hematopoiesis. (C) ...Missing: plasticity | Show results with:plasticity
  50. [50]
    Cbfβ-SMMHC induces distinct abnormal myeloid progenitors able to ...
    The acute myeloid leukemia (AML)-associated CBFβ-SMMHC fusion protein impairs hematopoietic differentiation and predisposes to leukemic transformation.
  51. [51]
    The assessment of the hematopoietic reservoir after ... - PubMed
    Burst forming unit-erythroid (BFU-E) and colony forming unit-granulocyte erythroid megakaryocyte macrophages (CFU-GEMM) numbers were reduced when compared with ...
  52. [52]
    Bone marrow MSCs in MDS: contribution towards dysfunctional ...
    Dec 21, 2018 · Frequencies for CFU-BFU were 2.9 ± 0.3 vs 14.3 ± 5.1, those for CFU-GM were 17.2 ± 1.2 vs. 39.3 ± 2.3 and those for CFU-GEMM were 6.6 ± 0.9 vs.
  53. [53]
    Impairment of GM‐CSF production in myelodysplastic syndromes
    Moreover, the number of multipotent (CFU-GEMM), granulocyte/macrophage (CFU-GM) and erythroid (BFU-E) progenitors was significantly impaired in myelodysplastic ...
  54. [54]
    Effect of intensified chemotherapy on the pluripotent haematopoietic ...
    At diagnosis, the number of CFU-GEMM/ml bone marrow aspirate was significantly reduced to 12.1 (95% confidence interval 2.1-70) as compared to healthy controls ...
  55. [55]
    RUNX1 mutations enhance self-renewal and block granulocytic ...
    Feb 1, 2019 · Key Points. RUNX1 mutations lead to a block in granulocytic differentiation that can be partially rescued by CEBPA overexpression.
  56. [56]
    Modeling Down Syndrome Myeloid Leukemia by Sequential ... - MDPI
    Feb 11, 2022 · Transcriptome analysis showed that GATA1 mutation resulted in downregulation of megakaryocytic and erythrocytic differentiation pathways and ...
  57. [57]
    Cord blood expansion. Pyrimidoindole derivatives are agonists of ...
    Sep 19, 2014 · The properties of UM171 make it a potential candidate for hematopoietic stem cell transplantation and gene therapy. Copyright © 2014 ...Missing: CFU- GEMM marrow engraftment
  58. [58]
    Pyrimidoindole derivatives are agonists of human hematopoietic ...
    Sep 19, 2014 · The properties of UM171 make it a potential candidate for hematopoietic stem cell transplantation and gene therapy. ... Bone Marrow Transplant.
  59. [59]
    Advances in ex vivo expansion of hematopoietic stem and ... - Frontiers
    In this review, we comprehensively view hematopoietic stem cells as therapeutic assets, going from fundamental to translational.
  60. [60]
    Correction of β-thalassemia major by gene transfer in ...
    Gene therapy, based on autologous transplantation of genetically corrected hematopoietic stem cells (HSCs), represents a promising alternative for patients ...
  61. [61]
    Fetal gene therapy of α-thalassemia in a mouse model - PNAS
    In this study, we investigated the efficacy of direct in utero delivery of a lentiviral-based human a-globin gene to a mouse model of α-thalassemia.Fetal Gene Therapy Of... · Results · Erythroid-Specific...
  62. [62]
    Human multipotential progenitor cells (CFU-GEMM) have ... - PubMed
    This study found a much higher replating efficiency for CB CFU-GEMM than previously reported, in terms of the percentage of colonies that could be replated.
  63. [63]
    Development of a gene edited next-generation hematopoietic cell ...
    To unlock the full potential of AML therapies, we used CRISPR-Cas9 to genetically ablate the myeloid protein CD33 from healthy donor hematopoietic stem and ...
  64. [64]
    Decoding functional hematopoietic progenitor cells in the adult ...
    May 1, 2025 · Here, in the adult human lung, we show that functional hematopoietic precursors reside in the extravascular spaces with a frequency similar to ...
  65. [65]
    GMP-like and MLP-like Subpopulations of Hematopoietic Stem ... - NIH
    Apr 29, 2025 · The majority of adult AML patients will experience relapse within the first two years from diagnosis, which makes relapse prevention an ...