Stem cell factor

Last updated
KITLG
Protein KITLG PDB 1exz.png
Available structures
PDB Ortholog search: PDBe RCSB
Identifiers
Aliases KITLG , FPH2, FPHH, KL-1, Kitl, MGF, SCF, SF, SHEP7, DCUA, KIT ligand, DFNA69, SLF
External IDs OMIM: 184745 MGI: 96974 HomoloGene: 692 GeneCards: KITLG
Orthologs
SpeciesHumanMouse
Entrez
Ensembl
UniProt
RefSeq (mRNA)

NM_003994
NM_000899

NM_013598
NM_001347156

RefSeq (protein)

NP_000890
NP_003985

NP_001334085
NP_038626

Location (UCSC) Chr 12: 88.49 – 88.58 Mb Chr 10: 99.85 – 99.94 Mb
PubMed search [3] [4]
Wikidata
View/Edit Human View/Edit Mouse

Stem cell factor (also known as SCF, KIT-ligand, KL, or steel factor) is a cytokine that binds to the c-KIT receptor (CD117). SCF can exist both as a transmembrane protein and a soluble protein. This cytokine plays an important role in hematopoiesis (formation of blood cells), spermatogenesis, and melanogenesis.

Contents

Production

The gene encoding stem cell factor (SCF) is found on the Sl locus in mice and on chromosome 12q22-12q24 in humans. [5] The soluble and transmembrane forms of the protein are formed by alternative splicing of the same RNA transcript, [6] [7]

Figure 1: Alternative splicing of the same RNA transcript produces soluble and transmembrane forms of stem cell factor (SCF). Scf alternative splicing.JPG
Figure 1: Alternative splicing of the same RNA transcript produces soluble and transmembrane forms of stem cell factor (SCF).

The soluble form of SCF contains a proteolytic cleavage site in exon 6. Cleavage at this site allows the extracellular portion of the protein to be released. The transmembrane form of SCF is formed by alternative splicing that excludes exon 6 (Figure 1). Both forms of SCF bind to c-KIT and are biologically active.

Soluble and transmembrane SCF is produced by fibroblasts and endothelial cells. Soluble SCF has a molecular weight of 18,5 KDa and forms a dimer. It is detected in normal human blood serum at 3.3 ng/mL. [8]

Role in development

SCF plays an important role in the hematopoiesis during embryonic development. Sites where hematopoiesis takes place, such as the fetal liver and bone marrow, all express SCF. Mice that do not express SCF die in utero from severe anemia. Mice that do not express the receptor for SCF (c-KIT) also die from anemia. [9] SCF may serve as guidance cues that direct hematopoietic stem cells (HSCs) to their stem cell niche (the microenvironment in which a stem cell resides), and it plays an important role in HSC maintenance. Non-lethal point mutants on the c-KIT receptor can cause anemia, decreased fertility, and decreased pigmentation. [10]

During development, the presence of the SCF also plays an important role in the localization of melanocytes, cells that produce melanin and control pigmentation. In melanogenesis, melanoblasts migrate from the neural crest to their appropriate locations in the epidermis. Melanoblasts express the KIT receptor, and it is believed that SCF guides these cells to their terminal locations. SCF also regulates survival and proliferation of fully differentiated melanocytes in adults. [11]

In spermatogenesis, c-KIT is expressed in primordial germ cells, spermatogonia, and in primordial oocytes. [12] It is also expressed in the primordial germ cells of females. SCF is expressed along the pathways that the germ cells use to reach their terminal destination in the body. It is also expressed in the final destinations for these cells. Like for melanoblasts, this helps guide the cells to their appropriate locations in the body. [9]

Role in hematopoiesis

SCF plays a role in the regulation of HSCs in the stem cell niche in the bone marrow. SCF has been shown to increase the survival of HSCs in vitro and contributes to the self-renewal and maintenance of HSCs in-vivo. HSCs at all stages of development express the same levels of the receptor for SCF (c-KIT). [13] The stromal cells that surround HSCs are a component of the stem cell niche, and they release a number of ligands, including SCF.

Figure 2: A diagram of a hematopoietic stem cell (HSC) inside its niche. It is adjacent to stromal cells that secrete ligands, such as stem cell factor (SCF). Scf in hsc niche.JPG
Figure 2: A diagram of a hematopoietic stem cell (HSC) inside its niche. It is adjacent to stromal cells that secrete ligands, such as stem cell factor (SCF).

In the bone marrow, HSCs and hematopoietic progenitor cells are adjacent to stromal cells, such as fibroblasts and osteoblasts (Figure 2). These HSCs remain in the niche by adhering to ECM proteins and to the stromal cells themselves. SCF has been shown to increase adhesion and thus may play a large role in ensuring that HSCs remain in the niche. [9]

A small percentage of HSCs regularly leave the bone marrow to enter circulation and then return to their niche in the bone marrow. [14] It is believed that concentration gradients of SCF, along with the chemokine SDF-1, allow HSCs to find their way back to the niche. [15]

In adult mice, the injection of the ACK2 anti-KIT antibody, which binds to the c-Kit receptor and inactivates it, leads to severe problems in hematopoiesis. It causes a significant decrease in the number HSC and other hematopoietic progenitor cells in the bone marrow. [16] This suggests that SCF and c-Kit plays an important role in hematopoietic function in adulthood. SCF also increases the survival of various hematopoietic progenitor cells, such as megakaryocyte progenitors, in vitro. [17] In addition, it works with other cytokines to support the colony growth of BFU-E, CFU-GM, and CFU-GEMM4. Hematopoietic progenitor cells have also been shown to migrate towards a higher concentration gradient of SCF in vitro, which suggests that SCF is involved in chemotaxis for these cells.

Fetal HSCs are more sensitive to SCF than HSCs from adults. In fact, fetal HSCs in cell culture are 6 times more sensitive to SCF than adult HSCs based on the concentration that allows maximum survival. [18]

Expression in mast cells

Mast cells are the only terminally differentiated hematopoietic cells that express the c-Kit receptor. Mice with SCF or c-Kit mutations have severe defects in the production of mast cells, having less than 1% of the normal levels of mast cells. Conversely, the injection of SCF increases mast cell numbers near the site of injection by over 100 times. In addition, SCF promotes mast cell adhesion, migration, proliferation, and survival. [19] It also promotes the release of histamine and tryptase, which are involved in the allergic response.

Soluble and transmembrane forms

The presence of both soluble and transmembrane SCF is required for normal hematopoietic function. [6] [20] Mice that produce the soluble SCF but not transmembrane SCF suffer from anemia, are sterile, and lack pigmentation. This suggests that transmembrane SCF plays a special role in vivo that is separate from that of soluble SCF.

c-KIT receptor

Figure 3: c-Kit expression in hematopoietic cells Kit expression in hematopoietic cells.JPG
Figure 3: c-Kit expression in hematopoietic cells

SCF binds to the c-KIT receptor (CD 117), a receptor tyrosine kinase. [21] c-Kit is expressed in HSCs, mast cells, melanocytes, and germ cells. It is also expressed in hematopoietic progenitor cells including erythroblasts, myeloblasts, and megakaryocytes. However, with the exception of mast cells, expression decreases as these hematopoietic cells mature and c-KIT is not present when these cells are fully differentiated (Figure 3). SCF binding to c-KIT causes the receptor to homodimerize and auto-phosphorylate at tyrosine residues. The activation of c-Kit leads to the activation of multiple signaling cascades, including the RAS/ERK, PI3-Kinase, Src kinase, and JAK/STAT pathways. [21]

Clinical relevance

SCF may be used along with other cytokines to culture HSCs and hematopoietic progenitors. The expansion of these cells ex-vivo (outside the body) would allow advances in bone marrow transplantation, in which HSCs are transferred to a patient to re-establish blood formation. [13] One of the problems of injecting SCF for therapeutic purposes is that SCF activates mast cells. The injection of SCF has been shown to cause allergic-like symptoms and the proliferation of mast cells and melanocytes. [9]

Cardiomyocyte-specific overexpression of transmembrane SCF promotes stem cell migration and improves cardiac function and animal survival after myocardial infarction. [22]

Interactions

Stem cell factor has been shown to interact with CD117. [23] [24]

Related Research Articles

<span class="mw-page-title-main">Haematopoiesis</span> Formation of blood cellular components

Haematopoiesis is the formation of blood cellular components. All cellular blood components are derived from haematopoietic stem cells. In a healthy adult human, roughly ten billion to a hundred billion new blood cells are produced per day, in order to maintain steady state levels in the peripheral circulation.

<span class="mw-page-title-main">Granulocyte colony-stimulating factor</span> Mammalian protein found in Homo sapiens

Granulocyte colony-stimulating factor, also known as colony-stimulating factor 3, is a glycoprotein that stimulates the bone marrow to produce granulocytes and stem cells and release them into the bloodstream.

<span class="mw-page-title-main">Hematopoietic stem cell</span> Stem cells that give rise to other blood cells

Hematopoietic stem cells (HSCs) are the stem cells that give rise to other blood cells. This process is called haematopoiesis. In vertebrates, the very first definitive HSCs arise from the ventral endothelial wall of the embryonic aorta within the (midgestational) aorta-gonad-mesonephros region, through a process known as endothelial-to-hematopoietic transition. In adults, haematopoiesis occurs in the red bone marrow, in the core of most bones. The red bone marrow is derived from the layer of the embryo called the mesoderm.

<span class="mw-page-title-main">Interleukin 3</span> Protein-coding gene in the species Homo sapiens

Interleukin 3 (IL-3) is a protein that in humans is encoded by the IL3 gene localized on chromosome 5q31.1. Sometimes also called colony-stimulating factor, multi-CSF, mast cell growth factor, MULTI-CSF, MCGF; MGC79398, MGC79399: the protein contains 152 amino acids and its molecular weight is 17 kDa. IL-3 is produced as a monomer by activated T cells, monocytes/macrophages and stroma cells. The major function of IL-3 cytokine is to regulate the concentrations of various blood-cell types. It induces proliferation and differentiation in both early pluripotent stem cells and committed progenitors. It also has many more specific effects like the regeneration of platelets and potentially aids in early antibody isotype switching.

<span class="mw-page-title-main">CD34</span> Cluster of differentiation protocol that identifies cell surface antigens.

CD34 is a transmembrane phosphoglycoprotein protein encoded by the CD34 gene in humans, mice, rats and other species.

<span class="mw-page-title-main">Extramedullary hematopoiesis</span> Medical condition

Extramedullary hematopoiesis refers to hematopoiesis occurring outside of the medulla of the bone. It can be physiologic or pathologic.

<span class="mw-page-title-main">CD44</span> Cell-surface glycoprotein

The CD44 antigen is a cell-surface glycoprotein involved in cell–cell interactions, cell adhesion and migration. In humans, the CD44 antigen is encoded by the CD44 gene on chromosome 11. CD44 has been referred to as HCAM, Pgp-1, Hermes antigen, lymphocyte homing receptor, ECM-III, and HUTCH-1.

<span class="mw-page-title-main">Endothelial stem cell</span> Stem cell in bone marrow that gives rise to endothelial cells

Endothelial stem cells (ESCs) are one of three types of stem cells found in bone marrow. They are multipotent, which describes the ability to give rise to many cell types, whereas a pluripotent stem cell can give rise to all types. ESCs have the characteristic properties of a stem cell: self-renewal and differentiation. These parent stem cells, ESCs, give rise to progenitor cells, which are intermediate stem cells that lose potency. Progenitor stem cells are committed to differentiating along a particular cell developmental pathway. ESCs will eventually produce endothelial cells (ECs), which create the thin-walled endothelium that lines the inner surface of blood vessels and lymphatic vessels. The lymphatic vessels include things such as arteries and veins. Endothelial cells can be found throughout the whole vascular system and they also play a vital role in the movement of white blood cells

<span class="mw-page-title-main">CD90</span> Mammalian protein found in Homo sapiens

Thy-1 or CD90 is a 25–37 kDa heavily N-glycosylated, glycophosphatidylinositol (GPI) anchored conserved cell surface protein with a single V-like immunoglobulin domain, originally discovered as a thymocyte antigen. Thy-1 can be used as a marker for a variety of stem cells and for the axonal processes of mature neurons. Structural study of Thy-1 led to the foundation of the Immunoglobulin superfamily, of which it is the smallest member, and led to some of the initial biochemical description and characterization of a vertebrate GPI anchor and also the first demonstration of tissue specific differential glycosylation.

Lymphopoiesis (lĭm'fō-poi-ē'sĭs) is the generation of lymphocytes, one of the five types of white blood cells (WBCs). It is more formally known as lymphoid hematopoiesis.

<span class="mw-page-title-main">KIT (gene)</span> Mammalian protein and protein-coding gene

Proto-oncogene c-KIT is the gene encoding the receptor tyrosine kinase protein known as tyrosine-protein kinase KIT, CD117 or mast/stem cell growth factor receptor (SCFR). Multiple transcript variants encoding different isoforms have been found for this gene. KIT was first described by the German biochemist Axel Ullrich in 1987 as the cellular homolog of the feline sarcoma viral oncogene v-kit.

<span class="mw-page-title-main">CD135</span> Protein-coding gene in the species Homo sapiens

Cluster of differentiation antigen 135 (CD135) also known as fms like tyrosine kinase 3, receptor-type tyrosine-protein kinase FLT3, or fetal liver kinase-2 (Flk2) is a protein that in humans is encoded by the FLT3 gene. FLT3 is a cytokine receptor which belongs to the receptor tyrosine kinase class III. CD135 is the receptor for the cytokine Flt3 ligand (FLT3L).

<span class="mw-page-title-main">Granulocyte-macrophage colony-stimulating factor receptor</span> Protein-coding gene in the species Homo sapiens

The granulocyte-macrophage colony-stimulating factor receptor also known as CD116, is a receptor for granulocyte-macrophage colony-stimulating factor, which stimulates the production of white blood cells. In contrast to M-CSF and G-CSF which are lineage specific, GM-CSF and its receptor play a role in earlier stages of development. The receptor is primarily located on neutrophils, eosinophils and monocytes/macrophages, it is also on CD34+ progenitor cells (myeloblasts) and precursors for erythroid and megakaryocytic lineages, but only in the beginning of their development.

<span class="mw-page-title-main">FMS-like tyrosine kinase 3 ligand</span> Protein-coding gene in the species Homo sapiens

Fms-related tyrosine kinase 3 ligand (FLT3LG) is a protein which in humans is encoded by the FLT3LG gene.

<span class="mw-page-title-main">HOXA9</span> Protein-coding gene in humans

Homeobox protein Hox-A9 is a protein that in humans is encoded by the HOXA9 gene.

<span class="mw-page-title-main">Sean J. Morrison</span>

Sean J. Morrison is a Canadian-American stem cell biologist and cancer researcher. Morrison is the director of Children's Medical Center Research Institute at UT Southwestern, a nonprofit research institute established in 2011 as a joint venture between Children’s Health System of Texas and UT Southwestern Medical Center. The CRI was established in 2011 by Morrison with the mission to perform transformative biomedical research at the interface of stem cell biology, cancer, and metabolism to better understand the biological basis of disease. He is a Howard Hughes Medical Institute Investigator and member of the National Academy of Medicine. From 2015 to 2016 Morrison served as the president of the International Society for Stem Cell Research.

<span class="mw-page-title-main">CFU-GEMM</span>

CFU-GEMM is a colony forming unit that generates myeloid cells. CFU-GEMM cells are the oligopotential progenitor cells for myeloid cells; they are thus also called common myeloid progenitor cells or myeloid stem cells. "GEMM" stands for granulocyte, erythrocyte, monocyte, megakaryocyte.

<span class="mw-page-title-main">Haematopoietic system</span>

The haematopoietic system is the system in the body involved in the creation of the cells of blood.

Many human blood cells, such as red blood cells (RBCs), immune cells, and even platelets all originate from the same progenitor cell, the hematopoietic stem cell (HSC). As these cells are short-lived, there needs to be a steady turnover of new blood cells and the maintenance of an HSC pool. This is broadly termed hematopoiesis. This event requires a special environment, termed the hematopoietic stem cell niche, which provides the protection and signals necessary to carry out the differentiation of cells from HSC progenitors. This niche relocates from the yolk sac to eventually rest in the bone marrow of mammals. Many pathological states can arise from disturbances in this niche environment, highlighting its importance in maintaining hematopoiesis.

Since haematopoietic stem cells cannot be isolated as a pure population, it is not possible to identify them in a microscope. Therefore, there are many techniques to isolate haematopoietic stem cells (HSCs). HSCs can be identified or isolated by the use of flow cytometry where the combination of several different cell surface markers are used to separate the rare HSCs from the surrounding blood cells. HSCs lack expression of mature blood cell markers and are thus, called Lin-. Lack of expression of lineage markers is used in combination with detection of several positive cell-surface markers to isolate HSCs. In addition, HSCs are characterised by their small size and low staining with vital dyes such as rhodamine 123 or Hoechst 33342.

References

  1. 1 2 3 GRCh38: Ensembl release 89: ENSG00000049130 - Ensembl, May 2017
  2. 1 2 3 GRCm38: Ensembl release 89: ENSMUSG00000019966 - Ensembl, May 2017
  3. "Human PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  4. "Mouse PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  5. Geissler EN, Liao M, Brook JD, Martin FH, Zsebo KM, Housman DE, Galli SJ (March 1991). "Stem cell factor (SCF), a novel hematopoietic growth factor and ligand for c-kit tyrosine kinase receptor, maps on human chromosome 12 between 12q14.3 and 12qter". Somat. Cell Mol. Genet. 17 (2): 207–14. doi:10.1007/BF01232978. PMID   1707188. S2CID   37793786.
  6. 1 2 Flanagan JG, Chan DC, Leder P (March 1991). "Transmembrane form of the kit ligand growth factor is determined by alternative splicing and is missing in the Sld mutant". Cell. 64 (5): 1025–35. doi:10.1016/0092-8674(91)90326-t. PMID   1705866. S2CID   11266238.
  7. Anderson DM, Williams DE, Tushinski R, Gimpel S, Eisenman J, Cannizzaro LA, Aronson M, Croce CM, Huebner K, Cosman D (August 1991). "Alternate splicing of mRNAs encoding human mast cell growth factor and localization of the gene to chromosome 12q22-q24". Cell Growth Differ. 2 (8): 373–8. PMID   1724381.
  8. Langley KE, Bennett LG, Wypych J, Yancik SA, Liu XD, Westcott KR, Chang DG, Smith KA, Zsebo KM (February 1993). "Soluble stem cell factor in human serum". Blood. 81 (3): 656–60. doi: 10.1182/blood.V81.3.656.656 . PMID   7678995.
  9. 1 2 3 4 Broudy VC (August 1997). "Stem cell factor and hematopoiesis". Blood. 90 (4): 1345–64. doi: 10.1182/blood.V90.4.1345 . PMID   9269751.
  10. Blouin R, Bernstein A (1993). "The White spotting and Steel hereditary anaemias of the mouse". In Freedman MH, Feig SA (eds.). Clinical disorders and experimental models of erythropoietic failure. Boca Raton: CRC Press. ISBN   0-8493-6678-X.
  11. Wehrle-Haller B (June 2003). "The role of Kit-ligand in melanocyte development and epidermal homeostasis". Pigment Cell Res. 16 (3): 287–96. doi:10.1034/j.1600-0749.2003.00055.x. PMID   12753403.
  12. Rossi P, Sette C, Dolci S, Geremia R (October 2000). "Role of c-kit in mammalian spermatogenesis" (PDF). J. Endocrinol. Invest. 23 (9): 609–15. doi:10.1007/bf03343784. hdl: 2108/65858 . PMID   11079457. S2CID   43786244.
  13. 1 2 Kent D, Copley M, Benz C, Dykstra B, Bowie M, Eaves C (April 2008). "Regulation of hematopoietic stem cells by the steel factor/KIT signaling pathway". Clin. Cancer Res. 14 (7): 1926–30. doi: 10.1158/1078-0432.CCR-07-5134 . PMID   18381929.
  14. Méndez-Ferrer S, Lucas D, Battista M, Frenette PS (March 2008). "Haematopoietic stem cell release is regulated by circadian oscillations". Nature. 452 (7186): 442–7. Bibcode:2008Natur.452..442M. doi:10.1038/nature06685. PMID   18256599. S2CID   4403554.
  15. Nervi B, Link DC, DiPersio JF (October 2006). "Cytokines and hematopoietic stem cell mobilization". J. Cell. Biochem. 99 (3): 690–705. doi: 10.1002/jcb.21043 . PMID   16888804. S2CID   40354996.
  16. Ogawa M, Matsuzaki Y, Nishikawa S, Hayashi S, Kunisada T, Sudo T, Kina T, Nakauchi H, Nishikawa S (July 1991). "Expression and function of c-kit in hemopoietic progenitor cells". J. Exp. Med. 174 (1): 63–71. doi:10.1084/jem.174.1.63. PMC   2118893 . PMID   1711568.
  17. Keller JR, Ortiz M, Ruscetti FW (September 1995). "Steel factor (c-kit ligand) promotes the survival of hematopoietic stem/progenitor cells in the absence of cell division". Blood. 86 (5): 1757–64. doi: 10.1182/blood.V86.5.1757.bloodjournal8651757 . PMID   7544641.
  18. Bowie MB, Kent DG, Copley MR, Eaves CJ (June 2007). "Steel factor responsiveness regulates the high self-renewal phenotype of fetal hematopoietic stem cells". Blood. 109 (11): 5043–8. doi: 10.1182/blood-2006-08-037770 . PMID   17327414.
  19. Okayama Y, Kawakami T (2006). "Development, migration, and survival of mast cells". Immunol. Res. 34 (2): 97–115. doi:10.1385/IR:34:2:97. PMC   1490026 . PMID   16760571.
  20. Brannan CI, Lyman SD, Williams DE, Eisenman J, Anderson DM, Cosman D, Bedell MA, Jenkins NA, Copeland NG (June 1991). "Steel-Dickie mutation encodes a c-kit ligand lacking transmembrane and cytoplasmic domains". Proc. Natl. Acad. Sci. U.S.A. 88 (11): 4671–4. Bibcode:1991PNAS...88.4671B. doi: 10.1073/pnas.88.11.4671 . PMC   51727 . PMID   1711207.
  21. 1 2 Rönnstrand L (October 2004). "Signal transduction via the stem cell factor receptor/c-Kit". Cell. Mol. Life Sci. 61 (19–20): 2535–48. doi:10.1007/s00018-004-4189-6. PMID   15526160. S2CID   2602233.
  22. Xiang FL, Lu X, Hammoud L, Zhu P, Chidiac P, Robbins J, Feng Q (September 2009). "Cardiomyocyte-specific overexpression of human stem cell factor improves cardiac function and survival after myocardial infarction in mice". Circulation. 120 (12): 1065–74, 9 p following 1074. doi: 10.1161/CIRCULATIONAHA.108.839068 . PMID   19738140.
  23. Lev S, Yarden Y, Givol D (May 1992). "A recombinant ectodomain of the receptor for the stem cell factor (SCF) retains ligand-induced receptor dimerization and antagonizes SCF-stimulated cellular responses". J. Biol. Chem. 267 (15): 10866–73. doi: 10.1016/S0021-9258(19)50098-9 . PMID   1375232.
  24. Blechman JM, Lev S, Brizzi MF, Leitner O, Pegoraro L, Givol D, Yarden Y (Feb 1993). "Soluble c-kit proteins and antireceptor monoclonal antibodies confine the binding site of the stem cell factor". J. Biol. Chem. 268 (6): 4399–406. doi: 10.1016/S0021-9258(18)53623-1 . PMID   7680037.

Further reading