You are on page 1of 22

Stem Cell Factor

Jonathan Roy Keller1,* and Diana Marie Linnekin2


1
Intramural Research and Support Program-SAIC, Frederick Cancer Research and
Development Center-NCI, Building 560, Room 12-03, PO Box B, Frederick, MD 21702-1201, USA
2
Basic Research Laboratory-DBS, Frederick Cancer Research and Development Center-NCI,
Building 567, PO Box B, Frederick, MD 21702-1201, USA
* corresponding author tel: 301-846-1461, fax: 301-846-6646, e-mail: kellerj@mail.ncifcrf.gov
DOI: 10.1006/rwcy.2000.09003.

SUMMARY stromal cells (macrophages, fibroblasts, endothelial


cells, and adipocytes) in the bone marrow and else-
Stem cell factor (SCF) and its receptor, c-Kit, are where, which interact with specific receptors
essential for hematopoietic, melanocyte, and germ cell expressed on hematopoietic cells. The story of the dis-
development. In the hematopoietic compartment, SCF covery of one HGF/receptor pair, stem cell factor
has been shown to act on cells at multiple stages of (SCF) and SCF receptor (SCFR), illustrates the
development, including effects on primitive stem cells importance of HGFs in hematopoietic development.
and their more differentiated progeny. In particular, This story began with studies of inbred mouse
SCF can function to promote stem/progenitor cell strains that have naturally occurring mutations affect-
survival, proliferation and differentiation (especially ing SCF or its receptor that were initiated 20 years
in combination with other growth factors), adhesion, ago. Specifically, homozygote offspring from W mouse
activation, and migration/chemoattraction. While c- strains (white spotting locus on chromosome 5), or Sl
Kit is highly expressed on differentiated mast cells mouse strains (steel locus on chromosome 10) showed
and SCF is a potent mast cell growth factor, c-Kit varying degrees of impaired hematopoietic develop-
expression is downregulated during the maturation of ment (hypoplastic with macrocytic anemia), altera-
all other hematopoietic lineages. SCF can exist in tions in coat color or pigmentation (lack of cutaneous
both soluble and membrane forms which may have melanocytes), and defects in gametogenesis (sterility)
distinct biological functions in vivo. Further under- (Russell, 1979; Silvers, 1979). Bone marrow trans-
standing of this important hematopoietic regulator plantation and embryo fusion studies using W or Sl
will provide important insights into the biology, mutant mouse strains led to the seminal observation
biochemistry, and molecular biology of hematopoetic that the W mutant defects were intrinsic to hemato-
stem cells. poietic stem and mast cells, while the Sl mutant
defects were extrinsic to hematopoietic cells in that
bone marrow stromal cells were required to support
hematopoietic development (Russell et al., 1959;
BACKGROUND Altus et al., 1971; McCulloch et al., 1964, 1965).
Because both W and Sl mice showed defects in the
Discovery same three cell lineages (hematopoietic, melanocyte,
and germ cell) Elisabeth Russell speculated that the
Definitive hematopoiesis is a complex cellular process W locus might encode a growth factor receptor and
that is regulated, in part, by hematopoietic growth the Sl locus its complementary ligand (Russell, 1979).
factors (HGFs) and the bone marrow microenviron- This hypothesis was confirmed, in part, by the
ment, which act to promote the survival, prolifera- observation that the W locus encoded the proto-
tion, and differentiation of hematopoietic stem cells oncogene c-kit (SCFR), a transmembrane tyrosine
and their progeny (Spangrude, 1991; Ogawa, 1993; kinase, which was largely deleted in one strain of W
Metcalf, 1993). HGFs are produced by accessory or mice (W/W) mice and results in the most severe
878 Jonathan Roy Keller and Diana Marie Linnekin

phenotype in homozygous offspring (prenatal letha- Figure 1 Soluble and transmembrane forms of SCF.
lity). W mouse strains that produce viable homo- The amino acid sequences encoded by exon 6 are
zygous offspring do not show deleted SCFR, but shown in light blue, which contains the major
rather have mutations in the kinase domain that proteolytic site (purple arrow) that results in the
affect the kinase enzymatic activity (Charbot et al., production of soluble SCF164. Soluble SCF164 forms
1988; Geissler et al., 1988). SCFR is structurally rela- nonconvalently linked dimers using the cysteine
residues indicated. SCF220 lacks exon 6 and the pri-
ted to other transmembrane tyrosine kinases, including mary proteolytic site and remains membrane bound.
c-fms, the receptor for macrophage colony-stimulat- The signal peptide sequence is shown in purple and the
ing factor (M-CSFR), and platelet-derived growth transmembrane domain is shown as a green box. (Full
factor receptor (PDGFR) (Qui et al., 1988). colour figure can be viewed online.)
The search for the SCFR ligand ended several years
later when three groups simultaneously purified it
from medium conditioned by fibroblasts (stromal
cells) or a rat liver-derived cell by assaying the pro-
liferative response of cell lines or normal bone
marrow cells to SCFR ligand (Huang et al., 1990;
Zsebo et al., 1990a; Williams et al., 1990). The proteins
were purified to homogeneity, sequenced and used
to clone the corresponding cDNAs (Anderson et al.,
1990; Copeland et al., 1990; Martin et al., 1990).

Alternative names
SCF is also known as steel factor (SF) because it was
mapped to the Sl locus, Kit ligand (KL), and mast
cell growth factor (MCGF) (Anderson et al., 1990; germ, and melanocyte cell development during both
Copeland et al., 1990; Huang et al., 1990; Martin et al., embryonic development and adult life (Bernstein
1990; Williams et al., 1990; Zsebo et al., 1990b). et al., 1991b; Besmer et al., 1993). In particular, SCF
is expressed along the migratory pathways for pri-
mordial germ cells and melanocytes as well as sites of
Structure active hematopoiesis (fetal liver). SCF is essential for
the migration and homing of stem cells to their appro-
SCF is produced in both soluble and transmembrane priate developmental sites; moreover, SCF directly
forms that are encoded by two distinct mRNAs promotes the survival of these stem cell populations
generated from the same gene by alternate splicing of and greatly enhances their proliferation in response to
the primary RNA transcript (Flannagan et al., 1991; other growth factors. While SCF is critical for the
Anderson et al., 1990, 1991). Both isoforms of SCF development of a number of tissues, this review will
are initially produced as transmembrane proteins that only focus on hematopoietic cells and will refer to
contain a 25 amino acid signal peptide sequence cells from other tissue where appropriate.
(SCF248 or SCF220). Transmembrane SCF248 can be
proteolytically cleaved to give rise to soluble SCF,
which contains the first 164 amino acids of SCF248 GENE AND GENE REGULATION
(Figure 1). The alternatively spliced isoform, SCF220,
encodes a shorter 220 amino acid transmembrane Accession numbers
protein identical to SCF248 except that it lacks amino
acids 147±177, the region containing the proteolytic SCF cDNAs have been cloned and sequenced from
cleavage site. Little or no soluble SCF is produced a variety of species including mouse (GenBank:
from SCF220. U44725) (Anderson et al., 1990; Huang et al., 1990;
Zsebo et al., 1990b), human (GenBank: M59964), rat
(GenBank: M59964) (Martin et al., 1990), pig (L07786)
Main activities and (Zhang and Anthony, 1994), chicken (D13516) (Zhou
pathophysiological roles et al., 1993), quail (GenBank: U43079) (Pettite and
Kulik, 1996), dog (S53329) (Shull et al., 1992), cat
SCF plays essential roles in the survival, growth, and (Dunham and Onions, 1995) (DDBJ: D50833), and
differentiation of cells responsible for hematopoietic, horse (GenBank: AF053498).
Stem Cell Factor 879

binding motif for the transcription factors TFIID and


Chromosome location SP1 (Bedell et al., 1996; Taylor et al., 1996; Jiang et al.,
1997). The human SCF promoter contains a TATA
Mouse SCF has been mapped to chromosome 10 in a
box beginning at ÿ38 bp and a CCAAT box at
region between the peptidase 2 (Pep-2) and phenylhy-
ÿ71 bp (Figure 2). The major transcription initiation
droxylase (Pah) genes. Comparing the maps of mouse
start site for SCF in mouse brain, lung, kidney, heart,
and human chromosomes indicated that the human
ovary, and testes is located 28 nucleotides down-
SCF gene might be located on the distal end of the
stream from the consensus TATA motif in the murine
long arm of human chromosome 12 (Copeland et al.,
promoter (Bedell et al., 1996). There are also three
1990; Huang et al., 1990; Zsebo et al., 1990b). This was
ATGs located at positions ‡88, ‡123, and ‡193,
later confirmed by in situ hybridization where it was
with the latter encoding the initiator methionine for
shown that human SCF mapped to chromosome
SCF, however; initiation of translation from ‡88 or
12q22 (Anderson et al., 1991) and somatic cell hybrids
‡123 would produce peptides of 37 and 7 amino acids
which mapped SCF to 12q14.3-qter (Geissler et al.,
in length respectively. Other important sites in the
1991).
human promoter include SP1 sites at ÿ224, ÿ139,
The mouse SCF gene spans roughly 50 kb and is
and ÿ50, AP-2 sites at ÿ202 and ÿ91, and AP-1 and
composed of nine exons (Martin et al., 1990; Brannan
cAMP-response elements further upstream. In com-
et al., 1992). The locations of the intron and exon
parison, the mouse promoter contains SP1 sites, a
boundaries are highly conserved between species
hepatocyte-acute phase factor 1 motif (H-APF-1), a
including human, rat, and mouse. The first exon of
negative regulatory element-box 1 (NRE-1), a reverse
the mouse encodes 198 nucleotides of the 50 UTR and
TPA responsive element (TGAGTA), TRE/Rev sites,
the first five amino acids of the signal peptide. Exons
NFB elements, as well as AP-2 and AP-1 sites
2±6 encode the extracellular domain of the transmem-
(Taylor et al., 1996; Jiang et al., 1997). A functional
brane-bound ligand, while exon 7 encodes a portion
analysis of the rat SCF promoter sequences per-
of the extracellular domain and 23 amino acids of the
formed by transfecting a reporter plasmid-containing
membrane-spanning region. Exons 8 and 9 encode 23
promoter sequences into SCF-expressing and non-
amino acids and 22 amino acids of the cytoplasmic
SCF-expressing cell lines showed that sequences from
domain, respectively.
ÿ119 to ‡43 nucleotides contained the SCF core
There are two major RNA transcripts for SCF
promoter activity (Jiang et al., 1997). Transfection of
detected by northern blot analysis that range in size
plasmids containing additional 50 sequence (up to
from 5.5 kb to 6.5 kb for the larger message and from
ÿ1461 nucleotides) had no further effect on reporter
3 kb to 4.6 kb for the smaller mRNA. The difference
gene expression. However, SCF core promoter acti-
in the size of the mRNA transcripts is due to the
vity could be greatly enhanced by treating cells with
length of the 30 UTR which is determined by the use
cAMP or forskolin (previously shown to increase
of one of two polyadenylation sites (Anderson et al.,
SCF expression in Sertoli cells), suggesting that cAMP-
1990; Bedell et al., 1996). The largest SCF mRNA
dependent regulatory elements might play a role in
transcript predicted by sequence analysis is roughly
SCF gene activation. Human SCF promoter sequen-
5.4 kb and contains 197 nucleotides of 50 UTR, an
ces were also cloned into luciferase reporter constructs
ORF of 818 nucleotides and a 30 UTR of 4432
and analyzed for function by deletion analysis. The
nucleotides. The smaller mRNA differs in the length
shortest promoter construct that encompassed the
of the 30 UTR region which is 2732 nucleotides. There
core promoter activity activated by cAMP or forskolin
are 14 ATTA motifs in the 30 UTR of the SCF
contained sequences from ÿ167 to ‡109 nucleotides.
mRNA which have been shown to affect HGF mRNA
In comparison, promoter constructs that contained
stability and thereby could greatly affect protein levels
additional 50 sequences (from ÿ853 or ÿ2185 nucleo-
(Shaw and Kamen, 1986; Bedell et al., 1996).
tides) showed the same increase in reporter activity in
cells treated with cAMP; however, basal levels of
reporter activity were significantly reduced, suggest-
Regulatory sites and corresponding ing the potential for negative regulatory elements.
transcription factors
A comparison of the 50 flanking sequences of mouse,
rat, chicken, pig, and human SCF show a high degree Cells and tissues that express
of homology (82±94% identity) within a 110 nucleo- the gene
tide interval upstream of the initiator methionine that
includes a GGCGGG motif representing the core See section on Cellular sources that produce SCF.
880 Jonathan Roy Keller and Diana Marie Linnekin

Figure 2 Human SCF promoter DNA sequence. The DNA sequence of the upstream region of the SCF gene is shown,
including the first five codons and corresponding amino acids of Exon 1 is underlined. DNA sequence elements that are
homologous to transcription factor-binding elements are also underlined and indicated above the sequence. TATA and
CAT box sequences are underlined and indicated above the sequence. The transcription start site is shown as a red arrow.

PROTEIN and the transmembrane region are shown in the


boxed areas. SCF protein sequences that are con-
served between species are shown in red shaded
Accession numbers letters. Black shaded letters indicate conservative
amino acid differences between species while all other
SCF cDNAs isolated from a variety of species encode amino acid differences are shown as nonshaded black
proteins of roughly 273 amino acids including mouse letters. Mouse and human proteins show an overall
(SwissProt: P20826) (Anderson et al., 1990; Huang amino acid identity of 82% with 92% identity in the
et al., 1990; Zsebo et al., 1990a), human (SwissProt: cytoplasmic domain.
P21583) (Martin et al., 1990), and rat (SwissProt:
P21581) (Martin et al., 1990), pig (SwissProt: Q29030)
(Zhang and Anthony, 1994), chicken (SwissProt:
Q09108) (Zhou et al., 1993), dog (SwissProt: Q06220)
Description of protein
(Shull et al., 1992).
SCF protein is produced in both soluble and mem-
brane-bound forms (Anderson et al., 1990, 1991).
Sequence These isoforms arise from two differentially spliced
cDNAs. One cDNA encodes a 273 amino acid pre-
The amino acid sequences for SCF from human, cursor that contains a cleavable 25 amino acid N-
mouse, pig, cat, and dog have been aligned and show terminal signal peptide and gives rise to the 248 amino
a strong degree of conservation across species acid transmembrane protein SCF248 also known as
(Figure 3). The 25 amino acid signal peptide sequence KL-1 or SCF-1. SCF248 is comprised of a 189 amino
Stem Cell Factor 881

Figure 3 Alignment of SCF amino acid sequences. Human, murine, porcine, feline and
canine SCF amino acid sequences were aligned. The signal peptide and transmembrane
regions of SCF are shown in the boxed areas. Identical SCF amino acids between species are
shown in red shaded letters. Black shaded letters indicate conservative amino acid changes
between species, while black nonshaded letters indicate amino acid divergence between
species. The four cysteines involved in the intrachain disulfide bonds are shown in shaded
yellow letters. (Full colour figure can be viewed online.)
882 Jonathan Roy Keller and Diana Marie Linnekin

acid extracellular peptide domain, a 22 amino acid membrane-bound ligands). While it is not known
membrane-spanning region (from amino acids 190 to whether the levels of soluble SCF are significantly
212 of SCF248) and a short 36 amino acid intracellular reduced in Sl d homozygous mice compared with wild-
cytoplasmic tail (amino acids 212±248) (see Figure 1). type mice and might also contribute to the phenotype,
SCF248 can be proteolytically cleaved to give rise to administration of purified SCF to Sl d homozygous
soluble SCF, which contains the first 164 amino acids mice increases the hematocrit, and mast cell numbers
of SCF248. The alternatively spliced form of SCF at the site of injection, suggesting that a portion of the
cDNA encodes a shorter 220 amino acid transmem- hematological effects in Sl d homozygous mice can
brane protein SCF220, also known as KL-2 or SCF-2, be reversed by administration of soluble SCF (Zsebo
that is identical to SCF248 except that it lacks amino et al., 1990b). Furthermore, studies comparing the
acids 147±177 (encoded by exon 6), which contains a expression of soluble and membrane-bound isoforms
proteolytic cleavage site important for the production of SCF in vitro have shown that human hematopoie-
of soluble SCF. This transcript arises from the use tic cell growth can be maintained for longer periods
of an alternative 30 splice acceptor site in the RNA, on stromal cells that express transmembrane SCF
which skips the 84 nucleotides contained within (SCF220) in comparison with stromal cells that
exon 6. A second proteolytic cleavage site in mouse express SCF248 which is cleaved to produce soluble
SCF has been identified between amino acids 178±181 SCF (Toksoz et al., 1992). In addition, the survival
(12 amino acids in exon 7) that can be used in vitro to and proliferation of factor-dependent hematopoietic
generate soluble SCF; however, whether or not this cell lines are enhanced when they are co-cultured on
site is used in vivo is unknown (Huang et al., 1992; stromal cells that express a noncleavable variant of
Toksoz et al., 1992; Majumdar et al., 1994). transmembrane SCF (SCFx9/D3) versus SCF248 (Kapur
The soluble form of the SCF protein (164 amino et al., 1998). Thus, transmembrane SCF may be
acids) forms noncovalently linked homodimers that critical for the maintenance and survival of primitive
have extensive N- and O-linked glycosylation with an hematopoietic progenitor cells and for mast cell
apparent molecular mass of 50±60 kDa. SCF dimers accumulation and proliferation. Finally, the mem-
are required for SCF receptor dimerization, receptor brane-bound isoform of SCF can also function as an
transphosphorylation, activation, and signal trans- adhesion molecule for mast cells and hematopoietic
duction (Lev et al., 1992; Heldin, 1995; Hsu et al., 1997). progenitor cells (Kaneko et al., 1991; Adachi et al.,
In this regard, dimerization defective isoforms of SCF 1992; Avraham et al., 1992; Long et al., 1992; Kodama
show greatly reduced biological activity including the et al., 1994).
ability to bind SCFR. The majority of soluble SCF
exists as a monomer in serum because it is in low
concentration; however, it is predicted that trans- Discussion of crystal structure
membrane SCF exists predominantly as dimers
because of the enhanced probability for SCF mono- The structure of SCF has not been directly deter-
mers to interact with each other and form dimers in mined; however, a model has been predicted based on
the membrane. the published structural model for M-CSF that was
The exact physiological role(s) of soluble and determined at 2.5 AÊ by X-ray crystallography (Bazan,
membrane-bound SCF are not fully understood. 1991; Pandit et al., 1992) (Figure 4). There are four
However, analysis of mouse strains with naturally antiparallel (helical bundles (I, II, III, and IV) which
occurring mutations in the SCF gene have provided are connected by two overhand loops. Thus far, all
some insights into the importance of these two other examples of four helix proteins have been
isoforms (also described in the knockout and trans- identified as cytokines including GM-CSF, growth
gene sections below). In particular, a mutation of hormone, and IL-4 (Bazan, 1991). The four cysteine
SCF in one mouse strain, steel-dickie (Sl d ), results in residues in SCF are proposed to be involved in intra-
the production of soluble but not transmembrane molecular disulfide bonds, which contribute to the
SCF (Flannagan and Leder, 1990; Brannan et al., three-dimensional structure of the protein (Figure 4).
1991). Homozygous Sl d mice are viable but they have The intramolecular disulfide pairs are Cys4±Cys89
complete lack of skin pigmentation, are sterile, show and Cys43±Cys138, both of which are critical for full
severe anemia, and are mast cell deficient. Thus, it is biological activity (Nishikawa et al., 1992; Langley
speculated that the membrane-associated form of et al., 1994; Jones et al., 1996). The same four Cys
SCF is critical for normal mouse development, sug- residues are conserved between other family mem-
gesting that some of the important biological effects bers, including M-CSF and Flt-3 ligand (Flt-3L).
of SCF are mediated through juxtacrine stimulation However, M-CSF contains three additional Cys resi-
of target cell populations (stimulation through dues, two that are involved in another intramolecular
Stem Cell Factor 883

Figure 4 A molecular model of human SCF. This that share significant structural homologies as
model is based on the published structure of human discussed in the crystal structure section above. These
M-CSF (reprinted with permission from Broudy, 1997, include conserved cysteine residues, the four helix
copyright 1997, Blood). The SCF four helical bundles bundle structure and the transmembrane domain.
are shown (I, II, III, IV) and the location of the
intramolecular bonds is indicated with yellow balls.
(Full colour figure can be viewed online.)
Posttranslational modifications
Mouse and human SCF are extensively glycosylated
and contain both N- and O-linked sugars (Anderson
et al., 1990; Huang et al., 1990; Arakawa et al., 1991).
In particular, the predicted molecular mass of
monomeric SCF is 18,589 kDa, which is close to the
molecular weight of E. coli-derived SCF which
migrates at an apparent molecular weight of 18.5 kDa
on SDS-PAGE (Arakawa et al., 1991). In compari-
son, natural and CHO-derived monomeric forms of
SCF migrate on SDS-PAGE with an apparent
molecular mass of 28±35 kDa, which can be reduced
to 18.5 kDa by removal of both O- and N-linked
sugars, indicating extensive glycosylation (30% by
weight) (Zsebo et al., 1990a; Arakawa et al., 1991; Lu
et al., 1992; Langley et al., 1992).

CELLULAR SOURCES AND


disulfide bond and one that is involved in an
intermolecular disulfide bond (between M-CSF TISSUE EXPRESSION
monomers) (Bazan, 1991; Pandit et al., 1992). Thus far,
no intermolecular bonds between SCF monomers Cellular sources that produce
have been identified, however, it is speculated that
there is a large area of contact between SCF mono- Analysis of SCF gene expression in the developing
mers which is sufficient for stable dimer formation. embryo suggests that this gene has multiple functional
Truncation mutational analysis of SCF protein roles in development. For example, the SCF gene is
indicates that the majority of the extracellular se- expressed along the migratory pathways for stem cells
quences (those present in the soluble form of SCF) are that give rise to hematopoietic, melanocyte, and germ
required for biological activity (first 141 of 164 cell lineages as indicated by in situ hybridization
residues) (Nishikawa et al., 1992; Langley et al., 1994). and northern blot analysis (Matsui et al., 1990; Motro
Studies of mouse and human SCF chimeras et al., 1991; Keshet et al., 1991). Specifically, SCF is
demonstrate that SCF residues 1±35 and 79±97 expressed in the genital ridge of day 10 embryos,
located in the first and third helix are required for while the receptor (SCFR) is expressed in the migrat-
SCF's ability to synergize with GM-CSF (Matous ing germ cells. SCF expression in the genital ridge
et al., 1996). In agreement with this study, the se- decreases after day 11.5 of gestation but remains high
quences recognized by one neutralizing SCF mono- in the developing gonads during sexual differential
clonal mapped to residues Leu79 and Asn97. Another (both in the testes and ovary). This pattern of expres-
region between 121 and 128 in the fourth helix is also sion suggests that SCF is involved in regulating the
required for synergistic responses with GM-CSF. migration, proliferation, and differentiation of germ
cells, which is in agreement with the sterile phenotype
of Sl mice. Similarly, SCF is expressed in mesenchy-
Important homologies mal cells located in the limb bud where melanocyte
precursors colonize while SCFR is expressed on the
SCF, Flt-3 ligand, and M-CSF are all type I trans- migrating cells. The expression of SCF persists during
membrane proteins with short cytoplasmic domains and after stem cell colonization of the limb buds. SCF
884 Jonathan Roy Keller and Diana Marie Linnekin

is also expressed in fetal liver, which is the migratory follicle-stimulating hormone (FSH), growth hor-
site for hematopoietic stem cells in the developing mone-releasing hormone (GHRH), forskolin, and
embryo between embryonic days 12 and 17. As would cholera toxin.
be predicted, there is a marked reduction in the Both TGF and TNF have been shown to
number of SCFR-positive stem cells that migrate to directly inhibit SCF-mediated growth and survival
the fetal livers of Sl/Sl embryos. SCF mRNA trans- effects on hematopoietic cell progenitors in vitro and
cripts were also detected in other tissues in the in vivo (Keller et al., 1992; McNiece et al., 1992;
developing embryo, including the spinal cord, fore- Jacobsen et al., 1994, 1995a, 1995b, 1996). Local
brain, cerebellum, and olfactory bulbs, suggesting administration of glucocorticoids at sites of allergic
that SCF might play a role in the developing CNS. inflammation in vivo significantly decreases mast cell
While there are no gross neurological defects in Sl numbers and inhibits SCF production (Finotto et al.,
mutant mice, Sl/Sl d mutant mice show a defect in 1997). The effect of glucocorticoids on mast cell
hippocampal-dependent learning (spatial learning) numbers was reversed by exogenous SCF, suggesting
(Motro et al., 1996). that SCF is required for the growth and survival of
SCF is expressed by stromal or accessory cells in mast cells at local sites of allergy in vivo.
the adult where these cells constitute the hematopoie- SCF mRNA levels are increased in bone marrow
tic microenvironment and include endothelial cells, and spleen cells from mice that were previously
monocytes, and fibroblasts (Flannagan and Leder, treated with 5-fluorouracil (5-FU), a myeloablative
1990; McNiece et al., 1991a; Aye et al., 1992; Heinrich chemotherapeutic agent (Hunt et al., 1992; van Os
et al., 1993; Linenberger et al., 1995; Broudy et al., et al., 1997). SCF expression was also increased in
1996). SCF is also produced by intestinal epithelial bone marrow cells from mice that had received a
cells, Sertoli cells, and follicular cells that surround sublethal dose of gamma-irradiation, suggesting that
oocytes in the gonads, in thymic stroma and brain increased SCF mRNA and protein levels may be a
cells, including those that constitute the olfactory crucial part of the host response to hematopoietic
bulb, thalamus, cerebellum, and brainstem (Tajima injury (Limmani et al., 1995). In this regard, admini-
et al., 1991; deCastro et al., 1994; Klimpel et al., 1995). stration of antibodies that inhibit SCF's ability to
SCF expression has also been detected in human bind to its receptor greatly increases the sensitivity of
CD34-positive cells and keratinocytes in the skin mice to irradiation, suggesting that SCF may play a
(Longley et al., 1993; Ratajczak et al., 1995). While crucial role in recovery of hematopoietic cells from
mRNA levels for the alternatively spliced variants of radiation-induced hypoplasia. In this regard, admin-
SCF, SCF220, and SCF248 vary considerably between istration of SCF alone has been shown to protect
tissues, SCF248 expression is the predominant species mice from the lethal effects of irradiation (Zsebo et al.,
in fibroblasts, brain, thymus, spleen, and bone 1992). Furthermore, it is known that IL-1 or TNF
marrow, while SCF220 is prominent in placenta, cere- can protect mice from the lethal effects of irradiation,
bellum, and testes (Huang et al., 1992; Brannan et al., and that IL-1-mediated radioprotection can be pre-
1991). The mechanisms responsible for regulating the vented by administration of antibodies which block
levels of alternatively spliced SCF mRNA are unknown, SCF/SCFR binding, indicating that IL-1's effects are
as are the enzyme(s) responsible for the proteolytic mediated, in part, through SCF (Neta et al., 1993).
cleavage of SCF248 that produce soluble SCF. While the levels of SCF mRNA expression have not
been determined in cells from patients that have
received myeloablative chemotherapy or radiation
therapy, the concentration of soluble SCF in normal
Eliciting and inhibitory stimuli, human serum ranges from roughly 1 to 3 ng/mL and
is not increased in the serum from patients with
including exogenous and myelodysplasia or aplastic anemia (Langley et al., 1993;
endogenous modulators Bowen et al., 1993; Testa et al., 1994; Abkowitz et al.,
1996). Furthermore, the levels of SCF expression do
Inflammatory stimuli including IL-1, TNF , and not significantly differ between stromal cells from
bacterial pathogens can induce SCF production aplastic anemic patients and stromal cells from
in vitro by bone marrow stromal cells including normal donors, suggesting that levels of soluble
endothelial cells and fibroblasts (Anderson et al., SCF are not predictive for those hematopoietic dis-
1990; Koenig et al., 1994; Linenberger et al., 1995). orders examined. In summary, as is the case for many
In other tissues, SCF can be induced in Sertoli cells HGFs, the physiological mechanisms and signals
with dibutyryl cAMP, and agents that increase the responsible for regulating SCF levels in vivo are
production of intracellular cAMP levels, including unknown.
Stem Cell Factor 885

RECEPTOR UTILIZATION combinations consistently reduces the lymphoid


repopulating ability of the surviving PHSC popula-
SCF utilizes the SCF receptor, which is member of tion, while combinations of SCF and IL-11 maintain
the type III receptor tyrosine kinase family that PHSC activity in liquid culture (Hirayama et al.,
includes c-Fms and PDGFR and Flk-2/Flt-3 recep- 1992, 1994; Holyoake et al., 1996). SCF by itself is a
tors (Besmer et al., 1986; Yarden et al., 1987; Qiu potent survival factor for primitive PHSCs and can be
et al., 1988; Ullrich and Schlessinger, 1990). The SCFR used to transiently maintain the survival of PHSCs
receptor has been given the designation CD117. and their more committed progeny in vitro (Leary
et al., 1992; Li and Johnson, 1994; Keller et al., 1995).
Thus far, SCF does not affect the self-renewal
capability of stem cells alone or in combination with
other HGFs in vitro.
IN VITRO ACTIVITIES Mouse PHSCs and human PHSCs can be main-
tained in co-cultures with stromal, adherent, or
In vitro findings accessory cell populations for extended periods in vitro
(Dexter et al., 1977). The presence of PHSCs in these
SCF alone has little or no direct effect on the cultures is monitored by the continued production of
proliferation of primitive and more committed progenitor cells [CFU ± spleen (CFU-s) or cells that
human and mouse hematopoietic progenitor cells, form macroscopic spleen colonies when transplanted
or stem cells in other tissues. However; in the presence into irradiated mouse recipients, CFU ± culture
of other HGFs, SCF stimulates the proliferation of (CFU-c), and their differentiated progeny (total cell
hematopoietic cells in soft agar (size and number of output)] over time in the nonadherent fraction of
colonies) and liquid cultures (Broxmeyer et al., these cultures. Antibodies that block SCF/SCFR
1991a,b; de Vries et al., 1991; Lowry et al., 1991; interactions inhibit the production of CFU-s, CFU-c,
Metcalf and Nicola, 1991; Migliaccio et al., 1991a,b; and total cell number to nearly undetectable
Tsuji et al., 1991; Williams et al., 1992; Ku et al., levels indicating that SCF is absolutely required to
1996; Ramsfjell et al., 1996, 1997). For example, SCF maintain hematopoietic development in these cultures
directly synergizes with (a) erythropoietin (EPO) to (Kodama et al., 1992; Wineman et al., 1993; Miller
promote unilineage erythroid colony formation et al., 1997).
(burst-forming units ± erythroid, BFU-E), (b) IL-3 Interestingly, the survival and maintenance of the
to promote multilineage colony formation (colony- PHSC population was not impaired in the continued
forming unit ± multipotential, CFU-multi) containing presence of the SCF-blocking antibodies since hema-
granulocytes, macrophages, mast cells, and megakar- topoiesis could be restored in these cultures when the
yocytes (and erythroid cells in the presence of EPO), antibodies were removed. The presence of PHSCs was
(c) GM-CSF to promote granulocyte±macrophage also confirmed in these cultures by transplanting cells
(GM) colony formation, (d) M-CSF or G-CSF to into lethally irradiated mouse recipients in competi-
promote unilineage CFU-G and CFU-M colony tive repopulating assays, which are designed to detect
formation respectively, and (e) thrombopoietin (TPO) PHSC activity. PHSCs can also be maintained on
to stimulate megakaryocytic colony formation in stromal cells that do not produce SCF. Taken together,
soft agar. SCF can promote the survival of PHSCs and is
SCF in combination with multiple HGF combina- absolutely required to maintain hematopoietic
tions that include IL-3, IL-6, and G-CSF can support development in vitro and in vivo, however, there are
the expansion of progenitor cells in liquid culture other HGF or microenvironmental signals that can
including BFU-E, CFU-multi, CFU-G, CFU-M, and support the survival and development of the SCFR-
CFU-GM (Bernstein et al., 1991a; Migliaccio et al., expressing PHSCs (Kodoma et al., 1992; Wineman
1992; Haylock et al., 1992; Brandt et al., 1992). While et al., 1993; Ortiz et al., 1999).
the combination of HGFs (SCF, IL-3, G-CSF, IL-6 SCF can synergize with IL-7 to stimulate pro-B cell
(or IL-11)) can promote the expansion of committed proliferation, however, SCF is not required for pre-B
progenitor cells in vitro, it does not significantly cells to further differentiate and it does not induce the
expand the number of pluripotential hematopoietic proliferation of B220‡ cytoplasmic ‡ pre-B cells
stem cells (PHSCs), which are capable of reconstitut- (Rolink et al., 1991; McNiece et al., 1991b; Billips
ing the entire hematopoietic system, including et al., 1992; Funk et al., 1993). Further, while SCF
lymphoid cells, myeloid cells, erythroid cells, and can affect primitive B cell progenitor cell growth
megakaryocytes when transplanted in vivo (Peters in vitro, there is not a strict requirement for SCF in B
et al., 1996). In fact, inclusion of IL-3 in these HGF cell development because, (1) pro-B cell numbers can
886 Jonathan Roy Keller and Diana Marie Linnekin

expand on stromal cells that do not produce SCF,


and (2) W/W fetal liver cells that lack SCFR expres-
Regulatory molecules: Inhibitors
sion can differentiate into B cells in RAG2 knockout and enhancers
mice, which lack mature B cells (Rolink et al., 1991;
Faust et al., 1993; Takeda et al., 1997). In compari- See section on Eliciting and inhibitory stimuli, includ-
son, there is a critical role for SCF in early T cell ing exogenous and endogenous modulators.
development. SCF can synergize with IL-7 to
stimulate the proliferation of primitive CD4ÿ CD8ÿ
CD3ÿ thymocytes but not more differentiated single Bioassays used
CD4‡ or CD8‡ cells (Godfrey et al., 1992; deCastro
et al., 1994; Morrissey et al., 1994). Furthermore, T The majority of murine factor-dependent cell lines
cell development is also defective in W/W and Sl/Sl derived from mouse hematopoietic tissues are IL-3-
mice where the genes for SCFR and SCF have been dependent and require IL-3 for their continued
completely deleted respectively. In addition, W/W proliferation in vitro. However, some of these cell
fetal liver cells do not give rise to thymocytes when lines also proliferate in response to murine SCF,
transplanted into RAG2 knockout mice (Rodewald including the mast cell lines MC/9 and H-7 originally
et al., 1995; Takeda et al., 1997). used to purify SCF, and some subclones of FDC-P1
The receptor for SCF, SCFR, is expressed on the that were also used to purify IL-3 (Dexter et al., 1980;
majority of hematopoietic progenitors but is not Martin et al., 1990). In addition, there are SCF-
expressed on their differentiated progeny including dependent cell lines, including EML (Tsai et al.,
mature myeloid and monocytic cells or lymphoid and 1994), that require SCF for their continued growth
erythroid cell populations; however, its expression is and proliferation. Human factor-dependent cell lines
maintained at high levels on mast cells. In this regard, have been established by culturing patient leukemic
SCF promotes the survival, proliferation and differ- cells in the presence of HGF in vitro and include
entiation of mast cellsin vitro (Tsai et al., 1991a, the cell lines MO7e and MB-O2 (Avanzi et al., 1990;
1991b; Iemura et al., 1994). In addition, SCF in Hendrie et al., 1991; Broudy et al., 1993). These cell
combination with IL-3 and IL-4 promotes the lines proliferate in response to SCF in a dose-
proliferation of mast cells and their progenitors dependent manner and represent sensitive and reliable
(Kirshenbaum et al., 1992; Rennick et al., 1995). SCF bioassays to determine the specific activity of mouse
can promote the growth and differentiation of natural and human SCF. It should be noted that rodent SCF
killer (NK) cells in combination with IL-2, IL-7, or is active on human cells, while human SCF is 100-fold
IL-15in vitro (Matos et al., 1993; Silva et al., 1994; less active on rodent cell lines, therefore, bioassays
Shibuya et al., 1995; Mrozek et al., 1996). SCF can require mouse or human cell lines where appropriate
also promote the proliferation of the highly effective (Martin et al., 1990).
antigen-presenting dendritic cell population in re-
sponse to GM-CSF or GM-CSF plus TNF (Young
et al., 1995; Szabolcs et al., 1995).
SCF can promote the adhesion of mast cells, IN VIVO BIOLOGICAL
hematopoietic progenitor cells and cell lines to ACTIVITIES OF LIGANDS IN
fibronectin or vascular cell adhesion molecule 1
(VCAM-1) expressed on endothelial cells by activat- ANIMAL MODELS
ing VLA-4 and VLA-5 integrin expression on pro-
genitor cells (Kinashi and Springer, 1994; Dastych Normal physiological roles
and Metcalfe, 1994; Kovach et al., 1995; Levesque,
1996). Membrane-bound SCF can also function as A single injection of SCF to normal mice promotes a
an adhesion molecule for mast cells and progenitor dose-dependent increase in total number of peripheral
cells which express SCFR (Kaneko et al., 1991; Long blood leukocytes, including neutrophils and imma-
et al., 1992; Kodama et al., 1994; Broudy et al., 1996). ture myeloid cells but not circulating platelets or red
As discussed above, SCF is expressed along the cells (Ulich et al., 1991). While total bone marrow
migratory routes for germ cells, melanocytes and cellularity was not significantly altered by this treat-
hematopoietic cells, allowing these cells to home to ment, bone marrow morphology appeared more
proper developmental sites. In this regard, SCF is also primitive with a decrease in the number of mature
a potent chemoattractant for mast cells and progeni- neutrophils and an increase in the number of
tor cells (Meininger et al., 1992; Nilsson et al., 1994; primitive myeloid and erythroid cells (Molineux
Okumura et al., 1996). et al., 1991; Ulich et al., 1991). Daily injections of
Stem Cell Factor 887

SCF for 2 weeks in rodents and nonhuman primates antibody injection while the number of progenitors
promoted a mast cell hyperplasia in many tissues that respond to IL-7 increased. The majority of the
including the bone marrow (Tsai, 1991b; Galli et al., committed CFU-s day 8 and more primitive CFU-s
1993). Daily injections of SCF in nonhuman primates day 12 colonies were also eliminated from the bone
resulted in increased numbers of neutrophils and marrow by this treatment in vivo, however, CFU-s
lymphocytes; however, in contrast to the rodents, day 12 progenitors were not completely eliminated. In
increased numbers of basophils, eosinophils, and similar experiments looking at other organ systems,
monocytes were also observed (Andrews et al., 1991). adult spermatogenesis and melanocyte development
Administration of SCF to rodents also increases the were also blocked by the administration of these
number of progenitors, including CFU-s, short-term antibodies in vivo (Nishikawa et al., 1991). These
reconstituting cells (STRCs ± radioprotective when antibodies also inhibited SCFR/SCF expression in
transplanted in vivo), and PHSCs in the bone marrow, fetal tissues and specifically blocked fetal thymic
peripheral blood, and spleen (increased spleen size colonization by triple negative fetal liver progenitors
and cell number) (Molineux et al., 1991; Fleming in vitro (Godfrey et al., 1994). Furthermore, these
et al., 1993; Bodine et al., 1993; Briddell et al., 1993; antibodies also inhibit the development of CD4‡
de Revel et al., 1994; Yan et al., 1995). This is a con- CD8‡ cells in fetal thymus reconstitution assays
sequence of SCF-induced progenitor/stem cell mobi- (Hozumi et al., 1994). Taken together, these data
lization and redistribution as well as effects on suggest that SCF is critical for the development of
proliferation and expansion of progenitor cells includ- most hematopoietic cell lineages except B cells.
ing PHSCs (Bodine et al., 1993; Fleming et al., 1993;
Harrison et al., 1994).
Increased numbers of hematopoietic progenitor Species differences
cells, including those capable of engrafting lethally
irradiated baboons, were observed in the peripheral Although mouse and human SCF are roughly 82%
blood and bone marrow of nonhuman primates identical at the amino acid level, human SCF is
treated with SCF and could be increased up to 100± 100-fold less active on mouse cells, while mouse
1000-fold in the circulation (Andrews et al., 1991, and rat SCF are equally active on both human and
1992). SCF in combination with G-CSF was more rodent cells.
effective in mobilizing progenitor/stem cells capable
of protecting lethally irradiated baboons; however,
whether these progenitors include PHSCs remains to Knockout mouse phenotypes
be determined (Andrews et al., 1994, 1995; McNiece
and Briddell, 1995). In comparison, the combination The phenotypes of mice with naturally occurring
of SCF and G-CSF showed a rapid increase in the mutations in SCF or the Sl locus demonstrate that the
number of PHSCs in the peripheral blood and bone SCF/SCFR axis is critical for hematopoiesis, mela-
marrow of mice after 14 days (Bodine et al., 1996). nogenesis, and gametogenesis. Sl mutant mice that
Thus, SCF alone or in combination with G-CSF have completely deleted the coding sequences for
effectively mobilizes stem cells into the periphery SCF do not produce viable homozygous offspring;
and provides a convenient source of stem cells for embryos only survive to roughly 15 days gestation. In
transplantation. comparison, Sl mutant mice where SCF sequences are
SCF has also been shown to be critical for main- present, produce viable homozygous offspring that
taining constitutive hematopoiesis in studies where show defects in hematopoietic development, pigment-
animals were administered antibodies that block SCF ation, and germ cell development (sterility) depending
function by inhibiting the binding of SCF to its on the severity of the mutation. Thus, null alleles are
receptor. Treatment of mice with 1 mg of antibody lethal, while viable alleles retain some of the normal
every other day for 2 weeks resulted in the elimination function of SCF (summarized in Table 1). For
of erythroid cells followed by myeloid cells, while example, Sl gb, Sl J, Sl10H, Sl, Sl 8H, Sl 12H, and Sl 18H
total bone marrow cellularity remained unchanged. have complete deletions of the SCF gene and all
However, the majority of cells (90%) found in the homozygous embryos die at roughly 15 days gesta-
bone marrow were B220-positive (normally 30±40%), tion with severe defects in fetal liver hematopoiesis
including pre-B cells that express cytoplasmic  without effects on other organ systems (Russell, 1979;
chains, and IgM-bearing B lymphocytes (Ogawa et al., Silvers, 1979; Copeland et al., 1990; Huang et al.,
1991; Rico-Vargas et al., 1994). Myeloid progenitor 1990; Zsebo et al., 1990b). The smallest complete
cell populations including those that respond to IL-3, deletion in SCF coding sequences occurs in Sl gb mice
GM-CSF, and M-CSF declined after a single where the total genomic deletion is roughly 120 kb.
888 Jonathan Roy Keller and Diana Marie Linnekin

Table 1 Summary of Steel locus mutants

Gene symbol Gene name Mutation Consequence of mutation Phenotype of


homozygous mice

Sl gb Steel-Grizzle Belly 120 kb deletion Absence of SCF Prenatal lethality


J
Sl Steel-J 650 kb deletion Absence of SCF Prenatal lethality
10H
Sl Steel-10H 680 kb deletion Absence of SCF Prenatal lethality
Sl Steel > 810 kb deletion Absence of SCF Prenatal lethality
Sl d Steel dickie 4 kb intragenic deletion Lack of membrane-bound Severely anemic
SCF
Lack of pigmentation
Sterility in both sexes
17H
Sl Steel-17H Point mutation Absence of normal Macrocytic anemia
in splice acceptor cytoplasmic SCF domain
White spotting
Male sterility
Sl pan Steel-Panda Rearrangement/ Decreased SCF expression Mild anemia
inversion >100 kb Decreased pigmentation
50 SCF gene
Female sterility
con
Sl Steel-contrasted Rearrangement Decreased SCF expression Mild anemia
0
> 100 kb 5 SCF gene Decreased pigmentation
Female sterility

This deletion begins in the 30 untranslated region of the development of three distinct cell lineages,
SCF and includes the entire SCF coding sequences including hematopoietic cells.
of roughly 50 kb, as well as 60 kb of DNA sequence Another informative viable mutant of the Sl locus
50 of the SCF coding region (Bedell et al., 1996). is the Sl17H mutation, which is a splice site mutation
Furthermore, the deletions detected in Sl J and Sl10H that causes exon 8 to be skipped, resulting in the
(larger than the deletion in Sl gb) are also smaller splicing of exon 7 with exon 9. The deletion of exon 8
deletions than those seen in the original Sl allele which encodes the cytoplasmic domain of SCF results
where lethality occurred at the same time during in a frameshift mutation that replaces the normal
gestation. In summary, late gestation lethality appears SCF cytoplasmic domain with extraneous amino
to be the true null phenotype; however, whether other acids before a stop codon is encountered (Brannan
genes are deleted in Sl mice with larger genomic dele- et al., 1992). This mutation does not affect the ability
tions remains to be determined. of SCF to form dimers but slows the rate of SCF
The best-characterized mouse strain with a natu- transport to the cell surface and subsequent stability
rally occurring viable mutation of SCF is the Sl d/Sl d (Tajima et al., 1998a). Total bone marrow cellularity,
mouse. These mice have a 4 kb intragenic deletion leukocyte counts, platelets and hematocrit are un-
which removes the exons encoding the transmem- affected in these mice. However, myeloid progenitor
brane and cytoplasmic domains of SCF resulting in cells and tissue mast cells are slightly decreased. long-
the Sl d transcript, which encodes the soluble isoform term bone marrow culture cells (LTBMCs) estab-
of SCF (Brannan et al., 1991; Flannagan et al., 1991). lished from these mice show a greatly reduced ability
These mice are viable but have severe macrocytic to support the production of total hematopoietic cell
anemia with mast cell deficiencies, sterility in both output. Furthermore, these mice have reduced levels
sexes, and lack skin pigmentation. The bone marrow of CFU-s progenitor cells. Further, male mice are
of Sld is hypocellular with both myeloid and erythroid sterile while female mice are unaffected.
progenitor cell deficiencies. Thus, it is speculated that Two other Sl mouse strains including Sl pan and
the membrane-associated form of SCF is critical for Sl con have DNA rearrangements roughly 100 kb 50 of
Stem Cell Factor 889

the SCF coding sequences (Bedell et al., 1995). These maturation, suggesting that melanoblasts, dermal
mutations affect SCF mRNA expression in the mast cells, and thymocyte progenitors are not able to
gonads and lead to reduced numbers of primordial migrate to the appropriate sites, or are unable to
germ cells and result in female sterility (male sterility survive and proliferate once they arrive. Transgenic
is not observed). mouse strains were constructed which restricted
the expression of SCF248 or SCF220 transgenes to
keratinocytes in normal mice (Kunisada et al., 1998).
These studies found that expression of SCF248 in
Transgenic overexpression keratinocytes results in cutaneous mastocytosis and
epidermal hyperpigmentation due to the maintenance
To better understand the physiological roles of of melanocytes and melanin production in the epi-
soluble versus membrane isoforms of SCF, Sl d dermis. In comparison, expression of the membrane-
mice were engineered to stably express soluble and restricted form of SCF, SCF220, in keratinocytes
membrane-restricted SCF transgenes. Both Sl d promoted epidermal melanocytosis and melanin
transgenic mouse strains showed increases in the production without mastocytosis. This suggests that
number of multipotential and more committed soluble SCF is required for dermal mast cell migra-
myeloid progenitor cells in the bone marrow and tion and/or proliferation while membrane-associated
increased numbers of peripheral blood leukocytes SCF is required for melanocyte growth and differ-
(mainly neutrophils) (Kapur et al., 1998). However, entiation. In other studies, expression of SCF248 in
only Sl d mice that expressed the membrane-restricted keratinocytes also promoted pigmentation in sites
isoform of SCF showed (1) a reversal in the severe which normally do not contain melanocytes or their
runting phenotype, (2) an increase in bone marrow precursors, suggesting that SCF can also stimulate the
cellularity, and (3) an increase in the hematocrit and migration of melanocytes in vivo (Kunisada et al.,
red cell count. However, neither transgene could 1998).
rescue the defects in germ cells, melanocytes, or mast
cells in Sl d mice. The lack of effect of either SCF
transgene on other organ systems may be due to the Pharmacological effects
choice of promoter in these constructs which could
have restricted the transgene expression to specific None reported for mice.
cell types (Kapur et al., 1998). In other experiments,
transgenic mouse strains were engineered to exclu-
sively produce the membrane-restricted form of SCF Interactions with cytokine network
(SCF220 or KL-2) by homologous recombination
in embryonic stem cells. No effect on hematopoiesis None reported in vivo.
including peripheral blood counts or bone marrow
cellularity or progenitor numbers were observed in
these mice (Tajima et al., 1998b). Furthermore, there Endogenous inhibitors and
were no observable effects on gametogenesis since enhancers
male and female mice produced normal numbers of
offspring. However, these mice showed defects in the See section on Eliciting and inhibitory stimuli, includ-
production of dermal mast cells, and were also ing exogenous and endogenous modulators.
sensitive to sublethal doses of gamma-irradiation,
suggesting a role for soluble SCF in hematopoietic
recovery following radiation injury.
Because human SCF can act as an antagonist of PATHOPHYSIOLOGICAL ROLES
mouse SCF, transgenic mice were produced that IN NORMAL HUMANS AND
express human membrane-restricted SCF (SCF220) DISEASE STATES AND
to separate the effects of soluble and transmembrane
SCF (Kapur et al., 1997, 1998). No significant effect DIAGNOSTIC UTILITY
on hematopoiesis was observed in these mice,
including total leukocyte counts and red cell numbers Normal levels and effects
in the peripheral blood, or bone marrow cellularity
and progenitor numbers. However, these mice have a Normal plasma levels of human SCF range from 1
coat color defect, have greatly reduced numbers of to 5 ng/mL (Langley et al., 1993). In addition, SCF
dermal mast cells, and show abnormal thymocyte levels do not significantly vary in patients with a
890 Jonathan Roy Keller and Diana Marie Linnekin

variety of hematological disorders including aplastic blood. Therefore, it is speculated that the major effect
anemia and myelodysplastic syndromes (McNiece of SCF treatment in Sl/Sl d mice is to promote the
and Briddell, 1995; Kumar and Alter, 1998). proliferation of immature megakaryocytes and their
precursors, while other factors such as thrombopoietin
are critical for the final stages of platelet maturation.
Role in experiments of nature and
disease states Pharmacokinetics
While no abnormalities involving the SCF genetic loci Baseline SCF serum levels in patients that were to
have been reported, locally high concentrations of receive SCF treatment in a phase I clinical trial were
soluble SCF have been found in lesions of human roughly 1 ng/mL (Glaspy, 1996). SCF serum levels
cutaneous mastocytosis (Longley et al., 1993, 1995). reached a maximum 13±14 hours after the first dose
This disease is characterized by accumulations of of SCF. Maximum serum SCF levels were observed
mast cells as well as increases in the production of between 10 and 14 hours after subsequent doses and
epidermal melanin similar to that observed in trans- SCF. Adsorption of SCF was relatively low after the
genic animals that expressed SCF transgenes in first dose of SCF.
keratinocytes (described above). This has led to the
hypothesis that locally produced SCF can promote
mast cell hyperplasia. Toxicity
The use of SCF in vivo has been limited due to adverse
IN THERAPY side effects. These included dermatological reactions
such as pruritic wheals and edema at the site of
injection as well as increased melanization (pigmenta-
Preclinical ± How does it affect tion) of the epidermis at doses above 25 mg/kg body
disease models in animals? weight over a 14-day period (Demetri et al., 1993;
Costa et al., 1996; Weaver et al., 1996; Moskowitz
SCF administration to Sl/Sl d mice reverses the et al., 1997). In addition, 10±20% of the patients
macrocytic anemia resulting in increased mean red treated with SCF developed allergic-like reactions
blood cell volume and red blood cell counts that are characterized by urticaria with respiratory symptoms
within 70±85% of their littermate control levels. This that required discontinuation of treatment (Grichnik
effect was reversed when SCF treatment was stopped et al., 1995; Costa et al., 1996). Because mast cell
(Hunt et al., 1992; Galli et al., 1994). Increased hyperplasia was suspected in these cases, a prophy-
numbers of lymphocytes and platelets were also lactic antihistamine treatment was incorporated into
observed in Sl/Sl d mice treated with SCF, which were subsequent protocols (McNiece and Briddell, 1995).
also observed in normal mice treated with SCF.
Injection of SCF into Sl/Sl d mice caused a two-fold
and 20-fold increase in the number of primitive Clinical results
progenitors (CFU-s day 12) found in the bone
marrow and spleen, respectively (Bodine et al., 1992). A phase I clinical trial of SCF was conducted in lung
Sl/Sl d mice do not experience a rebound in platelet cancer and breast cancer patients who received 10±
counts after 5-FU treatment, while normal mice show 50 mg/kg/day SCF by subcutaneous injection for 14
an overshoot between 10 and 20 days post 5-FU days (Glaspy, 1996; Demitri et al., 1993). An increase
treatment (Hunt et al., 1992). To determine whether in the total number of peripheral blood leukocytes
SCF was critical for rebound (overshoot) thrombocy- was observed prior to chemotherapy and some
tosis after 5-FU treatment in Sl/Sl d mice, mice were acceleration in platelet and leukocyte numbers was
treated with SCF after 5-FU. Administration of SCF seen after chemotherapy. Daily administration of
to 5-FU-treated Sl/Sl d mice promoted the same over- SCF to patients at dosages of up to 50 mg/kg for 14
shoot as that seen in control 5-FU-treated mice, days did not affect the number of peripheral blood
demonstrating the importance of SCF in megakaryo- CD34‡ cells; however, increased numbers of CD34‡
cyte growth and development. Megakaryocyte pro- cells, that included both committed and more
genitor (CFU-MK) numbers are increased in the bone primitive hematopoietic progenitor cells, were de-
marrow and spleen of normal mice treated with SCF, tected in the bone marrow (Tong et al., 1993). SCF
while platelet counts are not increased in the peripheral therapy also promoted an increase in bone marrow
Stem Cell Factor 891

cellularity with significant increases in promyelocytes (1992). The ligand for c-kit, stem cell factor, stimulates the
without apparent effects on other marrow hemato- circulation of cells that engraft lethally irradiated baboons.
Blood 80, 2715.
poietic cells (Orazi et al., 1995). Andrews, R. G., Briddell, R. A., Knitter, G. H., Opie, T.,
Because SCF was shown to potently mobilize Bronsden, M., Myerson, D., Appelbaum, F. R., and
progenitor cells in mice and nonhuman primates, McNiece, I. K. (1994). In vivo synergy between recombinant
subsequent clinical trials have focused on the ability human stem cell factor and recombinant human granulocyte
of SCF (in lower doses) in combination with G-CSF colony-stimulating factor in baboons: enhanced circulation of
progenitor cells. Blood 84, 800.
to mobilize peripheral blood progenitor cells for Andrews, R. G., Briddell, R. A., Knitter, G. H., Rowley, S. D.,
transplantation. A phase I/II trial in non-Hodgkin's Appelbaum, F. R., and McNiece, I. K. (1995). Rapid engraft-
lymphoma patients was performed to compare ment by peripheral blood progenitor cells mobilized by recom-
mobilized peripheral blood progenitor cells for binant human stem cell factor and recombinant human
autologous transplantation from patients treated granulocyte colony-stimulating factor in nonhuman primates.
Blood 85, 15.
with the combination of SCF plus G-CSF or G- Arakawa, T., Yphantis, D. A., Lary, J. W., Narhi, L. O., Lu, H. S.,
CSF alone (Moskowitz et al., 1997). Patients trans- Prestrelski, S. J., Clogston, C. L., Zsebo, K. M., Mendiaz, E. A.,
planted with G-CSF plus SCF-mobilized peripheral and Wypych, J. (1991). Glycosylated and unglycosylated recom-
blood cells showed a decreased time to normal binant-derived human stem cell factors are dimeric and have
platelet levels following chemotherapy compared with extensive regular secondary structure. J. Biol. Chem. 266, 18942.
Avanzi, G.C. Brizzi, M. F., Ginnoti, J., Ciarletta, A., Yang, Y. C.,
transplanted G-CSF-mobilized peripheral blood. In Pegararo, L., and Clark, S. C. (1990). M-O7e human leukemic
another phase II trial, fewer apheresis procedures factor-dependent cell line provides a rapid and sensitive bioas-
were required to collect sufficient numbers of peri- say for the human cytokines GM-CSF and IL-3. J. Cell. Phys.
pheral blood progenitor cells from breast cancer 145, 458.
patients for transplantation studies (Kumar and Alter, Avraham, H., Vannier, E., Cowley, S., Jiang, S. X., Chi, S.,
Dinarello, C. A., Zsebo, K. M., and Groopman, J. E. (1992).
1998). In a phase I study of patients with refractory Effects of the stem cell factor, c-kit ligand, on human megakar-
aplastic anemia, SCF was well tolerated at all doses yocytic cells. Blood 79, 365.
and patients that received SCF showed neutrophil Aye, M. T., Hashemi, S., Leclari, B., Zeibdawi, A., Trudel, E.,
improvements that were improved by the combina- Halpenny, M., Fuler, V., and Cheng, G. (1992). Expression of
tion with G-CSF (Kumar and Alter, 1998). stem cell factor and c-kit mRNA in cultured endothelial cells,
monocytes and cloned human bone marrow stromal cells
(CFU-RF). Exp. Hematol. 20, 523.
Bazan, J. F. (1991). Genetic and structural homology of stem cell
References factor and macrophage colony-stimulating factor. Cell 65, 9.
Bedell, M. A., Brannan, C. I., Evans, E. P., Copeland, N. G.,
Abkowitz, J. L., Hume, H., Yancik, S. A., Bennett, L. G., and Jenkins, N. A., and Donovan, P. J. (1995). DNA rearrange-
Matsumoto, A. M. (1996). Stem cell factor serum levels may not ments located over 100 kb 5 of the Steel (Sl)-coding region in
be clinically relevant. Blood 87, 4017. Steel-panda and Steel-contrasted mice deregulate Sl expression
Adachi, S., Ebi, Y., Nishikawa, S., Yamazaki, M., and and cause female sterility by disrupting ovarianfollicle develop-
Kasugai, T. (1992). Necessity of extracellular domain of W ment. Genes Dev. 9, 4.
(SCFR) receptors for attachment of murine cultured mast Bedell, M. A., Copeland, N. G., and Jenkins, N. A. (1996).
cells to fibroblasts. Blood 79, 650. Multiple pathways for Steel regulation suggested by genomic
Altus, M. S., Bernstein, S. E., Russell, E. S., Carsten, A. L., and and sequence analysis of the murine Steel gene. Genetics 142,
Upton, A. C. (1971). Defect extrinsic to stem cells in spleens of 927.
Steel anemic mice. Proc. Soc. Exp. Biol. Med. 138, 985. Bernstein, I. D., Andrews, R. G., and Zsebo, K. M. (1991a).
Anderson, D. M., Lyman, S. D., Baird, A., Wignall, J. M., Recombinant human stem cell factor enhances the formation
Eisenman, J., Rauch, C., March, C. J., Boswell, H. S., of colonies by CD34‡ and CD34‡lin cells, and the generation
Gimpel, S. D., and Cosman, D. (1990). Molecular cloning of of colony-forming cell progeny from CD34‡lin cells cultured
mast cell growth factor, a hematopoietin that is active in both with interleukin-3, granulocyte colony-stimulating factor, or
membrane bound and soluble forms. Cell 63, 235. granulocyte-macrophage colony-stimulating factor. Exp.
Anderson, D. M., Williams, D. E., Tushinski, R., Gimpel, S., Hematol. 77, 2316.
Eisenman, J., Cannizzaro, L. A., Aronson, M., Croce, C. M., Bernstein, A., Forrester, L., Reith, A. D., Dubreuil, P., and
Huebner, K., and Cosman, D. (1991). Alternate splicing of Rottapel, R. (1991b). The murine W/SCFR and Steel loci and
mRNAs encoding human mast cell growth factor and localiza- the control of hematopoiesis. Semin. Hematol. 28, 138.
tion of the gene to chromosome 12q22-q24. Cell Growth Differ. Besmer, P., Murphy, J. E., George, P. C., Qiu, F. H.,
2, 373. Bergold, P. J., Lederman, L., Snyder, H.W. Jr,, Brodeur, D.,
Andrews, R. G., Knitter, G. H., Bartelmez, S. H., Langley, K. E., Zuckerman, E. E., and Hardy, W. D. (1986). A new acute
Farrar, D., Hendren, R. W., Appelbaum, F. R., Bernstein, I. D., transforming feline retrovirus and relationship to its oncogene
and Zsebo, K. M. (1991). Recombinant human stem cell factor, v-kit with the protein kinase gene family. Nature 320, 415±421.
a SCFR ligand, stimulates hematopoiesis in nonhuman pri- Besmer, P., Manova, K., Duttlinger, R., Huang, E. J., Packer, A.,
mates. Blood 78, 1975. Gyssler, C., and Bachvarova, R. F. (1993). The kit-ligand (steel
Andrews, R. G., Bensinger, W. I., Knitter, G. H., Bartelmez, S. H., factor) and its receptor SCFR/W: Pleiotropic roles in gameto-
Longin, K., Bernstein, I. D., Appelbaum, F. R., and Zsebo, K. M. genesis and melanogenesis. Development 125,
892 Jonathan Roy Keller and Diana Marie Linnekin

Billips, L. G., Petitte, D., Dorshkind, K., Narayanan, R., Copeland, N. G., Gilbert, D. J., Cho, B. C., Donovan, P. J.,
Chiu, C. P., and Landreth, K. S. (1992). Differential roles of Jenkins, N. A., Cosman, D., Anderson, D., Lyman, S. D.,
stromal cells, interleukin-7, and kit-ligand in the regulation of B and Williams, D. E. (1990). Mast cell growth factor maps
lymphopoiesis. Blood 79, 1185. near the steel locus on mouse chromosome 10 and is deleted
Bodine, D. M., Orlic, D., Birkett, N. C., Seidel, N. E., and in a number of steel alleles. Cell 63, 175.
Zsebo, K. M. (1992). Stem cell factor increases colony-forming Costa, J. J., Demetri, G. D., Harrist, T. J., Dvorak, A. M.,
unit-spleen number in vitro in synergy with interleukin-6, and Hayes, D. F., Merica, E. A., Menchaca, D. M., Gringeri, A. J.,
in vivo in Sl/Sld mice as a single factor. Blood 79, 913. Schwartz, L. B., and Galli, S. J. (1996). Recombinant human
Bodine, D. M., Seidel, N. E., Zsebo, K. M., and Orlic, D. (1993). stem cell factor (Kit ligand) promotes human mast cell and
In vivo administration of stem cell factor to mice increases the melanocyte hyperplasia and funcational activation in vivo.
absolute number of pluripotent hematopoietic stem cells. Blood J. Exp. Med. 183, 2681.
82, 445. Dastych, J., and Metcalfe, D. D. (1994). Stem cell factor induces
Bodine, D. M., Seidel, N. E., and Orlic, D. (1996). Bone marrow mast cell adhesion to fibronectin. J. Immunol. 152, 213.
collected 14 days after in vivo administration of granulocyte de Vries, P., Brasel, K. A., Eisenman, J. R., Alpert, A. R., and
colony-stimulating factor and stem cell factor to mice has 10- Williams, D. E. (1991). The effect of recombinant mast cell
fold more repopulating ability than untreated bone marrow. growth factor on purified murine hematopoietic stem cells.
Blood 88, 89. J. Exp. Med. 173, 1205.
Bowen, D., Yancik, S., Bennett, L., Culligna, D., and Resser, K. de Revel, T., Appelbaum, F. R., Storb, R., Schuening, F.,
(1993). Serum stem cell factor concentration in patients with Nash, R., Deeg, J., McNiece, I., Andrews, R., and Graham, T.
myelodysplastic syndromes. Br. J. Haematol. 85, 63. (1994). Effects of granulocyte colony-stimulating factor and
Brandt, J., Briddell, R. A., Srour, E. F., Leemhuis, T. B., and stem cell factor, alone and in combination, on the mobilization
Hoffman, R. (1992). Role of SCFR ligand in the expansion of of peripheral blood cells that engraft lethally irradiated dogs.
human hematopoietic progenitor cells. Blood 79, 634. Blood 83, 3795.
Brannan, C. I., Lyman, S. D., Williams, D. E., Eisenman, J., deCastro, C. M., Denning, S. M., Langdon, S., Vandenbark, G. R.,
Anderson, D. M., Cosman, D., Bedell, M. A., Jenkins, N. A., Kurtzberg, J., Scearce, R., Haynes, B. F., and Kaufman, R. E.
and Copeland, N. G. (1991). Steel-dickie mutation encodes a (1994). The SCFR proto-oncogene receptor is expressed on a
SCFR ligand lacking transmembrane and cytoplasmic domains. subset of human CD3-CD4-CD8- (triple-negative) thymocytes.
Proc. Natl Acad. Sci. USA 88, 4671. Exp. Hematol. 22, 1025.
Brannan, C. I., Bedell, M. A., Resnick, J. L., Eppig, J. J., Demetri, G., Costa, J., Hayes, D., Sledge, G., Galli, S.,
Handel, M. A., Williams, D. E., Lyman, S. D., Donovan, P. J., Hoffman, R., Merica, E., Rich, W., Harkins, B., McGuire, B.,
Jenkins, N. A., and Copeland, N. G. (1992). Developmental and Gordon, M. (1993). A phase I trial of recombinant methio-
abnormalities in Steel17H mice result from a splicing defect in nyl human stem cell factor (SCF) in patients with advanced
the steel factor cytoplasmic tail. Genes Dev. 6, 1832. breast carcinoma pre- and post-chemotherapy with cyclopho-
Briddell, R. A., Hartley, C. A., Smith, K. A., and McNiece, I.K sphamide and doxorubicin. Proc. Am. Assoc. Clin. Oncol. 12,
(1993). Recombinant rat stem cell factor synergizes with recom- A367.
binant human granulocyte colony-stimulating factor in vivo in Dexter, T. M., Allen, T. D., and Lajtha, L. G. (1977). Conditions
mice to mobilize peripheral blood progenitor cells that have controlling the proliferation of hematopoietic stem cells in vitro.
enhanced repopulating potential. Blood 82, 1720. J. Cell. Physiol. 91, 335.
Broudy, V. C. (1997). Stem cell factor and hematopoiesis. Blood Dexter, T. M., Garland, J., Scott, D., Scolnick, E., and
90, 1345. Metcalf, D. (1980). Growth of factor-dependent precursor cell
Broudy, V. C., Morgan, D. A., Lin, N., Zsebo, K. M., lines. J. Exp. Med. 152, 1036.
Jacobsen, F. W., and Papayannopoulou, T. (1993). Stem cell Dunham, S. P., and Onions, D. E. (1995). The cloning and sequenc-
factor influences the proliferation and erythroid differentiation ing of cDNAs encoding two isoforms of feline stem cell factor.
of MB-O2 human erythroleukemia cell line by binding to a DNA Sequence 6, 233.
high-affinity SCFR receptor. Blood 82, 436. Faust, E. A., Saffran, D. C., Toksoz, D., Williams, D. A., and
Broudy, V. C., Morgan, D. A., Lin, N., Zsebo, K. M., Witte, O. N. (1993). Distinctive growth requirements and gene
Jacobsen, F. W., and Papayannopoulou, T. (1996). Interaction expression patterns distinguish progenitor B cells from pre-B
of stem cell factor and its receptor SCFR mediates lodgment cells. J. Exp. Med. 177, 915.
and acute expansion of hematopoietic cells in the murine spleen. Finotto, S., Mekori, Y. A., and Metcalfe, D. D. (1997).
Blood 88, 75. Glucocorticoids decrease tissue mast cell number by reducing
Broxmeyer, H. E., Hangoc, G., Cooper, S., Anderson, D., the production of the SCFR ligand, stem cell factor, by resident
Cosman, D., Lyman, S. D., and Williams, D. E. (1991a). cells: in vitro and in vivo evidence in murine system. J. Clin.
Influence of murine mast cell growth factor (SCFR ligand) on Invest. 99, 1721.
colony formation by mouse marrow hematopoietic progenitor Flannagan, J. G., and Leder, P. (1990). The kit ligand: A
cells. Exp. Hematol. 19, 143. cell surface molecule altered in steel mutant fibroblasts. Cell
Broxmeyer, H. E., Cooper, S., Lu, L., Hangoc, G., Anderson, D., 63, 185.
Cosman, D., Lyman, S. D., and Williams, D. E. (1991b). Effect Flannagan, J. G., Chan, D. C., and Leder, P. (1991). Trans-
of murine mast cell growth factor (SCFR proto-oncogene membrane form of the kit ligand growth factor is determined
ligand) on colony formation by human marrow hematopoietic by alternative splicing and is missing in the Sld mutant. Cell
progenitor cells. Blood 77, 2142. 64, 1025.
Charbot, B., Stephenson, D. A., Chapman, V. M., Besmer, P., and Fleming, W. H., Alpern, E. J., Uchida, N., Ikuta, K., and
Bernstein, A. (1988). The protooncogene SCFR encoding a Weissman, I. L. (1993). Steel factor influences the distribution
transmembrane tyrosine kinase recpetor maps to the mouse W and activity of murine hematopoietic stem cells in vivo. Proc.
locus. Nature 355, 88. Natl Acad. Sci. USA 90, 3760.
Stem Cell Factor 893

Funk, P. E., Varas, A., and Witte, P. L. (1993). Activity of stem posttransplant and the ability to serially transplant marrow.
cell factor and IL-7 in combination on normal bone marrow B Blood 87, 4589.
lineage cells. J. Immunol. 150, 748. Hozumi, K., Kobori, A., Sato, T., Nozaki, H., Nishikawa, S.,
Galli, S. J., Iemura, A., Garlick, D. S., Gamba-Vitalo, C., Nishimura, T., and Habu, S. (1994). Pro-T cells in fetal thymus
Zsebo, K. M., and Andrews, R. G. (1993). Reversible expansion express SCFR and RAG-2 but do not rearrange the gene encod-
of primate mast cell populations in vivo by stem cell factor. ing the T cell receptor beta chain. Eur. J. Immunol. 24, 1339.
J. Clin. Invest. 91, 148. Hsu, Y., Wu, G. M., Mendiaz, E. A., Syed, R., Wypych, J.,
Galli, S. J., Zsebo, K. M., and Geissler, E. N. (1994). The kit Toso, R., Mann, M. B., Boone, T. C., Narhi, L. O., Lu, H. S.,
ligand, stem cell factor. Adv. Immunol. 55, 1±96. and Langley, K. E. (1997). The majority of stem cell factor
Geissler, E. N., McFarland, E. C., and Russell, E. S. (1981). exists as monomer under physiological conditions. J. Biol.
Analysis of pleiotropism at the dominant white-spotting (W) Chem. 272, 64.
locus of the house mouse: A description of ten new W alleles. Huang, E., Nocka, K., Beier, D. R., Chu, T. Y., Buck, J.,
Genetics 97, 337. Lahm, H. W., Wellner, D., Leder, P., and Besmer, P. (1990).
Geissler, E. N., Ryan, M. A., and Housman, D. E. (1988). The The hematopoietic growth factor KL is encoded by the Sl locus
dominant-white spotting (W) locus of the mouse encodes the and is the ligand of the SCFR receptor, the gene product of the
SCFR proto-oncogene. Cell 55, 185. W locus. Cell 63, 225.
Geissler, E. N., Liao, M., Brook, J. D., Martin, F. H., Huang, E. J., Nocka, K. H., Buck, J., and Besmer, P. (1992).
Zsebo, K. M., Housman, D. E., and Galli, S. J. (1991). Stem Differential expression and processing of two cell associated
cell factor (SCF ), a novel hematopoietic growth factor and forms of the kit-ligand: KL-1 and KL-2. Mol. Biol. Cell. 3, 349.
ligand for SCFR tyrosine kinase receptor, maps on human Hunt, P., Zsebo, K. M., Hokom, M. M., Hornkohl, A.,
chromosome 12 between 12q14.3 and 12qter. Somatic Cell. Birkett, N. C., del Castillo, J. C., and Martin, F. (1992).
Mol. Genet. 17, 207. Evidence that stem cell factor is involved in the rebound throm-
Glaspy, J. (1996). Clinical applications of stem cell factor. Curr. bocytosis that follows 5-Fluorouracil treatment. Blood 80, 904.
Opin. Hematol. 3, 223. Iemura, A., Tsai, M., Ando, A., Wershil, B. K., and Galli, S. J.
Godfrey, D. I., Zlotnik, A., and Suda, T. (1992). Phenotypic and (1994). The c-kit ligand, stem cell factor, promotes mast cell
functional characterization of SCFR expression during survival by suppressing apoptosis. Am. J. Pathol. 144, 321±328.
intrathymic T cell development. J. Immunol. 149, 2281. Jacobsen, S. E. W., Jacobsen, F. W., Fahlman, C., and
Godfrey, D. I., Kennedy, J., Gately, M. K., Hakimi, J., Rusten, L. S. (1994). TNF-alpha, the great imitator: Role of
Hubbard, B. R., and Zlotnik, A. (1994). IL-12 influences p55 and p75 TNF receptors in hematopoiesis. Stem Cells 12,
intrathymic T cell development. J. Immunol. 152, 2729. 111.
Grichnik, J. M., Crawford, J., Jimenez, F., Kurtzberg, J., Jacobsen, F. W., Stokke, T., and Jacobsen, S. E. W. (1995a).
Buchanan, M., Blackwell, S., Clark, R. E., and Hitchcock, M. G. Transforming growth factor-beta potently inhibits the viabi-
(1995). Human recombinant stem-cell factor induces melanocy- lity-promoting activity of stem cell factor and other cytokines
tic hyperplasia in susceptible patients. J. Am. Acad. Dermatol. and induces apoptosis of primitive murine hematopoietic pro-
33, 577. genitor cells. Blood 86, 2957.
Harrison, D. E., Zsebo, K. M., and Astle, C. M. (1994). Splenic Jacobsen, F. W., Dubois, C. M., Rusten, L. S., Veiby, O. P., and
primitive hematopoietic stem cell (PHSC) activity is enhanced Jacobsen, S. E. (1995b). Inhibition of stem cell factor-induced
by steel factor because of PHSC proliferation. Blood 83, 3146. proliferation of primitive murine hematopoietic progenitor cells
Haylock, D. N., To, L. B., Dowse, T. L., Juttner, C. A., and signaled through the 75-kilodalton tumor necrosis factor recep-
Simmons, P. J. (1992). Ex vivo expansion and maturation of tor. Regulation of SCFR and p53 expression. J. Immunol. 154,
peripheral blood CD34‡ cells into the myeloid lineage. Blood 3732.
80, 1405. Jacobsen, F. W., Veiby, O. P., Stokke, T., and Jacobsen, S. E.
Hendrie, P. C., Miyazawa, K., Yang, Y. C., Langefeld, C. D., and (1996). TNF-alpha bidirectionally modulates the viability of
Broxmeyer, H. E. (1991). Mast cell growth factor (SCFR primitive murine hematopoietic progenitor cells in vitro.
ligand) enhnaces cytokine stimulation of proliferation of the J. Immunol. 157, 1193.
human factor-dependent cell line, MO7e. Exp. Hematol. 19, Jiang, C., Hall, S. J., and Boekelheide, K. (1997). Cloning and
1031. characterization of the 50 flanking region of the stem cell factor
Heinrich, M. C., Dooley, D. C., Freed, A. C., Band, L., gene in rat sertoli cells. Gene 185, 285.
Hoatlin, M. E., Keeble, W. W., Peters, S. T., Silvey, K. V., Jones, M. D., Narhi, L. O., Change, W. C., and Lu, H. S. (1996).
Ey, F. S., and Kabat, D. (1993). Constitutive expression of Refolding and oxidation of recombinant human stem cell factor
steel factor gene by human stromal cells. Blood 82, 771. produced in Escherichia coli. J. Biol. Chem. 271, 11301.
Heldin, C. H. (1995). Dimerization of cell surface receptors in Kaneko, Y., Takenawa, J., Yoshida, O., Fujita, K., Sugimoto, K.,
signal transduction. Cell 80, 213. Nakayama, H., and Fujita, J. (1991). Adhesion of mouse mast
Hirayama, F., Shih, J. P., Awgulewitsch, A., Warr, G. W., cells to fibroblasts: Adverse effects of steel (Sl) mutation. J. Cell.
Clark, S. C., and Ogawa, M. (1992). Clonal proliferation of Physiol. 147, 224.
murine lymphohemopoietic progenitors in culture. Proc. Natl Kapur, R., Everett, E. T., Uffman, J., McAndrews-Hill, M.,
Acad. Sci. USA 89, 5907. Cooper, R., Ryder, J., Vik, T., and Williams, D. A. (1997).
Hirayama, F., Clark, S. C., and Ogawa, M. (1994). Negative Overexpression of human stem cell factor impairs melanocyte,
regulation of early B lymphopoiesis by interleukin 3 and inter- mast cell, and thymocyte development: a role for receptor tyr-
leukin 1. Proc. Natl Acad. Sci. USA 91, 469. osine kinase-mediated mitogen ativated protein kinase activa-
Holyoake, T. L., Freshney, M. G., McNair, L., Parker, A. N., tion in cell differentiation. Blood 90, 3018.
McKay, P. J., Steward, W. P., Fitzsimons, E., Graham, G. J., Kapur, R., Majumdar, M., Xiao, X., McAndrews-Hill, M.,
and Pragnell, I. B. (1996). Ex vivo expansion with stem cell Schindler, K., and Williams, D. A. (1998). Signaling through
factor and interleukin-11 augments both short-term recovery the interaction of membrane-restricted stem cell factor and
894 Jonathan Roy Keller and Diana Marie Linnekin

SCFR receptor tyrosine kinase: Genetic evidence for a differen- and Zsebo, K. M. (1993). Soluble stem cell factor in human
tial role in erythropoiesis. Blood 91, 879. serum. Blood 81, 656.
Keller, J. R., Jacobsen, Se. E., Dubois, C. M., Hestdal, K., and Langley, K. E., Mendiaz, E. A., Liu, N., Narhi, L. O., Zenis, L.,
Ruscetti, F. W. (1992). Transforming growth factor-beta: A Parseghian, C. M., Clogston, C. L., Leslie, I., Pope, J. A., and
bidirectional regulator of hematopoietic cell growth. Int. J. Lu, H. S. (1994). Properties of variant forms of human stem cell
Cell Cloning 10, 2. factor recombinantly expressed in Escherchia coli. Arch.
Keller, J. R., Ortiz, M., and Ruscett, I. F. W. (1995). Steel factor Biochem. Biophys. 311, 55.
(SCFR ligand) promotes the survival of hematopoietic stem/ Leary, A. G., Zeng, H. Q., Clark, S. C., and Ogawas, M. (1992).
progenitor cells in the absence of cell division. Blood 86, 1757. Growth factor requirements for survival in G0 and entry into
Keshet, E., Lyman, S. D., Williams, D. E., Anderson, D. M., the cell cycle of primitive human hemopoietic progenitors. Proc.
Jenkins, N. A., Copeland, N. G., and Parada, L. F. (1991). Natl Acad. Sci. USA 89, 4013.
Embryonic RNA expression patterns of the SCFR receptor Lev, S., Yarden, Y., and Givol, D. (1992). A recombinant ecto-
and its cognate ligand suggest multiple functional roles in domain of the receptor for the stem cell factor (SCF) retains
mouse development. EMBO J. 10, 2425. ligand-induced receptor dimerization and antagonizes SCF-
Kirshenbaum, A. S., Goff, J. P., Kessler, S. W., Mican, J. M., stimulated cellular responses. J. Biol. Chem. 267, 10866.
Zsebo, K. M., and Metcalfe, D. D. (1992). Effect of IL-3 and Levesque, J. P., Haylock, D. N., and Simmons, P. J. (1996).
stem cell factor on the appearance of human basophils and mast Cytokine regulation of proliferation and cell adhesion are cor-
cells from CD34‡ pluripotent progenitor cells. J. Immunol. 148, related events in human CD34‡ hematopoietic progenitors.
772. Blood 88, 1168.
Kinashi, T., and Springer, T. A. (1994). Steel factor and SCFR Li, C. L., and Johnson, G. R. (1994). Stem cell factor enhances the
regulate cell-matrix adhesion. Blood 83, 1033. survival but not the self-renewal of murine hematopoietic long-
Klimpel, G. R., Chopra, A. K., Langely, K. E., Wypych, J., term repopulating cells. Blood 84, 408.
Annable, C. A., Kaiserlian, D., Ernst, P. B., and Peterson, Limmani, A., Baker, W. H., Change, C. M., Seemann, R.,
J. W. (1995). A role for stem cell factor and SCFR in the murine Williams, D. E., and Patchen, M. L. (1995). SCFR ligand
intestinal tract secretory response to cholera toxin. J. Exp. Med. gene expression in normal and sublethally irradiated mice.
182, 1931. Blood 85, 2377.
Kodama, H., Nose, M., Yamaguchi, Y., Tsunoda, J., Suda, T., Linenberger, M. L., Jacobson, F. W., Bennett, L. G., Broudy, V. C.,
and Nishikawa, S. (1992). In vitro proliferation of primitive Martin, F. H., and Abkowitz, J. L. (1995). Stem cell factor
hemopoietic stem cells supported by stromal cells: Evidence production by human marrow stromal fibroblasts. Exp.
for the presence of a mechanism(s) other than that involving Hematol. 23, 1104.
SCFR receptor and its ligand. J. Exp. Med. 176, 351. Long, M. W., Briddell, R., Walter, A. W., Bruno, E., and
Kodama, H., Nose, M., Niida, S., Nishikawa, S., and Hoffman, R. (1992). Human hematopoietic stem cell adherence
Nishikawa, S. (1994). Involvement of the SCFR receptor in to cytokines and matrix molecules. J. Clin. Invest. 90, 251.
the adhesion of hematopoietic stem cells to stromal cells. Exp. Longley, B.J. Jr,, Morganroth, G. S., Tyrrell, L., Ding, T. G.,
Hematol. 22, 979. Anderson, D. M., Williams, D. E., and Halaban, R. (1993).
Koenig, A., Yakisan, E., Reuter, M., Huang, M., Sykora, K. W., Altered metabolism of mast-cell growth factor (SCFR ligand)
Corbacioglu, S., and Welte, K. (1994). Differential regulation of in cutaneous mastocytosis. N. Engl. J. Med. 328, 1302.
stem cell factor mRNA expression in human endothelial cells by Longley, B. J., Duffy, T. P., and Kohn, S. (1995). The mast cell
bacterial pathogens: an in vitro model of inflammation. Blood and mast cell disease. J. Am. Acad. Dermatol. 32, 545.
83, 2836. Lowry, P. A., Zsebo, K. M., Deacon, D. H., Eichman, C. E., and
Kovach, N. L., Lin, N., Yednock, M. T., Harlan, J. M., and Quesenberry, P. J. (1991). Effects of rrSCF on multiple cytokine
Broudy, V. C. (1995). Stem cell factor modulates avidity of 41 responsive HPP-CFC generated from SCA‡Lin murine hema-
and 51 integrins expressed on hematopoietic cell lines. Blood 85, topoietic progenitors. Exp. Hematol. 19, 994.
159. Lu, H. S., Clogston, C. L., Wypych, J., Parker, V. P., Lee, T. D.,
Ku, H., Yonemura, Y., Kaushansky, K., and Ogawa, M. (1996). Swiderek, K., Baltera, R.F. Jr., Patel, A. C., Chang, D. C., and
Thrombopoietin, the ligand for the Mpl receptor, synergizes Brankow, D. W. (1992). Post-translational processing of mem-
with steel factor and other early acting cytokines in supporting brane-associated recombinant human stem cell factor expressed
proliferation of primitive hematopoietic progenitors of mice. in Chinese hamster ovary cells. Arch. Biochem. Biophys. 298,
Blood 87, 4544. 150.
Kumar, M., and Alter, B. P. (1998). Hematopoietic growth factors McCulloch, E. A., Siminovitch, L., and Till, J. E. (1964). Spleen-
for the treatment of aplastic anemia. Curr. Opin. Hematol. 5, 226. colony formation in anemic mice of genotype WWV. Science
Kunisada, T., Yoshida, H., Yamazaki, H., Miyamoto, A., 144, 844.
Hemmi, H., Nishimura, E., Shultz, L. D., Nishikawa, S., and McCulloch, E. A., Siminovitch, L., Till, J. E., Russell, E. S., and
Hayashi, S. (1998). Transgene expression of steel factor in the Bernstein, S. E. (1965). The cellular basis of the genetically
basal layer of epidermis promotes survival, proliferation, differ- determined hemopoietic defects in anemic mice of genotype
entiation and migration of melanocyte precursors. Development Sl/Sld. Blood 26, 399.
125, 2915. McNiece, I. K., and Briddell, R. A. (1995). Stem cell factor.
Langley, K. E., Wypych, J., Mendiaz, E. A., Clogston, C. L., J. Leukoc. Biol. 58, 14.
Parker, V. P., Farrar, D. H., Brothers, M. O., Satygal, V. N., McNiece, I. K., Langley, K. E., and Zsebo, K. M. (1991a).
Leslie, I., and Birkett, N. C. (1992). Purification and character- Recombinant human stem cell factor synergizes with GM-
ization of soluble forms of human and rat stem cell factor CSF, G-CSF, IL-3 and Epo to stimulate human progenitor
recombinantly expressed by Escherichia coli and by Chinese cells of the myeloid and the erythroid lineages. Blood 19, 226.
hamster ovary cells. Arch. Biochem. Biophys. 295, 21. McNiece, I. K., Langley, K. E., and Zsebo, K. M. (1991b). The
Langley, K. E., Bennett, L. G., Wypych, J., Yancik, S. A., role of recombinant stem cell factor in early B cell development.
Liu, X. D., Westcott, K. R., Change, D. G., Smith, K. A., Synergistic interaction with IL-7. J. Immunol. 146, 3785.
Stem Cell Factor 895

McNiece, I. K., Bertoncello, I., Keller, J. R., Ruscetti, F. W., Jelaca-Maxwell, K., Nichols, C. R., Brown, S. L., Nimer, S. D.,
Hartley, C. A., and Zsebo, K. M. (1992). Transforming growth and Gabrilove, J. (1997). Recombinant methionyl human stem
factor beta inhibits the action of stem cell factor on mouse and cell factor and filgrastim for peripheral blood progenitor cell
human hematopoietic progenitors. Int. J. Cell Cloning 10, 80. mobilization and transplantation in non-Hodgkin's lymphoma
Majumdar, M. K., Feng, L., Medlock, E., Toksoz, D., and patients. Results of a phase I/II trial. Blood 89, 3136.
Williams, D. A. (1994). Identification and mutation of primary Motro, B., van der Kooy, D., Rossant, J., Reith, A., and
and secondary proteolytic cleavage sites in murine stem cell Bernstein, A. (1991). Contiguous patterns of SCFR and steel
factor cDNA yields biologically active, cell-associated protein. expression: analysis of mutations at the W and Sl loci.
J. Biol. Chem. 269, 1237. Development 113, 1207.
Martin, F. H., Suggs, S. V., Langley, K. E., Lu, H. S., Ting, J., Motro, B., Wojtowicz, J. M., Bernstein, A., and van der Kooy, D.
Okino, K. H., Morris, C. F., McNiece, I. K., Jacobsen, F. W., (1996). Steel mutant mice are deficient in hipocampal learning
and Mediaz, E. A. (1990). Primary structure and functional but not long-term potentiation. Proc. Natl Acad. Sci. USA 93,
expression of rat and humanstem cell factor DNAs. Cell 63, 1808.
203. Mrozek, E., Anderson, P., and Caligiuri, M. A. (1996). Role of
Matos, M. E., Schnier, G. S., Beecher, M. S., Ashman, L. K., interleukin-15 in the development of human CD56‡ natural
William, D. E., and Caligiuiri, M. A. (1993). Expression of a killer cells from CD34‡ hematopoietic progenitor cells. Blood
functional SCFR receptor on a subset of natural killer cells. 87, 2632.
J. Exp. Med. 178, 1079. Neta, R., Willims, D., Selzer, F., and Abrams, J. (1993). Inhibition
Matous, J. V., Langley, K., and Kaushansky, K. (1996). Structure- of SCFR ligand/steel factor by antibodies reduces survival of
function relationships of stem cell factor: An analysis based on a lethally irradiated mice. Blood 81, 324.
series of human-murine stem cell factor chimera and the map- Nilsson, G., Butterfield, J. H., Nilsson, K., and Siegbahn, A.
ping of a neutralizing monoclonal antibody. Blood 88, 437. (1994). Stem cell factor is a chemotactic for human mast cells.
Matsui, Y., Zsebo, K. M., and Hogan, B. L. M. (1990). J. Immunol. 153, 3717.
Embryonic expression of a haematopoietic growth factor Nishikawa, S., Kusakabe, M., Yoshinaga, K., Ogawa, M.,
encoded by the Sl locus and the ligand for SCFR. Nature 347, Hayashi, S., Kunisada, T., Era, T., Sakakura, T., and
667. Nisshikawa, S. (1991). In utero manipulation of coat color for-
Meininger, C. J., Yano, H., Rottapel, R., Bernstein, A., mation by a monoclonal anti-SCFR antibody: two distinct
Zsebo, K. M., and Zetter, B. R. (1992). The SCFR receptor waves of SCFR-dependency during melanocyte development.
ligand functions as a mast cell chemoattractant. Blood 79, 958. Nishikawa, M., Tojo, A., Ikebuchi, K., Katayama, K., Fujii, N.,
Metcalf, D., and Nicola, N. A. (1991). Direct proliferative actions Ozawa, K., and Asano, S. (1992). Deletion mutagenesis of stem
of stem cell factor on murine bone marrow cells in vitro: Effects cell factor defines the C-terminal sequences essential for its bio-
of combination with colony-stimulating factors. Proc. Natl logical activity. Biochem. Biophys. Res. Commun. 188, 292.
Acad. Sci. USA 88, 6239. Ogawa, M. (1993). Differentiation and proliferation of hemato-
Metcalf, D. (1993). Hematopoietic regulators: Redundancy or poietic stem cells. Blood 81, 2844.
subtlety? Blood 82, 3515. Ogawa, M., Matsuzaki, Y., Nishikawa, S., Hayashi, S.,
Migliaccio, G., Migliaccio, A. R., Druzin, M. L., Giardina, P. J., Kunisada, T., Sudo, T., Kina, T., Nakauchi, H., and
Zsebo, K. M., and Adamson, J. W. (1992). Long-term genera- Nishikawa, S. (1991). Expression and function of SCFR in
tion of colony-forming cells in liquid culture of CD34‡ cord hemopoietic progenitor cells. J. Exp. Med. 174, 63.
blood cells in the presence of recombinant human stem cell Okumura, N., Tsuji, K., Ebihara, Y., Tanaka, I., Sawai, N.,
factor. Blood 79, 2620. Koike, K., Komiyama, A., and Nakhata, T. (1996).
Migliaccio, G., Migliaccio, A. R., Valinksy, J., Langley, K., Chemotactic and chemokinetic activities of stem cell factor on
Zsebo, K., Visser, J. W., and Adamson, J. W. (1991a). Stem murine hematopoietic progenitor cells. Blood 87, 4100.
cell factor induces proliferation and differentiation of highly Orazi, A., Gordon, M. S., John, K., Sledge, G. Jr., Neiman, R. S.,
enriched murine hemopoietic cells. Proc. Natl Acad. Sci. USA and Hoffman, R. (1995). In vivo effects of recombinant human
88, 7420. stem cell factor treatment. A morphological and immunohisto-
Migliaccio, G., Migliaccio, A. R., Druzin, M. L., Giardina, P. J., chemical study of bone marrow biopsies. Am. J. Clin. Pathol.
Zsebo, K. M., and Adamson, J. W. (1991b). Effects of recom- 103, 177.
binant human stem cell factor (SCF) on the growth of human Ortiz, M., Wine, J. W., Lohrey, N., Ruscetti, F. W., Spence, S. E.,
progenitor cells in vitro. J. Cell. Physiol. 148, 503. and Keller, J. R. (1999). Functional characterization of a novel
Miller, C. L., Rebel, V. I., Helgason, C. D., Lansdorp, P. M., and hematopoietic stem cell and its place in the SCFR maturation
Eaves, C. J. (1997). Impaired steel factor responsiveness differ- pathway in bone marrow cell development. Immunity 10, 173.
entially affects the detection and long-term maintenance of fetal Pandit, J., Bohm, A., Janacarik, J., Halenbeck, R., and Koths, K.
liver hematopoietic stem cells in vivo. Blood 89, 1214. (1992). Three-dimensional structure of dimeric human
Molineux, G., Migdalska, A., Szmitkowski, M., Zsebo, K., and recombinant macrophage colony-stimulating factor. Science
Dexter, T. M. (1991). The effects on hematopoiesis of recombi- 258, 1358.
nant stem cell factor (ligand for SCFR) administered in vivo to Peters, S. O., Kittler, E. L., Ramshaw, H. S., and Quesenberry, P. J.
mice either alone or in combination with granulocyte colony- (1996). Ex vivo expansion of murine marrow cells with interleu-
stimulating factor. Blood 78, 961. kin-3 (IL-3), IL-6, IL-11, and stem cell factor leads to impaired
Morrissey, P. J., McKenna, H., Widmer, M. B., Braddy, S., engraftment in irradiated hosts. Blood 87, 30.
Voice, R., Charrier, K., Williams, D. E., and Watson, J. D. Pettite, J. N., and Kulik, M. J. (1996). Cloning and characteriza-
(1994). Steel factor (SCFR ligand) stimulates the in vitro growth tion of cDNAs encoding two forms of avian stem cell factor.
of immature CD3/CD4/CD8 thymocytes: synergy with IL-7. Biochim. Biophys. Acta 1307, 149.
Cell. Immunol. 157, 118. Qui, F., Ray, P., Brown, K., Barker, P. E., Jhanwar, S.,
Moskowitz, C. H., Stiff, P., Gordon, M. S., McNiece, I., Ho, A. D., Ruddle, F. H., and Besmner, P. (1988). Primary structure of
Costa, J. J., Broun, E. R., Bayer, R. A., Wyres, M., Hill, J., SCFR: relationslhip with the CSF-1/PDGF receptor kinase
896 Jonathan Roy Keller and Diana Marie Linnekin

family-oncogenic activation of v-kit involves deletion of extra- Szabolcs, P., Moore, M. A. S., and Young, J. W. (1995).
cellular domain and C-terminus. EMBO J. 7, 1003. Expansion of immunostimulatory dendritic cells among the
Ramsfjell, V., Borge, O. J., Veiby, O. P., Cardier, J., Murphy, M.J. myeloid progeny of human CD34‡ bone marrow precursors
Jr., Lyman, S. D., Lok, S., and Jacobsen, S. E. (1996). cultured with SCFR ligand, granulocyte-macrophage colony-
Thrombopoietin, but not erythropoietin, directly stimulates stimulating factor, and TNF . J. Immunol. 154, 5851.
multilineage growth of primitive murine bone marrow Takeda, S., Shimizu, T., and Rodewald, H. R. (1997). Interactions
progenitor cells in synergy with early acting cytokines: distinct between SCFR and stem cell factor are not required for B-cell
interactions with the ligands for SCFR and FLT3. Blood 88, development in vivo. Blood 89, 518.
4481. Tajima, Y., Onoue, H., Kitamura, Y., and Nishimune, Y. (1991).
Ramsfjell, V., Borge, O. J., Cui, L., and Jacobsen, S. E. (1997). Biologically active kit ligand growth factor is produced by
Thrombopoietin directly and potently stimulates multilineage mouse Sertoli cells and is defective in Sld mutant mice.
growth and progenitor cell expansion from primitive Development 113, 1031.
(CD34‡CD38) human bone marrow progenitor cells: Distinct Tajima, Y., Huang, E. J., Vosseller, K., Ono, M., Moore, M. A.,
and key interactions with the ligands for SCFR and flt3, and and Besmer, P. (1998a). Role of dimerization of the membrane
inhibitory effects of TGF- and TNF. J. Immunol. 158, 5169. associated growth factor kit ligand in juxtacrine signaling: The
Ratajczak, M. Z., Kuczynski, W. I., Sokol, D. L., Moore, J. S., Sl17H mutation affects dimerization and stability-phenotypes in
Pletcher, C.H. Jr., and Gewirtz, A. M. (1995). Expression and hematopoiesis. J. Exp. Med. 187, 1451.
physiologic significance of Kit ligand and stem cell tyrosine Tajima, Y., Moore, M. A., Soares, V., Ono, M., Kissel, H., and
kinase-1 receptor ligand in normal human CD34‡, SCFR‡ Besmer, P. (1998b). Consequences of exclusive expression in vivo
marrow cells. Blood 86, 2161. of kit-ligand lacing the major proteolytic cleavage site. Proc.
Rennick, D., Hunte, B., Holland, G., and Thompson-Snipes, L. Natl Acad. Sci. USA 95, 11903.
(1995). Cofactors are essential for stem cell factor-dependent Taylor, W. E., Najmabadi, H., Strathearn, M., Jou, N. T.,
growth and maturation of mast cell progenitors: Comparative Liebling, M., Rajavashisth, T., Chanani, N., Phung, L., and
effects of interleukin-3 (IL-3), IL-4, IL-10, and fibroblasts. Bhasin, S. (1996). Human stem cell factor promoter
Blood 85, 57. deoxyribonucleic acid sequence and regulation by cyclin
Rico-Vargas, S. A., Weiskopf, B., Nishikawa, S., and 30 ,50 -Adenosine monophosphate in a sertoli cell line.
Osmond, D. G. (1994). SCFR expression by B cell precursors Endocrinology 137, 5407.
in mouse bone marrow. Stimulation of B cell genesis by in vivo Testa, U., Martucci, R., Rutella, S., Scambia, G., Sica, S.,
treatment with anti-SCFR antibody. J. Immunol. 152, 2845. Benedetti Panici, P., Pierelli, L., Menichella, G., Leone, G.,
Rodewald, H.-R., Kretzschmar, K., Swat, W., and Takeda, S. and Mancuso, S. (1994). Autologous stem cell transplantation:
(1995). Intrathymically expressed SCFR ligand (stem cell Release of early and late acting growth factors relates with
factor) is a major factor driving expansion of very immature hematopoietic ablation and recovery. Blood 84, 3532.
thymocytes in vivo. Immunity 3, 313. Toksoz, D., Zsebo, K. M., Smith, K. A., Hu, S., Brankow, D.,
Rolink, A., Streb, M., Nishikawa, S., and Melchers, F. (1991). The Suggs, S. V., Martin, F. H., and Williams, D. A. (1992).
SCFR-encoded tyrosine kinase regulates the proliferation of Support of human hematopoiesis in long-term bone marrow
early pre-B cells. Eur. J. Immunol. 21, 2609. cultures by murine stromal cells selectively expressing the mem-
Russell, E. S., Bernstein, S. E., Lawson, F. A., and Smith, L. G. brane-bound and secreted forms of the human homolog of the
(1959). Long-continued function of normal blood-forming steel gene product, stem cell factor. Proc. Natl Acad. Sci. USA
tissue transplanted into genetically anemic hosts. J. Natl 89, 7350.
Cancer Inst. 23, 557. Tong, J., Gordon, M. S., Srour, E. F., Cooper, R. J., Orazi, A.,
Russell, E. S. (1979). Hereditary anemias of the mouse: A review McNiece, I., and Hoffman, R. (1993). In vivo administration of
for geneticists. Adv. Genet. 20, 357. recombinant methionyl human stem cell factor expands the num-
Shaw, G., and Kamen, R. (1986). A conserved AU sequence from ber of human marrow hematopoietic stem cells. Blood 82, 784.
the 30 untranslated region of GM-CSF mRNA mediates selec- Tsai, M., Takeishi, T., Thompson, H., Langley, K. E.,
tive mRNA degradation. Cell 46, 659. Zsebo, K. M., Metcalfe, D. D., Geissler, E. N., and Galli, S. J.
Shibuya, A., Nagayoshi, K., Nakamura, K., and Nakauchi, H. (1991a). Induction of mast cell proliferation, maturation, and
(1995). Lymphokine requirement for the generation of natural heparin synthesis by the rat SCFR ligand, stem cell factor. Proc.
killer cells from CD34‡ hematopoietic progenitor cells. Blood Natl Acad. Sci. USA 88, 6382.
85, 3538. Tsai, M., Shih, L. S., Newlands, G. F., Takeishi, T.,
Shull, R. M., Suggs, S. V., Langley, K. E., Okino, K. H., Langley, K. E., Zsebo, K. M., Miller, H. R., Geissler, E. N.,
Jacobsen, F. W., and Martin, F. H. (1992). Canine stem cell and Galli, S. J. (1991b). The rat SCFR ligand, stem cell
factor (SCFR ligand) supports the survival of hematopoietic factor, induces the development of connective tissue-type and
progenitors in long-term canine marrow culture. Exp. mucosal mast cells in vivo. Analysis by anatomical distribution,
Hematol. 20, 1118. histochemistry, and protease phenotype. J. Exp. Med. 174, 125.
Silva, M. R., Hoffman, R., Srour, E. F., and Ascensao, J. L. Tsai, S., Bartelmez, S., Sitnicka, E., and Collins, S. (1994).
(1994). Generation of human natural killer cells from immature Lymphohematopoietic progenitors immortalized by a retroviral
progenitors does not require marrow stromal cells. Blood 84, vector harboring a dominant-negative retinoic acid receptor can
841. recapitulate lymphoid, myeloid, and erythroid development.
Silvers, W. K. (1979). In ``Dominant Spotting, Patch, and Rump- Genes Dev. 8, 2831.
White'' (ed. W. K. Silvers), The coat colors of mice: a model for Tsuji, K., Zsebo, K. M., and Ogawa, M. (1991). Enhancement of
mammalian gene action and interaction pp. p. 206. Springer- murine blast cell colony formation in culture by recombinant
Verlag, New York. rat stem cell factor, ligand for SCFR. Blood 78, 1223.
Spangrude, G. J., Smith, L., Uchida, H., Ikuta, K., Heimfeld, S., Ulich, T. R., del Castillo, J., Yi, E. S., Yin, S., McNiece, I.,
Friedman, J., and Weissman, I. L. (1991). Mouse hematopoietic Yung, Y. P., and Zsebo, K. M. (1991). Hematological effects
stem cells. Blood 78, 1395. of stem cell factor in vivo and in vitro in rodents. Blood 78, 645.
Stem Cell Factor 897

Ullrich, A., and Schlessinger, J. (1990). Signal transduction by Zhou, J.-H., Ohtaki, M., and Sakurai, M. (1993). Sequence of a
receptors with tyrosine kinase activity. Cell 61, 203. cDNA encoding chicken stem cell factor. Gene 127, 269.
Van Os, R., Dawes, D., Mislow, J. M., Witsell, A., and Zsebo, K. M., Wypych, J., McNiece, I. K., Lu. H. S., Smith, K. A.,
Mauch, P. M. (1997). Host conditioning with 5-fluorouracil Karkare, S. B., Sachdev, R. K., Yuschenkoff, V. N.,
and kit-ligand to provide for long term bone marrow engraft- Birkett, N. C., and Williams, L. R. (1990a). Identification, pur-
ment. Blood 89, 2376. ification, and biological characterization of hematopoietic
Weaver, A., Ryder, D., Crowther, D., Dexter, T. M., and stem cell factor from buffalo rat liver-conditioned medium.
Testa, N. G. (1996). Increased numbers of long-term culture- Cell 63, 195.
initiating cells in the apheresis product of patients randomized Zsebo, K. M., Williams, D. A., Geissler, E. N., Broudy, V. C.,
to receive increasing doses of stem cell factor administered in Martin, F. H., Atkins, H. L., Hsu, R. Y., Birkett, N. C.,
combination with chemotherapy and a standard dose of gran- Okino, K. H., and Murdock, D. C. (1990b). Stem cell factor
ulocyte colony-stimulating factor. Blood 88, 3323. is encoded at the Sl locus of the mouse and is the ligand for the
Williams, D. E., Eisenman, J., Baird, A., Rauch, C., Van Ness, K., SCFR tyrosine kinase receptor. Cell 63, 213.
March, C. J., Park, L. S., Martin, U., Mochizuki, D. Y., and Zsebo, K. M., Smith, K. A., Hartley, C. A., Greenblatt, M.,
Boswel, H. S. (1990). Identification of a ligand for the SCFR Cooke, K., Rich, W., and McNiece, I. K. (1992). Radio-
proto-oncogene. Cell 63, 167. protection of mice by recombinant rat stem cell factor. Proc.
Williams, N., Bertoncello, I., Kavnoudias, H., Zsebo, K., and Natl Acad. Sci. USA 89, 9464.
McNiece, I. (1992). Recombinant rat stem cell factor stimulates
the amplification and differentiation of fractionated mouse stem
cell populations. Blood 79, 58.
Wineman, J. P., Nishikawa, S., and Muller-Sieburg, C. E. (1993). ACKNOWLEDGEMENTS
Maintenance of high levels of pluripotent hematopoietic stem
cells in vitro: Effect of stromal cells and SCFR. Blood 81, 365. The content of this publication does not necessarily
Yan, X. Q., Hatley, C., McElroy, P., Chang, A., McCrea, C., and
reflect the views or policies of the Department of
McNiece, I. (1995). Peripheral blood progenitor cells mobilized
by recombinant human granulocyte colony-stimulating factor Health and Human Services, nor does mention of
plus recombinant rat stem cell factor contain long-term engraft- trade names, commercial products, or organizations
ing cells capable of cellular proliferation for more than two imply endorsement by the U.S. Government.
years as shown by serial transplantation in mice. Blood 85, 2303. We are grateful for the critical review of this
Yarden, Y., Kuang, W. J., Yang-Feng, T., Coussens, L.,
manuscript by Drs Sally Spence, Jon Dermott, and
Munemitsu, S., Dull, T. J., Chen, E., Schlessinger, J.,
Francke, U., and Ulrich, A. (1987). Human proto-oncogene J.J. Oppenheim. The publisher or recipient acknowl-
SCFR: A new cell surface receptor tyrosine kinase for an un- edges right of the U.S. Government to retain a
identified ligand. EMBO J. 6, 3341. nonexclusive, royalty-free license in and to any copy-
Young, J. W., Szabolcs, P., and Moore, M. A. (1995). right covering the article. This project has been
Identification of dendritic cell colony-forming units among nor-
funded in whole or in part with Federal Funds from
mal human CD34‡ bone marrow progenitors that are
expanded by SCFR-ligand and yield pure dendritic cell colonies the national Cancer Institute, National Institutes of
in the presence of granulocyte/macrophage colony-stimulating Health, under contract number NO1-CO-56000.
factor and tumor necrosis factor. J. Exp. Med. 182, 1111.
Zhang, Z., and Anthony, R. V. (1994). Porcine stem cell factor/
SCFR ligand: its molecular cloning and localization with the
uterus. Biol. Reprod. 50, 95.

You might also like