Fact-checked by Grok 2 weeks ago

Stem cell factor

Stem cell factor (SCF), also known as kit ligand or steel factor, is a pleiotropic cytokine and growth factor that binds to and activates the receptor tyrosine kinase c-Kit (encoded by the KIT gene), playing a critical role in the survival, proliferation, migration, and differentiation of various cell types, including hematopoietic stem and progenitor cells, melanocytes, germ cells, and mast cells. SCF was identified in the late 1980s through genetic studies in mice, where it corresponds to the product of the Sl (steel) locus on chromosome 10, complementing the W locus that encodes c-Kit; the human SCF gene is located on chromosome 12q14.3-q21 and was cloned in 1990. The protein is synthesized as a 248-amino-acid transmembrane precursor that undergoes alternative splicing to produce either a membrane-bound form (lacking exon 6, ~220 amino acids) or a soluble form (including exon 6, which allows proteolytic cleavage by matrix metalloproteinases). Both forms function as noncovalently linked homodimers, with the soluble variant having a molecular weight of approximately 18.5 kDa (glycosylated to 28–40 kDa) and stabilized by intramolecular disulfide bonds. Upon binding to c-Kit, a type III featuring five extracellular immunoglobulin-like domains, a single transmembrane helix, and an intracellular split kinase domain, SCF induces receptor dimerization, autophosphorylation, and activation of downstream signaling pathways such as PI3K/Akt, MAPK/ERK, and PLCγ, which collectively promote , , and anti-apoptotic effects. SCF is primarily produced by stromal cells, endothelial cells, fibroblasts, and , with expression tightly regulated during development and in response to stress, such as or tissue injury; circulating soluble SCF levels in human serum average around 3.3 ng/mL. In hematopoiesis, SCF is indispensable for the maintenance and expansion of primitive hematopoietic stem cells, synergizing with other cytokines like erythropoietin, IL-3, G-CSF, and GM-CSF to enhance progenitor cell proliferation and colony formation, while its absence leads to severe macrocytic anemia and impaired erythropoiesis. Beyond blood cell development, SCF/c-Kit signaling supports melanocyte migration and pigmentation (mutations cause piebaldism), primordial germ cell survival and fertility, mast cell maturation and degranulation in immune responses, interstitial cells of Cajal for gastrointestinal motility, and neuronal precursor differentiation. Clinically, dysregulated SCF/c-Kit signaling contributes to pathologies including mast cell disorders like systemic mastocytosis, gastrointestinal stromal tumors (GISTs, where activating KIT mutations occur in 80–85% of cases), acute myeloid leukemia, and melanoma; targeted tyrosine kinase inhibitors such as imatinib effectively treat many of these by blocking c-Kit activation, though resistance via secondary mutations like D816V remains a challenge. Recombinant SCF has been explored in clinical trials for stem cell mobilization and hematopoietic recovery post-chemotherapy, demonstrating synergy with G-CSF but limited by potential toxicity like mast cell activation.

Molecular Structure and Production

Gene and Protein Characteristics

The KITLG , which encodes stem cell factor (SCF), is located on the long arm of human at position 12q21.32. In mice, the orthologous resides at the Steel (Sl) locus on chromosome 10. The human KITLG spans approximately 88 kb and consists of 10 s separated by 9 introns, with producing at least two major transcript variants. Transcript variant 1 (NM_003994.6) encodes the membrane-bound isoform and lacks exon 6, resulting in a 9-exon structure, while transcript variant 2 (NM_000899.5) includes exon 6, which encodes a proteolytic cleavage site, yielding the soluble isoform. The mouse Kitl exhibits a similar organization, with 9-10 s depending on the splice variant, reflecting conserved genomic architecture across mammals. The SCF protein is synthesized as a precursor polypeptide that undergoes processing to yield the mature form. The soluble isoform precursor comprises 273 amino acids, which is cleaved to produce the 165-amino-acid mature protein with a molecular weight of approximately 18.5 kDa. The membrane-bound isoform precursor is 245 amino acids long, processed to a 220-amino-acid transmembrane protein. Mature SCF functions as a noncovalent homodimer, with each monomer adopting an antiparallel four-helix bundle fold stabilized by two intramolecular disulfide bonds: Cys4–Cys89 and Cys43–Cys138. Additionally, the protein features three N-linked glycosylation sites at Asn65, Asn93, and Asn120, which contribute to its stability and bioactivity, though glycosylation is not strictly required for receptor binding. The crystal structure of the functional core of recombinant human SCF, determined at 2.3 Å resolution (PDB entry 1EXZ), reveals a noncovalent homodimeric assembly of two identical protomers, each comprising residues 1–141. The dimeric core is characterized by extensive polar and nonpolar interactions at the interface, where a loop from one monomer (residues 61–72) inserts into a groove between helices 1 and 4 of the adjacent monomer, forming a helix-loop-helix motif critical for dimer stability and subsequent receptor engagement. This structural arrangement positions hydrophobic crevices and charged regions on the protomer surfaces as key sites for interaction with the c-KIT receptor. In human , soluble SCF circulates at an average concentration of 3.3 ± 1.1 ng/mL, supporting basal hematopoietic maintenance. The KITLG gene and SCF protein exhibit high evolutionary conservation across mammals, with sequence identities exceeding 80% between human SCF and orthologs in species such as , , and bovine, underscoring its fundamental role in developmental processes.

Isoforms and Processing

Stem cell factor (SCF), also known as KIT ligand, is generated through of its primary transcript, yielding two principal isoforms: a full-length transmembrane form consisting of 220 and a precursor form of 273 that can be processed into a soluble isoform. The transmembrane isoform results from the exclusion of 6 during splicing, which removes the proteolytic cleavage site and anchors the protein in the via a hydrophobic and a short cytoplasmic tail. In contrast, inclusion of 6 in the precursor isoform introduces the cleavage site, enabling subsequent release of the soluble form comprising the first 165 of the extracellular . The precursor isoform undergoes proteolytic processing primarily by matrix metalloproteinases, including ADAM17, which cleave the protein near Pro165 at the bond between residues 165 and 166 within the stalk region adjacent to the . This ectodomain shedding is stimulated by agents such as phorbol esters and occurs with distinct kinetics, where ADAM17 predominates in regulated release; the resulting soluble SCF fragment retains full bioactivity. The transmembrane isoform is resistant to such cleavage due to the absence of the recognition site, maintaining its membrane-bound state unless under extreme conditions. These isoforms exhibit complementary functions, with the transmembrane form promoting direct cell-cell and localized juxtacrine signaling essential for processes like , while the soluble form facilitates paracrine diffusion for broader effects such as hematopoietic stimulation. Studies in models underscore their non-redundant roles: animals expressing only the transmembrane isoform (e.g., Sl^d mutants lacking 6) develop severe due to insufficient soluble SCF for , whereas models restricted to soluble SCF alone display sterility from impaired primordial development and survival, highlighting the necessity of both for complete physiological activity. Structural analyses confirm the soluble isoform's stable non-covalent dimerization, critical for receptor activation, with recent cryo-EM studies of SCF-KIT complexes (2023) revealing no substantial deviations from the canonical head-to-head dimeric architecture established by the 2000 of the active core, thereby affirming long-term dimer stability across solution and complexed states.

Receptor Interaction and Signaling

The c-KIT Receptor

The c-KIT receptor, also designated CD117, belongs to the class III family of receptor tyrosine kinases and is encoded by the KIT proto-oncogene located on the long arm of human at position 4q12. This spans approximately 83 kb and consists of 21 exons, producing a 145 kDa transmembrane . The receptor's structure includes an N-terminal extracellular ligand-binding domain comprising five immunoglobulin-like folds (D1–D5), where the first three domains (D1–D3) primarily mediate interactions with stem cell factor (SCF); a single α-helical transmembrane segment of about 23 ; and an intracellular portion with a juxtamembrane region, an ATP-binding domain interrupted by a kinase insert, and a C-terminal regulatory tail. Binding of SCF to the extracellular of c-KIT induces receptor dimerization, which stabilizes the active conformation and triggers trans-autophosphorylation of multiple residues in the cytoplasmic , including Y721 (which recruits PI3K) and Y730 (which binds PLCγ). This ligand-dependent activation is characterized by high , with a (Kd) of approximately 2 nM for soluble monomeric SCF, though dimeric membrane-bound SCF exhibits even higher due to bivalent engagement. The dimerization process involves SCF's dimeric bridging two c-KIT molecules, relieving autoinhibitory constraints in the kinase and enabling ATP-dependent phosphorylation. c-KIT exhibits a restricted expression pattern, with high levels observed on hematopoietic stem and progenitor cells, mast cells, melanocytes, and primordial germ cells, where it supports key developmental and migratory functions. Gain-of-function mutations in the KIT gene, particularly in exons 11 and 13, are prevalent in gastrointestinal stromal tumors (GIST) and promote constitutive, ligand-independent dimerization and kinase activation, driving oncogenesis without SCF stimulation. These mutations often occur in the juxtamembrane or kinase domains, enhancing autophosphorylation and signaling. Evolutionarily, the SCF/c-KIT ligand-receptor pair represents an ancient signaling module conserved from invertebrates to mammals, with the receptor belonging to a family of type III RTKs that arose through gene duplication events. The interaction has evolved a distinctive recognition mode, involving SCF's helix-loop-helix dimer interfacing with c-KIT's D1–D2 domains in a manner distinct from other growth factor-receptor complexes, such as PDGF/PDGFR. Species differences are subtle but notable; for instance, human SCF features an asparagine at position 10 in the receptor-binding loop, which is conserved across mammals but can vary to aspartate in some rodents, influencing binding kinetics without altering core pairing specificity. This activation of c-KIT upon SCF binding initiates diverse downstream signaling cascades, including MAPK/ERK and PI3K/AKT pathways.

Downstream Signaling Mechanisms

Upon binding of stem cell factor (SCF) to the c-KIT receptor, autophosphorylation of specific residues initiates multiple downstream signaling cascades that regulate , , and . Key sites include Tyr-721, which directly recruits the p85 regulatory subunit of 3-kinase (PI3K), leading to its activation and production of phosphatidylinositol-3,4,5-trisphosphate (PIP3). This, in turn, activates (AKT) through at Thr-308 and Ser-473, promoting anti-apoptotic effects by phosphorylating Bad at Ser-136 and , thereby enhancing expression of factors like and . Adapter proteins such as GAB2, phosphorylated by family kinases, provide an indirect route for PI3K activation, amplifying the pathway in certain contexts. The (MAPK)/extracellular signal-regulated kinase (ERK) pathway is engaged primarily through at Tyr-568/570 and Tyr-703, recruiting family kinases and the Grb2-Sos complex to activate Ras-Raf-MEK-ERK signaling. This cascade facilitates changes associated with , with Shc serving as an additional to enhance ERK1/2 via . In parallel, the /signal transducer and activator of transcription (JAK/STAT) pathway is activated, particularly STAT3 and STAT5, often requiring JAK2 association for and translocation, enabling with other cytokines. Cγ (PLCγ) binds to Tyr-730, hydrolyzing PIP2 into inositol-1,4,5-trisphosphate (IP3) and diacylglycerol (DAG), which mobilizes intracellular calcium and activates , contributing to cytoskeletal rearrangements and proliferation. Negative regulation maintains signaling fidelity, with SHP1 dephosphorylating key sites like Tyr-569 to attenuate PI3K and MAPK activation, forming loops that prevent excessive responses. Cross-talk with other receptors, such as the , enhances pathway synergy; for instance, SCF/c-KIT signaling cooperates with erythropoietin-induced IRS2 expression to amplify AKT and ERK outputs via IGF1R engagement. Quantitative differences arise from SCF presentation: transmembrane SCF sustains longer c-KIT and stronger PLCγ activation compared to soluble SCF, which induces transient signals favoring PI3K over PLCγ, as evidenced by slower kinetics and prolonged downstream ERK activity with membrane-bound forms. Recent studies highlight ERK's role in , where SCF/c-KIT-mediated ERK activation influences histone modifications to regulate fate decisions, such as through p38/ERK axis modulation of epigenetic marks in responsive cells.

Biological Roles

In Hematopoiesis

Stem cell factor (SCF) plays a critical role in supporting hematopoietic stem and progenitor cells (HSPCs) within the by promoting their self-renewal, survival, and mobilization. It acts as an essential survival factor that prevents and facilitates of primitive hematopoietic cells, often in with other cytokines such as interleukin-3 (IL-3), which can result in up to 100-fold of progenitor populations . This synergistic effect underscores SCF's importance in amplifying HSPC pools during stress or protocols. In the bone marrow niche, SCF is selectively secreted by arterial endothelial cells, which provide a supportive microenvironment for HSPC maintenance and differentiation. A 2018 study demonstrated that these cells express high levels of membrane-bound SCF, essential for retaining HSPCs in perivascular regions. Recent updates from 2024 highlight co-expression of SCF with in vascular endothelial cells, further stabilizing HSPC localization and function within the niche. Notably, fetal HSPCs exhibit approximately sixfold greater responsiveness to SCF compared to HSPCs, despite equivalent c-KIT receptor levels, contributing to the expansion observed during embryonic hematopoiesis. Genetic studies of SCF-null mice reveal profound defects in hematopoiesis, including severe , hypocellularity, and near-complete absence of erythroid and myeloid progenitors due to impaired HSPC survival and . These phenotypes confirm SCF's indispensable role in multilineage production from early developmental stages. Advancements from 2023 to 2025 have emphasized SCF's involvement in HSPC for transplantation, particularly through non-genotoxic strategies targeting the SCF/c-KIT axis. Antibody-drug conjugates against c-KIT enable safer and depletion of endogenous HSPCs, facilitating engraftment of donor cells without chemotherapy-induced toxicity, as shown in preclinical models and early clinical trials.

In Development and Tissue Maintenance

Stem cell factor (SCF) plays a crucial role in embryonic development by facilitating the migration of primordial germ cells (PGCs) to the gonadal ridges through its interaction with the c-KIT receptor expressed on PGCs and gonadal somatic cells. This guidance ensures proper colonization of the developing gonads, with disruptions in SCF/c-KIT signaling leading to impaired PGC survival and migration. Similarly, SCF supports the migration and colonization of precursors from the to the skin and hair follicles, promoting their proliferation and differentiation into pigment-producing cells essential for coat coloration. In ovarian development, SCF induces the formation and progression of primordial follicles by acting on oocytes and surrounding granulosa cells, initiating through enhanced cell survival and growth. In adult tissues, SCF maintains survival and proliferation in the testes via from Sertoli cells, supporting continuous and preventing . It also sustains skin pigmentation by regulating homeostasis in the and hair follicles, where SCF deficiencies result in . In mice with the Sl/Sl^d genotype, mutations in the SCF cause white spotting due to failed migration and survival during , leading to unpigmented patches on the coat. Beyond reproductive and integumentary systems, SCF is vital for the development and maintenance of (ICCs) in the , where SCF/c-KIT signaling coordinates pacemaker activity and smooth muscle coordination to regulate gut . Recent studies highlight SCF's involvement in bone regeneration, where it promotes the differentiation and survival of progenitors in the skeletal niche, enhancing repair and renewal through targeted paracrine effects. In wound healing, inhibition of soluble SCF isoforms accelerates mucosal repair in intestinal injuries by modulating and epithelial regeneration. SCF integrates with the Wnt/β-catenin pathway in niches to regulate regeneration, as SCF/c-KIT signaling in intestinal Paneth cells enhances renewal during , synergizing with Wnt to promote and repair. This underscores SCF's role in maintaining niche across diverse .

In Biology

Stem cell factor (SCF) plays a pivotal role in mast cell biology by interacting with the c-KIT receptor, which serves as a key surface marker on immature s and their progenitors. In humans, s arise from CD34-positive, c-KIT-positive hematopoietic progenitors in the and , where SCF binding to c-KIT drives and maturation of these precursors into functional s. This process is essential for mast cell lineage commitment, with KIT expression retained at high levels throughout mast cell development, distinguishing immature cells from other hematopoietic lineages. SCF significantly enhances numbers ; for instance, of recombinant SCF in leads to a reversible expansion of cutaneous populations, with increases exceeding 100-fold at injection sites due to promoted and of progenitors. In rodent models, such as mice and rats, SCF similarly induces in tissues like the skin and mucosa, amplifying progenitor differentiation into mature s. Regarding survival and adhesion, the transmembrane form of SCF, produced by fibroblasts and other stromal cells, anchors mast cells to extracellular matrix components like fibronectin via c-KIT-mediated signaling, thereby supporting long-term survival and tissue localization. Soluble SCF, in contrast, promotes mast cell survival by suppressing and enhances activation, potentiating IgE-dependent and the release of mediators such as , which contributes to allergic responses. This dual action of SCF isoforms ensures mast cell viability and responsiveness in inflammatory contexts. Pathophysiologically, SCF overexpression is linked to increased numbers and activation in conditions like urticaria and . In asthmatic airways, SCF levels are elevated, correlating with hyperplasia and enhanced degranulation, which is reversible with therapy. Similarly, in urticaria, upregulated SCF contributes to dermal accumulation and mediator release, exacerbating wheal formation and itch. Recent advancements (2023–2025) highlight KIT inhibitors targeting SCF signaling for therapeutic intervention in chronic urticaria. For example, the anti-KIT barzolvolimab, in phase 3 trials for , depletes s by blocking SCF/c-KIT interactions, yielding sustained improvements in urticaria control and reduced disease activity with manageable side effects like mild infusion reactions. Species differences underscore varying SCF dependency in mast cell maturation: human mast cells exhibit an absolute requirement for SCF, lacking support from interleukin-3 alone, whereas mast cells can mature with interleukin-3, though SCF potently enhances their development and survival.

Clinical and Pathological Implications

Therapeutic Applications

(SCF) plays a key role in cocktails for the expansion of hematopoietic stem cells (HSCs), often combined with FLT3 ligand (FLT3L) and thrombopoietin (TPO) to enhance and maintain repopulating capacity. In cultures of cord blood-derived HSCs, the combination of SCF, FLT3L, and TPO achieves up to 241-fold expansion of total nucleated cells while preserving multilineage engraftment potential in /SCID models, supporting short- and long-term reconstitution comparable to unexpanded cells. Similarly, co-culture with mesenchymal cells and these s yields 74-fold increases in +/- primitive cells, improving long-term culture-initiating cell output by over 5-fold and addressing limitations in blood transplantation volumes. These expanded HSCs demonstrate enhanced engraftment rates in preclinical models, with SCF and FLT3L alone maintaining 64% long-term engraftment in murine transplants after 72-hour culture, facilitating faster hematopoietic recovery post-transplant. In ischemia therapies, recombinant SCF (Ancestim) has been explored to mobilize s and promote cardiac repair following , primarily through and recruitment. show that SCF administration reduces infarct volume by up to 50% and improves left ventricular function via c-KIT-mediated endothelial and activation, enhancing neovascularization in ischemic tissues. Clinical trials combining Ancestim with G-CSF for stem cell mobilization in acute patients demonstrated feasibility and safety in early phases, with interim reports indicating potential for improved cardiac outcomes through mobilized cell homing. However, development was halted due to adverse effects, including urticaria and reactions, as noted in 2024 retrospectives reviewing cytokine-based regenerative approaches. Recent applications in regenerative medicine leverage SCF supplementation to activate ovarian follicles in reproductive disorders, promoting primordial follicle development via c-KIT signaling. In mouse models, SCF treatment accelerates primordial follicle activation and oocyte growth, increasing follicle survival and maturation rates through PI3K/AKT pathway stimulation, with potential translation to human infertility therapies. A 2022 study in aged mice demonstrated that SCF promotes primordial follicle activation via PI3K/AKT/mTOR signaling, enhancing follicle recruitment in ovarian failure models. For bone repair, targeting the SCF/c-KIT axis enhances osteogenesis by recruiting c-KIT+ progenitors to fracture sites, accelerating healing through Lnk-dependent signaling that boosts endochondral ossification. A 2024 review emphasizes SCF's therapeutic potential in bone renewal, with targeted delivery improving progenitor mobilization and tissue regeneration in preclinical defect models without systemic side effects. Emerging therapies also target SCF inhibition for mast cell-related disorders. As of 2025, CDX-622 (Celldex Therapeutics) is in Phase 1 trials, neutralizing SCF to reduce activation in inflammatory conditions like . Similarly, Jasper Therapeutics is developing SCF/c-Kit pathway blockers for systemic . The global SCF market is projected to grow at a compound annual growth rate (CAGR) of 11.4% through 2030, driven by expanding applications in biotechnology for HSC expansion and regenerative therapies.

Disease Associations

Dysregulation of stem cell factor (SCF), through genetic defects or overexpression, is implicated in various pathological conditions. In mice, hypomorphic alleles of the SCF gene, known as Steel mutations (e.g., Sl/Sld and Sl17H), lead to semidominant phenotypes including severe macrocytic anemia due to impaired erythropoiesis, sterility from defects in germ cell migration and survival, and depigmentation from melanocyte deficiencies. These mutations disrupt SCF production or processing, resulting in reduced ligand availability for the c-KIT receptor, which is essential for hematopoietic, gonadal, and pigment cell development. In humans, piebaldism—a congenital disorder characterized by leukoderma and white forelock—arises from dominant-negative or loss-of-function mutations in the KIT gene, the receptor for SCF, effectively mimicking SCF deficiency and causing impaired melanocyte migration and survival without affecting hematopoiesis or fertility. Overexpression of SCF contributes to oncogenesis by forming autocrine or paracrine loops with c-KIT in several malignancies. In gastrointestinal stromal tumors (GIST), co-expression of SCF and c-KIT within tumor cells promotes ligand-independent activation and tumor growth, as evidenced by SCF detection in primary GIST lesions. Similarly, in (AML), SCF/c-KIT sustains and of blasts, with KIT expression in up to 50% of cases driving oncogenic pathways. In systemic mastocytosis, activating KIT mutations (e.g., D816V) combined with elevated SCF levels lead to clonal expansion and tissue infiltration. Recent 2023 single-cell analyses have further revealed that SCF-mediated remodeling of the niche, including stromal alterations, facilitates progression by altering hematopoietic stem and progenitor cell (HSPC) interactions and promoting inflammatory microenvironments. Elevated SCF levels are associated with allergic and inflammatory disorders, particularly those involving accumulation. In , increased cutaneous SCF expression correlates with disease severity, enhancing survival and through c-KIT binding. In , SCF upregulation in airway and promotes hyperplasia and eosinophil recruitment, exacerbating bronchial inflammation, with levels decreasing after glucocorticoid therapy. A 2024 study highlighted SCF's role in , where it induces ERK pathway activation in tumor cells, driving epithelial-mesenchymal transition and via c-KIT signaling. SCF dysregulation also links to other conditions, including reproductive and renal pathologies. SCF deficiency contributes to by impairing primordial follicle development and survival, as seen in Steel mutant mice with ovarian failure; human parallels suggest similar mechanisms in premature ovarian insufficiency.

Molecular Interactions

Protein-Protein Interactions

Stem cell factor (SCF), also known as KIT ligand, primarily exerts its effects through high-affinity to the extracellular domain of the . This interaction occurs via a dimeric SCF structure that engages two c-KIT molecules, forming a 2:2 complex essential for receptor dimerization and . The interface is divided into three main sites: Site I in the D1 domain of c-KIT involves the αC-β2 loop of SCF; Site II spans the D2 domain and D2-D3 linker, featuring electrostatic interactions between basic residues in c-KIT (e.g., Arg122, Arg181) and acidic residues in SCF; and Site III in the D3 domain includes the N-terminal segment of SCF. Structural analyses reveal that loops in the SCF dimer, such as the extended loop (residues 95–104) and flapping loop, undergo conformational changes upon , contributing to a buried surface area exceeding 1,800 Ų across sites and enabling stable, high-affinity association with dissociation constants (K_d) typically in the low nanomolar range (e.g., 0.2–2 nM for wild-type SCF). In addition to direct receptor engagement, SCF interacts with proteoglycans (HSPGs) on surfaces, which modulate its presentation and bioavailability in the . These chains, particularly those with specific sulfation patterns (N-, 2-O-, and 6-O-sulfation), bind SCF and facilitate its localization near c-KIT-expressing , enhancing signaling efficiency by creating concentration gradients and protecting SCF from proteolytic degradation. This modulation is critical in hematopoietic niches, where HSPGs on stromal amplify SCF's role in maintenance without altering the core binding affinity to c-KIT. Cleaved, soluble forms of SCF, generated by proteolytic processing of the membrane-bound isoform, indirectly promote by activating downstream pathways that enhance integrin avidity. For instance, soluble SCF binding to c-KIT upregulates the adhesive function of such as α4β1 and α5β1 on hematopoietic and cells, facilitating interactions with components like in a dose- and time-dependent manner, though SCF itself does not directly bind . Several inhibitors target SCF-c-KIT interactions at the protein level. Monoclonal antibodies like ACK2 bind the extracellular domain of c-KIT, sterically blocking SCF access to its high-affinity sites and preventing receptor dimerization, as demonstrated in both and models of hematopoiesis and function. In contrast, small-molecule inhibitors such as bind the intracellular domain of c-KIT after SCF-induced dimerization, inhibiting ATP binding and autophosphorylation to disrupt downstream signaling without affecting the initial ligand-receptor interaction (IC_{50} ≈ 100 nM for c-KIT activity). Recent biophysical studies using (SPR) have quantified affinity variations among SCF isoforms and variants. For example, soluble monomeric SCF exhibits lower affinity (K_d ≈ 10–20 nM) compared to the dimeric form (K_d ≈ 0.1–1 nM), with engineered variants showing up to 3.7-fold improvements in binding kinetics due to optimized loop interactions; these differences influence signaling potency in isoform-specific contexts.

Functional and Genetic Interactions

Stem cell factor (SCF), also known as , primarily exerts its effects through binding to the c-Kit, initiating a cascade of downstream signaling that interacts with multiple pathways to regulate , , , and . Upon binding, SCF induces c-Kit dimerization and autophosphorylation on specific residues, recruiting adaptor proteins such as , Shc, and GAB2, which activate the /MAPK/ERK pathway for proliferation and the PI3K/Akt pathway for and anti-apoptosis (e.g., via of Bad at Ser-136). These interactions are modulated by Src family kinases (SFKs) binding to Tyr-568/570, enhancing ERK1/2 and JNK activation, while phospholipase C-γ (PLC-γ) at Tyr-730 generates diacylglycerol (DAG) to promote calcium mobilization and proliferation. Additionally, c-Cbl interacts with Tyr-568/936 to downregulate signaling via receptor , preventing excessive activation. Functionally, SCF/c-Kit signaling synergizes with other cytokines to amplify cellular responses. For instance, SCF cooperates with (EPO) through their respective receptors to enhance erythroid colony formation by integrating JAK2/STAT5 and MAPK pathways, essential for maturation. Similarly, SCF interacts with (GM-CSF) and (TNF) to promote development from hematopoietic progenitors, where SCF supports survival while GM-CSF drives . In angiogenesis, SCF/c-Kit signaling intersects with Gαi1 and Gαi3 G-proteins to promote endothelial and tube formation, highlighting its role in vascular remodeling. These functional partnerships underscore SCF's pleiotropic effects, where signaling amplitude tunes outcomes. Genetically, SCF and c-Kit genes exhibit strong interactions, as evidenced by mutations in mouse models. The c-Kit gene (W locus) and SCF gene (Sl locus) show epistatic relationships, where compound heterozygotes (W/Sl) display more severe phenotypes than single mutants, including profound , sterility, and lack of pigmentation due to additive defects in hematopoiesis, , and melanogenesis. Over 30 loss-of-function mutations in c-Kit, such as Wv (Thr660Met), cause and hematopoietic failure, while Sl mutations (e.g., Sl/Sld) disrupt SCF processing, leading to soluble isoform deficiencies that impair membrane-bound signaling critical for homing. In humans, gain-of-function c-Kit mutations (e.g., D816V in exon 17) drive and gastrointestinal stromal tumors (GISTs) by constitutive kinase activation, often interacting with local SCF expression to exacerbate oncogenesis. These genetic interactions highlight the ligand-receptor pair's coordinated role, with spectra influencing penetrance and therapeutic sensitivity (e.g., exon 11 mutations respond to , unlike exon 17 variants).

References

  1. [1]
  2. [2]
    Stem Cell Factor and Hematopoiesis | Blood - ASH Publications
    Aug 15, 1997 · This review will briefly discuss the production and structure of SCF and will focus on the physiologic role of SCF in hematopoiesis. The ...SCF STRUCTURE · SCF AND MAST CELLS · SCF AND HEMATOPOIETIC...
  3. [3]
    Crystal structure of human stem cell factor - PNAS
    Stem cell factor (SCF) plays important roles in hematopoiesis and the survival, proliferation, and differentiation of mast cells, melanocytes, ...Materials And Methods · Results And Discussion · A Receptor-Binding Region On...
  4. [4]
    4254 - Gene ResultKITLG KIT ligand [ (human)] - NCBI
    This gene encodes the ligand of the tyrosine-kinase receptor encoded by the KIT locus. This ligand is a pleiotropic factor that acts in utero in germ cell and ...Missing: structure exons
  5. [5]
    Stem cell factor is encoded at the Sl locus of the mouse ... - PubMed
    Oct 5, 1990 · Stem cell factor is encoded at the Sl locus of the mouse and is the ligand for the c-kit tyrosine kinase receptor.Missing: human 12q22- 12q24 Steel exon- intron structure alternative splicing
  6. [6]
    KITLG - Kit ligand - Homo sapiens (Human) - UniProt
    Ligand for the receptor-type protein-tyrosine kinase KIT. Plays an essential role in the regulation of cell survival and proliferation.Missing: exons intron<|separator|>
  7. [7]
    Implication for stem cell factor receptor dimerization and activation
    The soluble form of SCF has 165 amino acids ... Furthermore, M-CSF is a covalent homodimer linked by an intermolecular disulfide bond, whereas SCF is a ...
  8. [8]
    Structural basis for stem cell factor–KIT signaling and activation of ...
    Stem cell factor (SCF) binds to the extracellular domains of the KIT receptor tyrosine kinase (RTK) and plays a key role in diverse biological processes. KIT, ...
  9. [9]
    1EXZ: STRUCTURE OF STEM CELL FACTOR - RCSB PDB
    Jul 6, 2000 · In this report we describe the 2.3-A crystal structure of the functional core of recombinant human SCF. SCF is a noncovalent homodimer composed of two slightly ...Missing: loop- motifs
  10. [10]
    implication for stem cell factor receptor dimerization and activation
    In this report we describe the 2.3-A crystal structure of the functional core of recombinant human SCF. SCF is a noncovalent homodimer composed of two slightly ...Missing: resolution loop- motifs
  11. [11]
    Soluble stem cell factor in human serum - PubMed
    Soluble stem cell factor (SCF) is a proteolytically released form of SCF, active in early hematopoiesis, with an average serum level of 3.3 +/- 1.1 ng/mL. It ...Missing: evolutionary conservation mammals
  12. [12]
    Comparative genomics of human stem cell factor (SCF) - PMC - NIH
    We demonstrated the evolutionary view of SCF to find the closest organism to human by orthologous SCF. MATERIALS AND METHODS. The sequences of human SCF (Gene ...
  13. [13]
    Structure of the active core of human stem cell factor and ... - NIH
    Stem cell factor (SCF) is an early-acting hematopoietic cytokine that elicits multiple biological effects. SCF is dimeric and occurs in soluble and ...<|control11|><|separator|>
  14. [14]
    phorbol-ester-stimulated ectodomain shedding of Kitl1 by ADAM17 ...
    Proteolytic processing of transmembrane proteins to release their soluble extracellular domains from the membrane, a process termed ectodomain shedding, is ...
  15. [15]
    Cryo-EM analyses of KIT and oncogenic mutants reveal structural ...
    KIT molecules are expressed at the cell membrane as freely diffusing KIT monomers. Binding of SCF dimers to the ligand-binding region (D1–D3) of the ...
  16. [16]
    KIT gene: MedlinePlus Genetics
    Jul 13, 2021 · Genomic Location. The KIT gene is found on chromosome 4. Related Health Topics. Genes and Gene Therapy · Genetic Disorders. MEDICAL ENCYCLOPEDIA.
  17. [17]
    KIT KIT proto-oncogene, receptor tyrosine kinase [ (human)] - NCBI
    Aug 19, 2025 · The KIT gene encodes a receptor tyrosine kinase, a proto-oncogene, activated by stem cell factor (SCF), and involved in cell proliferation and ...
  18. [18]
    Structural basis for stem cell factor–KIT signaling and activation of ...
    Stem cell factor (SCF) binds to and activates the KIT receptor, a class III receptor tyrosine kinase (RTK), to stimulate diverse processes including ...
  19. [19]
    Structure of a c-Kit Product Complex Reveals the Basis for Kinase ...
    May 2, 2003 · The stem cell factor (SCF) receptor c-Kit is a type. III transmembrane RPTK comprised of five extracellular im- munoglobulin domains, a single ...<|control11|><|separator|>
  20. [20]
    The Strength and Cooperativity of KIT Ectodomain Contacts ...
    SCF binding induces KIT dimerization, which increases the local ... tyrosine autophosphorylation of unoccupied KIT (basal phosphorylation, Figure 6C).
  21. [21]
    Normal and Oncogenic Forms of the Receptor Tyrosine Kinase Kit
    SCF-induced STAT tyrosine phosphorylation has been associated with increased DNA binding [81, 82]. In addi- tion to tyrosine phosphorylation, SCF also induces ...
  22. [22]
    Characterization of a fusion protein composed of the extracellular ...
    Binding assay and cross-linking assay showed that the fusion protein retained high affinity for binding SCF (Kd = 0.3 nM). Addition of the chimeric protein ...
  23. [23]
    Affinities of SCF variants for c-Kit. The association and dissociation...
    Also, the affinity (K D value, 4 nM [27] or 570 pM [28]) between SCF and KIT is acceptable for a drug-delivery vector compared with therapeutic antibodies. SCF ...
  24. [24]
    Structural Basis for Activation of the Receptor Tyrosine Kinase KIT ...
    Stem Cell Factor (SCF) initiates its multiple cellular responses by binding to the ectodomain of KIT, resulting in tyrosine kinase activation.
  25. [25]
    KIT as a master regulator of the mast cell lineage - PMC
    Jun 1, 2023 · These include hematopoietic progenitors, melanocytes, germ cells, and mast cells ... c-kit receptor for stem cell factor that stimulates ...
  26. [26]
    A distinct expression pattern and point mutation of c-kit in papillary ...
    Apr 9, 2004 · KIT is expressed not only in tumors derived from hematopoietic stem cells, melanocytes, germ cells, mast cells, and interstitial cells of ...
  27. [27]
    KIT Activation Is a Ubiquitous Feature of Gastrointestinal Stromal ...
    In summary, we have shown oncogenic KIT gene mutations in 44 of 48 GISTs, and we found biochemical KIT activation in each of 25 GISTs. These data implicate KIT ...
  28. [28]
    A novel gain-of-function mutation of c-kit gene in gastrointestinal ...
    Mutations in the kinase domain both in c-kit and ret result in the ligand-independent and dimerization-independent activation of KIT and RET. 15 ...
  29. [29]
    KIT mutations and expression: current knowledge and new insights ...
    Feb 27, 2024 · Clinically, about 80-90% of treatment-naive GIST patients harbor primary KIT mutations, and special KIT-targeted TKI, imatinib (IM) showing ...
  30. [30]
    Review A holistic view on c-Kit in cancer: Structure, signaling ...
    Structure of c-Kit. The c-Kit receptor is encoded by a proto-oncogene present in the long-arm region of chromosome 4 (4q11–4q13) [30], spanning more than 34 ...
  31. [31]
    Epo-IGF1R cross talk expands stress-specific progenitors in ... - NIH
    Epo-induced IRS2 allows engagement of IGF1R signaling to expand a previously unrecognized progenitor population in erythropoietic stress.
  32. [32]
    Transmembrane stem cell factor protein therapeutics enhance ...
    May 6, 2022 · Stem cell factor (SCF) is a cytokine that regulates hematopoiesis and other biological processes. While clinical treatments using SCF would ...
  33. [33]
    Epigenetic histone modification by butyrate downregulates KIT and ...
    Aug 21, 2023 · Butyrate epigenetically modifies histones and downregulates the SCF/KIT/p38/Erk signalling axis, leading to the attenuation of MC function.
  34. [34]
    Stem cell factor as a survival and growth factor in human ... - PubMed
    Stem cell factor (SCF) preserves stem cell viability, influences cell cycle entry, facilitates proliferation, and prevents apoptotic death. It also stimulates ...
  35. [35]
    Expansion of human NOD/SCID-repopulating cells by stem cell ... - JCI
    The present study demonstrates that a combination of SCF, FL, TPO, and IL-6/sIL-6R stimulates the significant expansion of human HSCs capable of repopulating in ...
  36. [36]
    Stem cell factor is selectively secreted by arterial endothelial cells in ...
    Jun 22, 2018 · Stem cell factor is selectively secreted by arterial endothelial cells in bone marrow. Nat Commun 9, 2449 (2018). https://doi.org/10.1038 ...
  37. [37]
    Bone Marrow Niche in Cardiometabolic Disease: Mechanisms and ...
    Jan 30, 2025 · Vascular endothelial cells have been shown to produce critical factors for HSC maintenance, including CXCL (C-X-C motif ligand) 12 and SCF (stem ...Normal Hematopoiesis · The Stem Cell Niche · Hyperlipidemia And...<|control11|><|separator|>
  38. [38]
    Steel factor responsiveness regulates the high self-renewal ...
    Jun 1, 2007 · Abstract. Fetal hematopoietic stem cells (HSCs) regenerate daughter HSCs in irradiated recipients more rapidly than do adult HSCs.Missing: SCF | Show results with:SCF
  39. [39]
    Restricted Hematopoietic Progenitors and Erythropoiesis Require ...
    Jan 17, 2019 · Mice with a loss of Scf function have bone marrow hypocellularity, HSC depletion, and hypoplastic macrocytic anemia (Russell, 1979; Huang et al.
  40. [40]
    Fertility-preserving myeloablative conditioning using single-dose ...
    Oct 12, 2023 · The exploratory CD117-ADC targets CD117 (c-KIT), the receptor for stem cell factor (SCF) expressed primarily on HSCs and hematopoietic ...Results · Cd117-Adc Targets And... · Autologous Cd34+ Cell...
  41. [41]
    Targeted hematopoietic stem cell depletion through SCF-blockade
    Oct 29, 2024 · Robust phenotypic HSC depletion in the BM of treated mice was only observed with the antagonistic αCD117 + αCD110 mAb combination and the ...
  42. [42]
    advances and promises in the study of primordial germ cell migration
    Apr 12, 2024 · Another chemical cue commonly required in mammalian PGC migration and survival is kit ligand (KITL, also known as stem cell factor, or SCF, ...The somatic shepherd... · The PGC flock: cue responses... · When migration fails...
  43. [43]
    Stem cell factor and c-Kit in human primordial germ cells and fetal ...
    c-Kit and SCF play important roles during ascent of primordial germ cells towards the gonadal anlage, and during oogenesis and folliculogenesis in the human ...
  44. [44]
    Identifying the niche controlling melanocyte differentiation
    Melanocytes present in hair follicles are responsible for their pigmentation. Melanocyte differentiation and hair pigmentation depend on the stem cell factor ...Missing: colonization | Show results with:colonization
  45. [45]
    Kit-ligand/stem cell factor induces primordial follicle ... - PubMed
    The hypothesis tested in the current study was that kit-ligand/stem cell factor (KL) promotes the initiation and progression of primordial follicle development ...Missing: formation | Show results with:formation
  46. [46]
    Function of Stem Cell Factor as a Survival Factor of Spermatogonia ...
    Abstract. To address the possibility that stem cell factor (SCF) is a paracrine regulator of germ cell development in the adult rat testis, stage-specific.
  47. [47]
    Spotlight on Spotted Mice: A Review of White Spotting Mouse ...
    May 11, 2004 · White-spotting defects can also be associated with disorders in other cell populations, related either by a common neural crest origin or by ...
  48. [48]
    Interstitial cells of Cajal and human colon motility in health and ...
    Interaction between Kit and its ligand stem cell factor (SCF) from nerves or smooth muscle cells is essential for developing and maintaining several ICC ...
  49. [49]
    Unlocking the regenerative key: Targeting stem cell factors for bone ...
    Oct 27, 2024 · This review elucidates the sources, roles, mechanisms, and molecular strategies of SCF in bone renewal, offering a comprehensive overview of ...
  50. [50]
    Inhibition of Soluble Stem Cell Factor Promotes Intestinal Mucosal ...
    Jul 5, 2023 · Our investigation demonstrates that blocking cleavable SCF248 isoform by administration of specific stem cell factor antibodies enhances healing of the ...
  51. [51]
    The Niche that Fine Tunes Mammalian Intestinal Regeneration - PMC
    Paneth cells respond to inflammation and contribute to tissue regeneration by acquiring stem-like features through SCF/c-Kit signaling. Cell Rep 24: 2312–2328.
  52. [52]
    Two anatomically distinct niches regulate stem cell activity
    Sep 13, 2012 · Stem cell factor (SCF; c-Kit ... BMP signaling inhibits intestinal stem cell self-renewal through suppression of Wnt-beta-catenin signaling.
  53. [53]
  54. [54]
  55. [55]
    Stem cell factor expression, mast cells and inflammation in asthma
    SCF expression increases in the airways of asthmatic patients, and this is reversed after treatment with glucocorticoids.Missing: overexpression urticaria pathophysiological
  56. [56]
  57. [57]
    [PDF] ©Ferrata Storti Foundation - Haematologica
    Our observations, like those of previous studies, sup- port the use of SCF+FLT-3-L+TPO for clinical ex vivo expansion protocols of CB-derived primitive cells,31.<|separator|>
  58. [58]
    The Important Role of FLT3-L in Ex Vivo Expansion of ... - NIH
    FLT3-L could expand HSCs in the co-culture condition at a level of 20-fold equal to the presence of stem cell factor (SCF), thrombopoietin (TPO) and FLT3-L ...
  59. [59]
    Improved hematopoietic stem cell engraftment following ex vivo ...
    The data demonstrate the ability of SCF and Flt3L cytokine-stimulated BM cells to maintain short- and long-term engraftability.Missing: TPO | Show results with:TPO
  60. [60]
    Administration of Hematopoietic Cytokines in the Subacute Phase ...
    Feb 7, 2006 · Compared with the vehicle-treated (no-cytokine) group, administration of G-CSF and SCF significantly reduced the percentage infarct volume.
  61. [61]
    Stem Cell Factor Gene Transfer Promotes Cardiac Repair After ... - NIH
    There is growing evidence that the myocardium responds to injury by recruiting c-kit+ cardiac progenitor cells to the damage tissue.
  62. [62]
    Targeting stem cell niches and trafficking for cardiovascular therapy
    This approach is currently scrutinized in the SITAGRAMI trial on patients suffering from acute MI. A first ad-interim report indicates feasibility and safety of ...
  63. [63]
    In vivo promotion of primordial follicle activation by stem cell factor ...
    Dec 28, 2022 · We provide a series of experimental evidence that SCF is an efficient activator for improving PF activation in mouse ovaries.Missing: 2024 2025
  64. [64]
    Stem cell factor promotes in vitro ovarian follicle development in the ...
    Jun 9, 2016 · In the present study, we hypothesised that SCF promoted primordial follicle activation, specifically by activating the PI3K/AKT and/or MAPK ...Materials And Methods · Follicular Assessment · ResultsMissing: therapy | Show results with:therapy<|separator|>
  65. [65]
    Unlocking the regenerative key: Targeting stem cell factors for bone ...
    This review addressed the mechanisms through which SCF affects myriad biological processes, including cell migration, cell viability, bone repair, and cell ...
  66. [66]
    Stem Cell Factor (SCF) Market: Forecast to Grow at a 11.4% CAGR ...
    Jan 29, 2025 · The Global Stem Cell Factor (SCF) Market was valued at USD 12900 ... 2030 with a CAGR of 12.9% during the forecast period 2024-2030.
  67. [67]
    The Presence of Novel Amino Acids in the Cytoplasmic Domain of ...
    Stem cell factor (SCF) is expressed as an integral membrane growth factor that may be differentially processed to produce predominantly soluble (S) (SCF248) ...<|control11|><|separator|>
  68. [68]
    Mutation of the KIT (mast/stem cell growth factor receptor ... - NIH
    We identified a KIT gene mutation in a proband with classic autosomal dominant piebaldism. This mutation results in a Gly----Arg substitution at codon 664, ...
  69. [69]
    Novel mutations of the KIT (Mast/Stem cell growth factor receptor ...
    Piebaldism results from mutations of the KIT proto-oncogene, which encodes the cellular receptor transmembrane tyrosine kinase for mast/stem cell growth factor.
  70. [70]
    Co expression of SCF and KIT in gastrointestinal stromal tumours ...
    Thus, KIT activation in GISTs may in part result from the presence of SCF within the tumours. Keywords: gastrointestinal tumour, sarcoma, SCF, autocrine loop ...Missing: mastocytosis | Show results with:mastocytosis
  71. [71]
    Neutralization of KIT Oncogenic Signaling in Leukemia with ...
    The SCF/KIT axis works as an autocrine or paracrine loop sustaining proliferation and/or migration. Indeed, KIT is present on 50% of AML (17) and AML blasts ...
  72. [72]
    A Single-Cell Taxonomy Predicts Inflammatory Niche Remodeling to ...
    A cellular taxonomy of the bone marrow stroma in homeostasis and leukemia. ... Stem cell factor/c-kit up-regulates cyclin D3 and promotes cell cycle ...
  73. [73]
    Elevated cutaneous expression of stem cell factor in chronic ...
    Nov 22, 2024 · Elevated cutaneous expression of stem cell factor in chronic spontaneous urticaria: a prospective cohort study. Clin Exp Dermatol. 2024 Nov ...
  74. [74]
    Wnt/β-catenin-C-kit axis may play a role in adenoid cystic carcinoma ...
    Moreover, stem cell factor (SCF; also known as KIT ligand, KITL) induced ERK ... adenoid cystic carcinoma and non adenoid cystic carcinoma malignant tumors of the ...
  75. [75]
  76. [76]
  77. [77]
    Heparan sulfates and heparan sulfate proteoglycans in hematopoiesis
    Jun 20, 2024 · Heparan sulfates (HSs) are linear polysaccharide chains attached to select surface proteins (HS proteoglycans [HSPGs]) that are present in all eukaryotic cells.Review Article · Hematopoietic Stem And... · Heparanase And Hs...<|separator|>
  78. [78]
    Stem cell factor induces mast cell adhesion to fibronectin.
    Mast cell adhesion in the presence of SCF appeared to occur through an integrin receptor as adhesion was calcium dependent and could be blocked by an RGD ...
  79. [79]
    Stem Cell Factor Modulates Avidity of α4β1 and α5β1 Integrins ...
    These studies show that SCF modulates adhesive function in a dose- and time-dependent manner, but does not modulate expression of the integrins α4β1, and α4β1 ...
  80. [80]
    InVivoMAb anti-mouse c-Kit (CD117, Clone: ACK2) | Bio X Cell
    The ACK2 antibody has been reported to deplete c-Kit+ cells, including mast cells, when administered in vivo and neutralize c-Kit+ signaling when used in vitro.
  81. [81]
    Efficacy of SCF drug conjugate targeting c-KIT in gastrointestinal ...
    Aug 24, 2022 · Small molecule kinase inhibitor imatinib is effective in prolonging the survival of GIST patients by targeting KIT. However, drug resistance ...
  82. [82]
    Engineering Stem Cell Factor Ligands with Different c-Kit Agonistic ...
    To directly measure the affinity of each SCF protein for c-Kit, we used surface plasmon resonance (SPR) with the receptor c-Kit protein immobilized on the chip ...
  83. [83]
    Decoupling the Functional Pleiotropy of Stem Cell Factor by Tuning ...
    Mar 9, 2017 · The affinity of the SCF variants for c-Kit was measured by surface plasmon ... Surface Plasmon Resonance. Binding affinity and kinetics were ...
  84. [84]
    Functional interaction of erythropoietin and stem cell factor receptors ...
    Mar 4, 1997 · Functional interaction of erythropoietin and stem cell factor receptors is essential for erythroid colony formation · Abstract · Publication types.
  85. [85]
    Stem cell factor and the regulation of dendritic cell production from ...
    The interactions of SCF with GM-CSF, TNF, interleukin-3 (IL-3) and macrophage colony stimulating factor (M-CSF) in promoting CFU-DL development have now been ...<|control11|><|separator|>