HEMATOPOIESIS IS A life-long process responsible for replenishing both hematopoietic progenitor cells and mature blood cells from a pool of pluripotent, long-term reconstituting stem cells.1 The daily turnover in a normal adult of approximately 1012 blood cells is tightly regulated, involving, in part, a complex interaction between soluble and membrane-bound stimulatory and inhibitory cytokines and their corresponding receptors.2-4 The molecular cloning of these hematopoietic growth factors (HGFs) and their receptors has been instrumental in delineating the pathways that lead from a single hematopoietic stem cell to the various terminally differentiated cells in the hematopoietic system.

Although a number of cytokines have effects on progenitor and stem cells in vitro or in vivo, two cytokines discovered in the early 1990s, c-kit ligand and flt3 ligand, appear to have unique and nonredundant activities on primitive progenitor/stem cells.

Because of the broad range of hematopoietic activities mediated through interaction of c-kit ligand (KL) and flt3 ligand (FL) with their receptors, it is beyond the scope of this report to review the effects of these proteins outside of the hematopoietic system. Rather, we will focus on the discovery, structure, function, expression, and biological roles of these two ligand-receptor pairs. Special attention will be directed towards hematopoietic activities in which KL and FL show either distinct or synergistic effects. For a more detailed overview of other hematologic and immunologic effects of KL and FL, other reviews can be recommended.5-8 Two subjects have been deliberately left out of this report, because they are deserving of their own separate reviews (signal transduction pathways involving c-kit and flt3 and activities of KL and FL outside of the hematopoietic system).

The W (dominant White spotting) locus in mice was first described in the early 1900s.9,10 Mice afflicted with mutations at the W locus were originally identified, as the name implies, by the presence of a white spot on the bellies of pigmented mice. Detailed examination of these mice showed that the mutation was pleiotropic. The mice suffer from defects in germ cell development (manifested as reproductive difficulties) and in hematopoiesis (characterized by a macrocytic anemia). Over the years, at least 20 allelic variants of the W locus have been described; most have a similar, although not identical, phenotype.9,10 The W locus is on chromosome 5 and is one of the most mutable loci in mice.9,10 

A central question that remained was what kind of protein the Wlocus encoded, and how did it affect so many different tissues. A breakthrough came in 1988 when it was shown that the W locus encoded a tyrosine kinase receptor known as c-kit.11,12 The c-kit protein has the same general structure as four other tyrosine kinase receptors: c-fms, the receptor for macrophage colony-stimulating factor (M-CSF)13-15; flt316-19; and both of the receptors for platelet-derived growth factor (PDGF; designated as A and B).20-23 Each of these receptors is approximately 1,000 amino acids in length, has five Ig-like domains in the extracellular region, and contains a split catalytic domain in the cytoplasmic region that phosphorylates tyrosine residues in specific target proteins after activation of the receptor by ligand. The exact defect in the c-kit receptor has been identified at the molecular level for a number of alleles of theW locus24-28 (see section on genetic alterations in c-kit and KL genes).

Many years after the discovery of the W locus, a mutation in mice that had a phenotype virtually identical to W mice was identified.29 Despite the similarities in phenotype, this new mutation, designated Steel (Sl), was localized to mouse chromosome 10, so it was clearly not allelic with the W locus on chromosome 5.10,30 Because mutations on two different chromosomes had the same complex phenotype that affects pigmentation, germ cells, and hematopoiesis, researchers hypothesized that there would be some relationship between the proteins encoded at these two loci. Elizabeth Russell, who did much of the pioneering research on both of these mutations, suggested (years before the discovery that theW locus encoded c-kit and that c-kit was a receptor) that the W and Sl loci might encode a receptor and its cognate ligand.10 

With the recognition that the W locus encoded c-kit,11,12 the search for the c-kit ligand began in earnest. A number of approaches were undertaken to identify the protein encoded at the Sl locus, including chromosome walking31 and expression cloning. However, the successful approach turned out to be the purification of the Steel factor protein.

The cloning of a cDNA encoding the Steel factor was reported simultaneously by three different groups, each of which discovered a different source of the factor.32-34 All three groups used a similar approach; they first purified the protein from medium conditioned by a cell line, obtained N-terminal amino acid sequence, and then made degenerate oligonucleotide primers based on the protein sequence to isolate cDNA clones by polymerase chain reaction (PCR). The three groups named this protein mast cell growth factor, stem cell factor, and c-kit ligand (see below). In this review, we will use the name c-kit ligand (KL) for the protein that binds to the c-kit receptor and is encoded at the Sl locus on mouse chromosome 10 (see below).32,35,36 

Once the murine and rat KL cDNAs had been cloned, cross-species hybridization was used to clone KL cDNAs from a number of other species.33,37-40 The mouse and human proteins are 82% identical at the amino acid level.

In contrast to the discovery of c-kit, analysis of mouse mutations did not play a role in the discovery of the flt3 receptor. This receptor was isolated independently by two groups using distinct cloning strategies.18,19,41 One group used low stringency hybridization with a DNA probe from the M-CSF receptor (c-fms) to isolate a portion of a related DNA sequence that was named flt3 (fms-like tyrosine kinase 3).41 The partial clone was then used to isolate a full-length receptor clone.18 

A second group used degenerate oligonucleotides (based on conserved regions within the kinase domain of tyrosine kinase receptors) in a PCR-based strategy to isolate a novel receptor fragment from highly purified murine fetal liver stem cells.19 This fragment was used to isolate a full-length receptor clone given the name flk-2 (fetal liver kinase 2). The flt3/flk-2 receptor has also been referred to as Stk-1 (stem cell kinase-1),17 but this name is not widely used, perhaps because it has been previously designated to denote a gene regulating stem cell kinetics42 as well as a different receptor tyrosine kinase of the met/sea/ron family.43 

Comparison of the murine flt3 and flk-2 receptor sequences showed that these sequences differ by only two amino acids in their extracellular domains.44 In contrast, a large number of amino acid differences were seen in a region near their C-terminal ends. The murine flt3 receptor sequence has been independently confirmed by several groups,44-46 and the human receptor sequence is directly homologous to the murine flt3, but not the murine flk-2 sequence.16,17 No independent confirmation of the sequence of flk-2 has been reported. Differences between flt3 and flk-2 sequences are not a result of tissue-specific expression of distinct isoforms.46 The differences in the murine flt3 and flk-2 sequences have never been fully explained, and the validity of the sequence reported as flk-2 is still unclear.47 As a result of this, we refer to the receptor as flt3 and to its ligand as flt3 ligand (FL).

A soluble form of the flt3 receptor was the key reagent used by two groups to clone FL. Lyman et al48 screened a variety of cell lines to look for one that expressed a ligand on the cell surface that was capable of binding the soluble receptor. A murine T-cell line was identified that specifically bound the soluble flt3 receptor. The ligand was then cloned from a cDNA expression library made from mRNA isolated from these cells.

An alternative approach employed by Hannum et al49 used an affinity column made with the mouse flt3 receptor extracellular domain to purify FL from medium conditioned by a murine thymic stromal cell line. N-terminal sequencing of the purified protein generated a short amino acid sequence, which was then used to design degenerate oligonucleotide primers to amplify a portion of the FL gene by PCR. Isolation of this FL gene fragment led to the cloning of a full-length murine cDNA.

Once the murine FL cDNA had been isolated, it was used to isolate cDNAs encoding the human gene.49,50 The mouse and human FL proteins are 72% identical at the amino acid level; homology is greater in the extracellular region (73%) than in the cytoplasmic domain (57%).

No restriction in species specificity has been observed with regard to FL binding or biological activity. Both the mouse and human ligand proteins are fully active on cells bearing either the mouse or human receptors.51 The human FL protein has been found to stimulate mouse, cat (Janis Abkowitz, University of Washington, Seattle, WA, unpublished data), rabbit, nonhuman primate, and human cells. This lack of species specificity of FL is in marked contrast to KL, where the mouse protein is active on human cells but the human protein has limited activity on murine cells.33 Analysis of chimeric mouse/human KL proteins has helped define regions of the protein that regulate its species-specific action.52 

The murine and human c-kit receptors are each 976 amino acids in length, have nine potential sites for N-linked glycosylation in their extracellular domains,53,54 and are glycosylated at one or more of these sites.54,55 Immunoprecipitation shows two proteins of approximately 140 kD and 155 kD54; the predicted size of the protein backbone alone is approximately 108 kD. Pulse-chase analysis has shown that the larger 155-kD protein arises from the smaller protein,56 presumably due to glycosylational processing of the protein from one containing high mannose carbohydrates to one containing complex carbohydrates. Furthermore, cell surface iodination of c-kit-expressing cells radiolabels only the larger protein.54 The size of the c-kit protein varies between tissues,55 although whether this is due to differential glycosylation or expression of different isoforms is unclear (see below).

The murine (1,000 amino acids) and human (993 amino acids) flt3 receptors have 9 and 10 potential sites for N-linked glycosylation, respectively, in their extracellular domains16-19 and are also glycosylated at one or more of these sites.44Immunoprecipitation shows two proteins of 130-143 kD and 155-160 kD44,57,58; the predicted size of the protein backbone alone is approximately 110 kD. As with c-kit, pulse-chase analysis has shown that the larger protein arises from the smaller protein44; again, this most likely results from glycosylational processing. Consistent with this interpretation is the finding that only the 158-kD species is found on the cell surface.44 There do not appear to be any O-linked sugars on the protein.59 

A number of studies have measured the binding affinity of KL to the c-kit receptor60-64 and that of FL to the flt3 receptor.65 Both high (kd, 16 to 310 pmol/L) and low (kd, 11 to 65 nmol/L) affinity binding of KL to its receptor have been reported.60,61,63 Some primary cells and cell lines have only high- affinity sites, whereas others have both.61,63 Neither the number of receptors per cell nor the finding of one or two classes of receptors can be correlated with the ability of cells to proliferate in response to KL.60 

The binding affinity of human FL for the flt3 receptor on human myeloid leukemia cells has been estimated to be 200 to 500 pmol/L,65 and only high-affinity binding is seen. The high binding affinity of FL for the flt3 receptor is therefore in the same range of affinities as the binding of KL to c-kit.

The c-kit and flt3 receptors each have five Ig-like domains in their extracellular regions. Mutagenesis studies on c-kit have shown that the first three domains are both necessary and sufficient for binding of ligand66 and that the fourth Ig-like domain is required for dimerization of the receptor,66 although this has recently been called into question.67 Several models have been proposed for binding of KL to c-kit,66-71 but it is beyond our scope to review these studies. Whatever the mechanism responsible for the formation of the complex, the ultimate result is that a dimeric form of the ligand is associated with a dimeric form of the receptor, which results in signal transduction. Although similar studies have not been performed with FL and flt3 receptors, a similar process most likely occurs with this ligand-receptor pair.

Analysis of independently derived cDNA clones has shown that there are two isoforms of both the murine and human c-kit-encoded protein.72 These c-kit receptor isoforms differ by four amino acids (glycine-asparagine-asparagine-lysine, abbreviated GNNK) that are either present or absent just upstream of the transmembrane domain. The different isoforms result from alternative splicing of c-kit mRNAs at a cryptic splice donor site located at the 3′ end of exon 9.73 Although it is not clear if physiologic differences occur because of ligand signaling via one c-kit isoform versus another, ligand-independent constitutive phosphorylation of the receptor occurs only in the isoform missing these four amino acids.72 

Crosier et al74 examined expression of the two c-kitisoforms in both leukemic cell lines and in primary acute myeloid leukemias; both isoforms appeared to be expressed in all of the cells examined, with the ratio of GNNK to GNNK+isoforms ranging from 10:1 to 15:1. A second study confirmed the expression of both isoforms in a series of acute myeloid leukemias.75 

In addition to the isoforms discussed above, other variants have been seen in the c-kit receptor. Alternative splicing of mRNAs has been shown to insert an extra serine residue in the cytoplasmic domain at position 715; a survey of human cell lines and acute myeloid leukemia samples shows that both of these isoforms are normally expressed.74 

Finally, soluble c-kit receptors are produced by some hematopoietic cell lines in culture,64 and a soluble version of c-kit has been found in human serum at high levels (324 ± 105 ng/mL).76 How this soluble c-kitreceptor is generated is unknown, although it does appear capable of binding KL.60,64 In each of the cases described above, the physiologic significance, if any, of the receptor variant is unknown.

Fewer isoforms of the flt3 receptor have been reported than have been seen with c-kit. One isoform of the murine flt3 receptor is missing the fifth of the five Ig-like regions in the extracellular domain as a result of the skipping of two exons during transcription.77 This alternative isoform is present at lower levels than the wild-type receptor, although it is able to bind ligand and is phosphorylated as a result of this binding. Thus, the fifth Ig domain of flt3 is not required for either ligand binding or receptor phosphorylation. Similarly, the c-kit receptor requires only the first three Ig-like domains for ligand binding.66 The physiologic significance of this flt3 receptor isoform is presently unknown, and a soluble version has not yet been identified in human serum.

The KL and FL proteins are structurally similar to each other (as described below)48-50 and to M-CSF.78 The primary translation product of the KL gene is a type 1 transmembrane protein, ie, the N-terminus of the protein is located outside of the cell. This protein is biologically active on the cell surface.79 The murine and human KL proteins are each 273 amino acids in length, with a 25 amino acid leader, a 185 amino acid extracellular domain, a 27 amino acid transmembrane domain, and a 36 amino acid cytoplasmic tail.

The murine32,79 KL protein has four potential sites for N-linked sugar addition; the human protein has five. KL made by Buffalo rat liver cells is N-glycosylated in a heterogeneous fashion and probably contains O-linked sugars. Analysis of human KL produced by Chinese hamster ovary (CHO) cells shows that it is glycosylated in a somewhat different manner than the rat protein and that it also contains O-linked sugars.80 

Circular dichroism spectra of KL shows that it has considerable secondary structure, including both α helical and β sheets.80 There are four cysteine residues that are conserved between KL, FL, and M-CSF. In the case of KL, these form two intramolecular disulfide bonds that establish the three-dimensional structure of the protein.81 Although KL forms homodimers in solution, they are not covalently linked.80 KL is thus different from M-CSF, which contains three intramolecular disulfide bonds and an unpaired cysteine residue that forms an intermolecular disulfide bond.82 Preliminary data suggest that FL also contains three intramolecular disulfide bonds and exists as a noncovalently linked homodimer (Rick Remmele, Immunex, Seattle, WA; unpublished observation).

Mutagenesis studies of mouse and human KL have identified a core region that is required for biological activity; this region constitutes the major portion of the extracellular domain and encompasses all four of the cysteine residues conserved between KL, FL, and M-CSF.83,84 Neither the cytoplasmic, transmembrane, spacer, nor tether regions of KL (Fig 1) is required for biological activity. Similar studies on FL have yielded essentially identical results.85 

Fig. 1.

Sequence alignment of human FL and KL proteins. The figure illustrates that both colony-stimulating factors are type I transmembrane proteins with short cytoplasmic domains; both are likely to be four helix bundle proteins (based on x-ray crystallography data in the case of M-CSF82). The approximate positions of the four helices are shown. The vertical red lines show the locations of introns (to the nearest amino acid) within the genes33,93,95,104 and illustrate their common genomic structure and ancestral origin. Conserved cysteine residues are shaded in color to reflect the formation of proposed intramolecular disulfide bonds (3 in the case of FL and 2 in the case of KL). Possible sites for N-linked glycosylation are boxed. The alignment is based on the one originally proposed by Bazan78 for KL and M-CSF.

Fig. 1.

Sequence alignment of human FL and KL proteins. The figure illustrates that both colony-stimulating factors are type I transmembrane proteins with short cytoplasmic domains; both are likely to be four helix bundle proteins (based on x-ray crystallography data in the case of M-CSF82). The approximate positions of the four helices are shown. The vertical red lines show the locations of introns (to the nearest amino acid) within the genes33,93,95,104 and illustrate their common genomic structure and ancestral origin. Conserved cysteine residues are shaded in color to reflect the formation of proposed intramolecular disulfide bonds (3 in the case of FL and 2 in the case of KL). Possible sites for N-linked glycosylation are boxed. The alignment is based on the one originally proposed by Bazan78 for KL and M-CSF.

Close modal

The primary translation product of the FL gene is also a type 1 transmembrane protein. The mouse and human proteins contain 231 and 235 amino acids, respectively. The first 27 (mouse) or 26 (human) amino acids constitute a signal peptide that is absent from the mature protein, followed by a 161 (mouse) or 156 (human) amino acid extracellular domain, a 22 (mouse) or 23 (human) amino acid transmembrane domain, and a 21 (mouse) or 30 (human) amino acid cytoplasmic tail. The cytoplasmic domains of murine and human FL are only 52% identical and are much more divergent than the cytoplasmic domains of murine and human KL (92% identical). Why the cytoplasmic domains of mouse and human FL are so much more divergent in sequence than the cytoplasmic domains of mouse and human KL is unknown. The mouse and human FL proteins each contain two potential sites for N-linked glycosylation. The human FL protein contains N-linked sugars (Claudia Jochheim, Immunex; unpublished observation).

The mature mouse and human KL proteins (from which the amino acid signal sequence has been cleaved) undergo proteolytic cleavage to generate a soluble, biologically active, 164-165 amino acid protein.32,33,79,86 The primary site for proteolytic cleavage is encoded within exon six33; however, mutagenesis experiments have shown that there is a secondary proteolytic cleavage site just upstream of the transmembrane region within exon 7.87 This secondary site is used only if the primary site is missing, which can occur by splicing out the sixth exon.79,88,89 

Splicing has been suggested to be a method of regulating the generation of soluble versus membrane-bound forms of the protein. Alternative splicing of the sixth exon of the KL gene has been reported in both mouse and human cells.40,79,88,90,91 The cell-bound form of KL appears to be required for normal development in mice since a mutation (Sld) that eliminates the membrane-bound form of the factor, but still makes a biologically active soluble form, results in developmental abnormalities.88,92 Huang et al90 showed that there is tissue-specific expression of the different isoforms. The physiologic significance of these altered isoform ratios is unknown but presumably reflects the capacity of each tissue to produce a form of KL that is capable of interacting with specific c-kit-expressing cells.

It is unclear what regulates the proteolytic cleavage of KL, and what, if any, the physiologic effects of this process are. The protease responsible for cleavage of KL has not been identified, and it is unknown if it is the same protease that generates soluble, biologically active forms of M-CSF and FL.48,49,93 

Multiple isoforms of both mouse and human FL have been identified by analysis of multiple cDNA clones and PCR.48-50,94 The biological significance of these isoforms is presently unknown. The predominant isoform of human FL is the transmembrane protein that is biologically active on the cell surface.48-50 This isoform is also found in the mouse, although it is not the most abundant isoform in that species (see below). The transmembrane FL protein can be proteolytically cleaved to generate a soluble form of the protein that is also biologically active.48 Neither the protease responsible for this cleavage nor the exact site in the FL amino acid sequence where cleavage occurs has been identified.

The most abundant isoform of murine FL95 is an alternative, 220 amino acid form that is membrane bound, but is not a transmembrane protein.49,94 This form arises due to a failure to splice an intron from the mRNA. This leads to a change in the reading frame, which terminates in a stretch of hydrophobic amino acids that serve to anchor the protein in the membrane.50 This isoform is missing the spacer and tether regions that contain the proteolytic cleavage site seen in the transmembrane isoform. As a result, this membrane-associated isoform is resistant to proteolytic cleavage,94 although it is biologically active on the cell surface. This isoform has not been identified in any human FL cDNAs examined.

A third FL isoform identified in mouse94 and human95 tissues arises because of an alternatively spliced sixth exon. This exon introduces a stop codon near the end of the extracellular domain and thereby generates a soluble, biologically active protein that appears to be relatively rare compared with other isoforms.95 Another method of generating soluble FL in the human is to splice out the transmembrane domain,50 but the relative abundance of this isoform has not been quantitated.

There is a difference between KL and FL in regard to their alternatively spliced sixth exons. The amino acids in exon 6 of mouse and human KL are nearly identical, whereas those of mouse and human FL have virtually no homology.95 In the case of KL, the sixth exon is normally part of the transmembrane protein and contains the proteolytic cleavage site. In the case of FL, it is not a part of the transmembrane protein; introduction of the sixth exon results in the generation of a soluble protein due to a shift in the reading frame. Thus, evolution has made two different uses of the sixth exon of KL and FL, allowing the generation of a soluble protein by different mechanisms.

The genomic loci encoding the c-kit, flt3, and c-fms receptors share overall conservation of exon size, number, sequence, and exon/intron boundary positions,96 and these genes have likely arisen from a common ancestral gene. The genomic loci encoding the mouse97 and human98-100 c-kit receptors show clear evidence of evolutionary conservation. The coding region of the c-kitreceptor encompasses 21 exons, and both the mouse and human loci span more than 70 kb of genomic sequence.

The human flt3 receptor genomic locus is approximately 100 kb in size.101 The exon:intron structure of the entire receptor has been reported to contain 24 exons,102 but only the portion of the gene encoding the C-terminal domain has been published.

The genomic locus encoding KL has been cloned from the human,33 rat,33 and mouse.103 The human KL locus is more than 50 kb in length (Vann Parker, Amgen, Thousand Oaks, CA; personal communication) and consists of eight exons that contain the entire coding region of the protein. The intron:exon boundaries identified within the rat, human, and murine genes occur at identical positions. In the case of the mouse protein, a ninth exon is present and encodes the C-terminal end of the cytoplasmic domain.103 

The genomic loci encompassing the coding regions of mouse and human FL are approximately 4.0 kb and 5.9 kb, respectively; the coding region comprises 8 exons.95 The human FL locus is thus significantly smaller than the human KL locus. The sizes of the individual FL exons are well conserved between species,95although the intron sizes are much more variable.

The genomic locus encoding M-CSF also contains eight exons.104 A comparison of exon sizes between FL, KL, and M-CSF shows that identically numbered exons are similar in size in all three proteins.95 If the sizes of the exons are taken as a measure of overall relatedness, then M-CSF and KL are more closely related to each other than they are to FL. For example, the sizes of exons 3 and 4 are identical between M-CSF and KL, but are not the same as the corresponding exons in FL. The location of the introns in the three genes are also fairly well conserved, indicating that these proteins are probably ancestrally related.

The murine c-kit locus is located in the D-E region of mouse chromosome 511,12 near two other tyrosine kinase receptors (PDGF A and flk-1/KDR). The murine flt3 receptor gene is also on chromosome 5, but at the G region.41 The flt3 receptor105 is located less than 350 kb from the murine flt tyrosine kinase receptor106 but is separated from the clustered c-kit, PDGF A, and flk-1/KDR receptors.

The human c-kit locus is on the centromeric region of chromosome 4, in the area of 4q31-34,534q11-21,54 and 4q11-12.107 The gene encoding the human flt3 receptor maps to chromosome 13q12,41 again near the flt receptor locus. The flt3 and flt genes are linked105 in a head to tail fashion and are separated by about 150 kb.101 

The KL gene is, as expected, encoded on mouse chromosome 10 and is deleted in some, but not all, Sl alleles.32,35,36The FL gene maps to the proximal portion of mouse chromosome 7.94 

The gene encoding human KL has been mapped to chromosome 12q22-2440 and 12q14.3-qter108 in a region that is syntenic with mouse chromosome 10. The human FL gene maps to chromosome 19q13.3-13.4,94,109 which is syntenic with mouse chromosome 7. The chromosomal locations of KL, FL, M-CSF, and their receptors are summarized in Table 1.

Table 1.

Chromosomal Locations of the c-kit,c-fms, and Flt3 Receptors and Their Ligands

Mouse Human
Receptors  
 Flt3  5G  13q12 
 c-kit 5D-E  4q11-34  
 c-fms 18 5q32-33  
Ligands  
 FL  7  19q13.3-13.4  
 KL  10 12q14.3-qter  
 M-CSF  3  1p13-21 
Mouse Human
Receptors  
 Flt3  5G  13q12 
 c-kit 5D-E  4q11-34  
 c-fms 18 5q32-33  
Ligands  
 FL  7  19q13.3-13.4  
 KL  10 12q14.3-qter  
 M-CSF  3  1p13-21 

The exact defect in the c-kit receptor has now been identified at the molecular level for a number of alleles of the Wlocus.24-28 Most of the alleles result from point mutations in the cytoplasmic domain of the receptor; these changes decrease the tyrosine-phosphorylating activity of the protein. However, in several cases, the mutations appear to be of a regulatory instead of a structural nature and result in reduced expression of the c-kitreceptor.

There is a rare, autosomal dominant genetic disease in humans known as piebald trait. Affected individuals have a white forelock and large, nonpigmented patches on the chest and/or other areas. All cases of piebald trait that have been molecularly analyzed result from missense or frameshift mutations in the c-kit tyrosine kinase receptor (Ezoe110 and references therein). Affected individuals are heterozygous for defects in the c-kit protein; the dominant nature of the trait reflects the dominant-negative effects of the mutant c-kit allele. The dominant-negative effects of these mutations are thought to result because receptor dimerization is required for proper biological function.

Because pigmentation defects in W and Sl mice are often indistinguishable, it would be reasonable to expect that at least some cases of piebald trait in humans would arise from mutations in the KL gene, ie, from a defect in the ligand instead of the receptor. However, no defects in the KL gene have been reported in piebald humans. Piebald trait thus represents the human homologue of the W mutation in mice.

Mutations at the Steel locus35 have occurred spontaneously or have been induced by chemical mutagenesis, x-ray irradiation, or transgene insertion.111 In addition to theSld mutation (see above), the molecular defect responsible for three other Sl mutations has been identified. In the Sl17H mutation,103 the cytoplasmic tail of KL is altered as a result of a splicing defect; in contrast, the Slcon and Slpanmutations are of a regulatory nature and result in altered, tissue-specific expression of mRNAs encoding KL.112 

In contrast to the well-described mutations in the c-kitreceptor and its ligand (see above), there are no reports of any genetic defects associated with either the flt3 receptor or its ligand.

As described above, FL maps to human chromosome 19q13.3. Trisomy 19 is strongly associated with myeloid malignancies.113 However, whether overexpression of FL plays a role in the increased incidence of leukemia in trisomy 19 remains to be determined.

The expression of the c-kit and flt3 receptors, and not their ligands, is the key to understanding the function of these growth factors. Numerous studies have shown that both KL and FL are widely expressed in different tissues, in contrast to their receptors, which are expressed on a more limited number of cells, especially in the case of flt3. KL is widely expressed during embryogenesis,114-116 suggesting that KL may affect the growth, survival, and/or differentiation of cells in addition to the three lineages (hematopoietic cells, germ cells, and melanocytes) shown to be affected in both W and Slmutant mice. Cells expressing KL are frequently contiguous with cells expressing c-kit, ie, ligand and receptor expression are complementary. KL is expressed on stromal cells,117,118fibroblast,26,79,119 endothelial cells,117visceral yolk sac,115 and other places.

FL, like KL, is widely expressed in both murine and human tissues.49,50,94 Highest levels of FL mRNA on human tissue Northern blots are in peripheral blood mononuclear cells, but the ligand is also expressed in almost every tissue that has been examined.48-50 Mouse developmental in situ hybridization studies have not yet been performed with FL, although it would be interesting to see how the distribution of FL would compare with flt3 receptor.120 

Expression of the c-kit receptor has been extensively surveyed on mouse and human hematopoietic cell lines (Table 2). It is seen on only a small percentage of myeloid and myeloblastic cell lines.121-124In contrast, the majority of erythroid and erythroleukemia cell lines express c-kit,121-123,125 as do virtually all megakaryocytic cell lines.121,123,125 Mast cell lines generally express c-kit.51,126-128 In contrast, expression of c-kit is generally not seen on lymphoid leukemia cell lines (including pre-B, B, and T cells),121,123,125 on B-cell or T-cell lymphoma cell lines,121,122,125 or on myeloma cell lines.121 

Table 2.

Expression of c-kit and Flt3 Receptors on Murine and Human Cell Lines

c-kitFlt3
Myeloid  Few positive  Mostly positive* 
Monocytic  Few  About 50%  
Erythroid  Most Few  
Megakaryocytic  Most  Few  
Mast cell  All None  
Lymphoid  
 Pro-B  None  Most  
 Pre-B None  Most* 
 B  None  Few  
 T  None  Few 
 Mature NK  ND  None  
Lymphomas  None  About 25% 
Myeloma  None  None 
c-kitFlt3
Myeloid  Few positive  Mostly positive* 
Monocytic  Few  About 50%  
Erythroid  Most Few  
Megakaryocytic  Most  Few  
Mast cell  All None  
Lymphoid  
 Pro-B  None  Most  
 Pre-B None  Most* 
 B  None  Few  
 T  None  Few 
 Mature NK  ND  None  
Lymphomas  None  About 25% 
Myeloma  None  None 

Results tabulated from a large number of reports. For individual references, see the sections of this report detailing the expression patterns for each of these receptors.

Abbreviation: ND, not determined.

*

Different expression patterns have been reported on mouse versus human cells; see text for details.

Flt3 receptor expression on mouse and human cell lines is quite different from that of c-kit. No flt3 expression is seen on any of the mouse myeloid, macrophage, erythroid, megakaryocyte, or mast cell lines examined46,129 or most early mouse B-cell lines, but it has been reported on several mature B-cell lines.129This lack of expression is different from what is seen on most human pre-B-cell lines, which do express flt3 receptor.123,130 In addition, flt3 expression has been seen on only one mouse pro-T cell line, but not on any T-cell lines.46,129 

A number of studies have been published that show expression of flt3 receptor on a limited range of human cell lines. The flt3 receptor is found on a high percentage of human myeloid and monocytic cell lines,123,129,130 in contrast to mouse cell lines.46,129 No flt3 expression is seen on myeloma cell lines,129,130 and only a few megakaryocytic cell lines are positive.123,129,130 All erythroid and erythroblastic cell lines are flt3 negative as well.129,130 

Among lymphoid cell lines, pro-B as well as pre-B lines are flt3 receptor positive,129,130 whereas natural killer (NK) cell lines and Hodgkin's cell lines are negative,130 as are all T-cell lines.123,129,130 

Both the c-kit and flt3 receptors are frequently seen on acute myelogenous leukemia (AML) blasts. The c-kit protein is expressed on blast cells obtained from a high percentage of patients with AML from all French-American-British (FAB) subtypes.61,124,131-139 Receptor levels on AML blast cells are variable, but in general are similar to or less than c-kitlevels on normal stem and progenitor cells.140 

Expression of the flt3 receptor in primary leukemias has also been investigated and recently reviewed.141 As with c-kit, the majority of adult AML samples from all FAB classes are positive for flt3 receptor expression.57,142-146 

Among lymphoid leukemias, little or no expression of c-kit is observed on blast cells in acute lymphoblastic leukemia (ALL).133,143 c-kit is expressed on Reed-Sternberg cells in about half of Hodgkin's disease patients as well as on some anaplastic large-cell lymphoma samples.147 

All B-lineage ALL samples examined are flt3 receptor positive,142-144 as are most hybrid (also known as mixed or biphenotypic) leukemia samples.144 The greatest variability reported in flt3 receptor expression is on T-lineage ALL, which have been reported to be all negative,142 have a small percentage that are positive,143 or have about half of the samples positive.144 In contrast, both T-cell and B-cell lymphomas are negative for flt3 receptor expression.144Tandem in-frame duplications in the juxtamembrane region of the human flt3 receptor have been reported to be associated with both leukocytosis148 and leukemic transformation.149 

The c-kit receptor is expressed on a majority of samples from chronic myelogenous leukemia (CML) patients in blast crisis134,150 and at least some samples of chronic phase CML138 and CML in blast transition.151 In contrast, almost all chronic-phase or accelerated-phase CML samples are negative for flt3 receptor expression.143,144 However, about two thirds of the samples from CML patients in blast crisis are flt3 receptor positive.143,144 

AML.

Numerous studies have been performed on human leukemia samples to determine whether the cells proliferate in response to KL, FL, or other growth factors, although a lack of proliferation should not necessarily be considered negative expression. For example, a growth factor could drive differentiation or inhibit apoptosis; in fact, both KL152 and FL153 have been shown to have this latter effect. In the case of nonproliferative cells, the cells may be truly nonresponsive or may be producing endogenous ligand, and thus are refractory to exogenously added growth factor.

c-kit receptor expression is variable among AML FAB subtypes and does not predict responsiveness to KL.145 The majority of AML samples proliferate in response to KL.61,131,137,154,155 Many of these studies show that KL synergizes with other cytokines to enhance the proliferation of leukemic blast cells. Some AML cell lines express KL in addition to c-kit,140,156 suggesting that an autocrine loop may play a role in the transformation of these cells. However, the low level of KL expression on some AML cells has led one group to conclude that a c-kit and KL autocrine cycle is not common in AML.140 

Whether flt3 receptor or its ligand play a causal role in the development of human leukemias has not been determined. A large percentage of AML cells from children142 and adults145,146 proliferate (as measured by both [3H]-thymidine incorporation or colony formation) in response to FL. Within age groups (children or adults), some FAB subtypes show a greater response compared with others.142,146 It is unclear whether there is a difference in the FL responsiveness of flt3 receptor-positive AML samples of different FAB subtypes from children and adults because not enough samples of each FAB subtype have been analyzed.

Primary AML samples that proliferate in response to FL also frequently proliferate in response to granulocyte-macrophage colony-stimulating factor (GM-CSF), interleukin-3 (IL-3), and KL, and additive or synergistic responses are observed. Some AML cells are therefore similar to normal hematopoietic progenitor cells in that both show synergistic responses to FL in combination with other cytokines. Many of the AML samples that do not proliferate in response to FL do proliferate in response to other cytokines,142 indicating that the cells do not lack a general capacity to proliferate. In summary, flt3 receptor expression on AML samples is not predictive of FL responsiveness, just as c-kit expression is not predictive of KL responsiveness.

CML.

KL can weakly stimulate the proliferation of CML blast cells on its own and strongly stimulate them in the presence of IL-3 and/or GM-CSF.138 Culturing of bone marrow (BM) cells from CML patients in the presence of KL favors the growth of malignant progenitor cells.157 In contrast, preliminary results suggest that FL favors the outgrowth of benign progenitors from 5-FU-treated CD34+ CML BM cells at the expense of malignant cells158 and that FL generates a significantly greater percentage of normal progenitors (Philadelphia chromosome-negative cells) compared with KL.

ALL.

Because c-kit is not generally expressed on ALL cells,124,133,134,139 the capacity of these cells to proliferate in response to KL has not been examined. As mentioned above, all B-lineage ALL and some T-lineage ALL samples express flt3 receptor. However, only a small percentage of B-lineage ALL samples proliferate in response to FL.142 

In one study, pediatric T-lineage ALL samples did not proliferate in response to FL, but none of these samples was positive for flt3 expression.142 In a separate study on a variety of ALLs, several flt3 receptor-positive samples proliferated in FL.159 However, the majority of samples failed to proliferate in FL, even though they were flt3 receptor positive.159 Flt3 receptor expression is therefore not predictive for proliferation of ALL cells to FL in vitro.

Studies of cytokine receptor expression have proven valuable in pinpointing where specific ligand-receptor pairs have biological activities. Not only can such studies identify cell types in which a specific receptor might be important, they also allow functional characterization of distinct cell populations separated based on various levels of receptor expression. The expression of c-kitand flt3 in the hematopoietic system has been studied in detail, and in the following sections we review the findings of flt3 and c-kitexpression on various cell types (summarized in Fig 2), followed by the in vitro biological effects (summarized in Table 3) of FL and KL on the same cell types. It is important to emphasize that the extensive c-kitand flt3 expression studies to be described have inherent limitations. Most expression studies have been performed by flow cytometric evaluation of cell-surface c-kit and flt3 expression. Because flow cytometry has a rather high detection limit (∼500 molecules/cell), so- called c-kit and flt3 populations might prove to express low levels of c-kit and flt3, respectively. On the other hand, reverse transcriptase-PCR (RT-PCR) detection of c-kitand flt3 mRNA has much greater sensitivity, but unless performed at the single-cell level does not provide a quantitative measurement of c-kit+ and flt3+ cells. Thus, a minor contaminating (nonrelevant) cell type might account for detected expression (particularly relevant for heterogenous primary cell populations).

Fig. 2.

c-kit and Flt3 expression in the hematopoietic hierarchy. The figure indicates expression of c-kit (red, upper symbol on side of each cell) and flt3 (green, lower symbol on side of each cell) on various classes of hematopoietic stem and progenitor cells as well as mature blood cells, as described in the text. Because most hematopoietic cell populations are heterogeneous and hard to purify, it is not possible to exclude c-kit and/or flt3 expression on a minority of cells in the different cell populations. Therefore, the figure illustrates the c-kit and flt3 receptor status on the majority of cells within a specific population, based on studies of receptor expression and/or functional studies. As discussed in the text, the proposed hierarchy of pluripotent stem cells is based solely on different levels of c-kit and flt3 expression and does not take into account other stem cell antigens/characteristics, which are likely to uncover additional heterogeneity. Symbols: (−) most/all cells appear to lack c-kit or flt3 expression; (+) most/all cells appear to express c-kit or flt3; (+/−) the cell type appears to consist of significant receptor-positive as well as receptor-negative populations; (?) sufficient expression or functional data not available; (high and low) cell populations have been separated based on high and low levels of c-kit expression. Abbreviations: BFU, burst-forming units; CFU, colony-forming units; E, erythroid; Mk, mega karyocyte; G, neutrophilic progenitor; M, monocyte/macrophage; DC, dendritic cell; Baso, basophil; RBC, red blood cell; NK, natural killer cell.

Fig. 2.

c-kit and Flt3 expression in the hematopoietic hierarchy. The figure indicates expression of c-kit (red, upper symbol on side of each cell) and flt3 (green, lower symbol on side of each cell) on various classes of hematopoietic stem and progenitor cells as well as mature blood cells, as described in the text. Because most hematopoietic cell populations are heterogeneous and hard to purify, it is not possible to exclude c-kit and/or flt3 expression on a minority of cells in the different cell populations. Therefore, the figure illustrates the c-kit and flt3 receptor status on the majority of cells within a specific population, based on studies of receptor expression and/or functional studies. As discussed in the text, the proposed hierarchy of pluripotent stem cells is based solely on different levels of c-kit and flt3 expression and does not take into account other stem cell antigens/characteristics, which are likely to uncover additional heterogeneity. Symbols: (−) most/all cells appear to lack c-kit or flt3 expression; (+) most/all cells appear to express c-kit or flt3; (+/−) the cell type appears to consist of significant receptor-positive as well as receptor-negative populations; (?) sufficient expression or functional data not available; (high and low) cell populations have been separated based on high and low levels of c-kit expression. Abbreviations: BFU, burst-forming units; CFU, colony-forming units; E, erythroid; Mk, mega karyocyte; G, neutrophilic progenitor; M, monocyte/macrophage; DC, dendritic cell; Baso, basophil; RBC, red blood cell; NK, natural killer cell.

Close modal
Table 3.

In Vitro Effects of KL and FL in the Murine and Human Hematopoietic System

Cell Type Response KL FL
Primitive progenitors/candidate stem cells  Growth  Synergy  Synergy 
 Viability  +  +  
 Adhesion  +  ND 
Erythroid progenitors  
 BFU-E  Growth  Synergy  −  
 Adhesion  +  −  
 CFU-E  Growth  +  −  
Myeloid (GM) progenitors  Growth  Synergy Synergy  
 Viability  +  +  
 Adhesion +  ND  
Megakaryocytopoiesis  
 BFU-Mk/CFU-Mk  Growth +  +  
 Mk maturation  +  −  
Mast cells  Growth  +  −  
 Maturation  +  −  
 Adhesion  +  −  
 Migration +  −  
 Activation  +  −  
B lymphopoiesis  
 Murine stem cells  Growth/  commitment Weak  Strong  
 Murine pro-B cells  Growth  Synergy Synergy  
 Human pro-B cells  Growth  −  Synergy 
T lymphopoiesis  
 Murine pro-T cells  Growth  Synergy Synergy  
 Human pro-T cells Stromadependent  growth  Synergy  Synergy  
NK cells  
 NK cell progenitors  Growth  Synergy  Synergy 
 NK cells  Growth  Synergy  ND  
 Viability  +  ND  
Dendritic cells  
 DC progenitors  Growth  Synergy Synergy 
Cell Type Response KL FL
Primitive progenitors/candidate stem cells  Growth  Synergy  Synergy 
 Viability  +  +  
 Adhesion  +  ND 
Erythroid progenitors  
 BFU-E  Growth  Synergy  −  
 Adhesion  +  −  
 CFU-E  Growth  +  −  
Myeloid (GM) progenitors  Growth  Synergy Synergy  
 Viability  +  +  
 Adhesion +  ND  
Megakaryocytopoiesis  
 BFU-Mk/CFU-Mk  Growth +  +  
 Mk maturation  +  −  
Mast cells  Growth  +  −  
 Maturation  +  −  
 Adhesion  +  −  
 Migration +  −  
 Activation  +  −  
B lymphopoiesis  
 Murine stem cells  Growth/  commitment Weak  Strong  
 Murine pro-B cells  Growth  Synergy Synergy  
 Human pro-B cells  Growth  −  Synergy 
T lymphopoiesis  
 Murine pro-T cells  Growth  Synergy Synergy  
 Human pro-T cells Stromadependent  growth  Synergy  Synergy  
NK cells  
 NK cell progenitors  Growth  Synergy  Synergy 
 NK cells  Growth  Synergy  ND  
 Viability  +  ND  
Dendritic cells  
 DC progenitors  Growth  Synergy Synergy 

Cell types or responses in which neither KL nor FL are known to have an effect are not listed.

Abbreviations: −, no effect found on indicated response (in some cases not specifically investigated but cell type lacks receptor for indicated ligand); +, stimulatory effect of ligand alone on indicated response; synergy, effect predominantly through synergistic interaction with other cytokines; ND, not determined.

c-kit and flt3 expression in the hematopoietic system appear predominantly restricted to the progenitor/stem cell compartment (outlined in the following sections). However, some differentiated blood cells also express these receptors (Fig 2).

c-kit is expressed on primary mast cells as well as mast cell lines and primary neoplastic mast cells.160 In addition, c-kit is constitutively activated in a number of mast cell tumor lines (mastocytomas),127,161 but mast cells do not express flt3.128 

There are other differentiated hematopoietic cells that express c-kit and/or flt3, although the functional significance is less clear. In mouse BM, very low levels of c-kit can be detected on promyelocytes and myelocytes, but not on neutrophils.162 Approximately 50% of murine BM eosinophils and monocytes express low levels of c-kit.162 Seven percent of lymphocytes in murine BM express high levels of c-kit.162 However, still other studies suggest that mature B and T cells do not express c-kit; therefore, this small fraction of c-kit+ cells might represent B- and T-cell precursors/progenitors.163-165 

Similar studies have revealed that flt3 expression in murine BM is restricted to blast cells, monocytes, and a small fraction of lymphocytes.166 Nucleated murine erythroid cells lack both c-kit and flt3 expression.162,166 Early murine megakaryocytes (stage I and II) express c-kit, whereas the most mature (stage III) megakaryocytes appear to be c-kit.167 Also, human megakaryocytes express c-kit,61,168 but not flt3.169 In addition, activated but not resting platelets express c-kit.170 

Initial studies indicated that flt3 mRNA is expressed by murine B and T cells from thymus, spleen, and peripheral blood.18 However, several later studies of mature murine B and T cells suggest that these do not express flt3.166,171 Thus, the initial findings potentially were due to a small fraction of contaminating flt3+ cells, such as more primitive B- and T-cell progenitors.

Peripheral human blood cells contain less than 0.1% c-kit+ cells, suggesting that very few mature human blood cells express c-kit.172-174 c-kit is constitutively expressed on a small subset of resting human NK cells in peripheral blood that are characterized by high CD56 expression, whereas c-kit is not expressed on the larger fraction of more differentiated NK cells with low CD56 expression.175 These c-kit+ NK cells appear to be the only mature, resting lymphocytes that constitutively express c-kit.

No expression of flt3 mRNA has been reported on mature lympohematopoietic cells fractionated from human peripheral blood17 or B cells, T cells, monocytes, or granulocytes.144 However, in other studies, monocytes and granulocytes have been shown as weakly positive at the mRNA and cell-surface level.16,176 

The effects of KL on mast cell populations have been extensively reviewed6 and will be only briefly summarized here. KL regulates the migration, maturation, proliferation, and activation of mast cells in vivo.6 Injection of recombinant KL into rodents,86,177 primates,178 or humans179 results in an increase in mast cells at both the site of injection and at distant sites. Treatment of rats with KL generates both connective tissue mast cells and mucosal mast cells.177 Animals treated with KL generally do not appear to suffer from serious adverse events despite the large-scale expansion of mast cells in vivo.178 However, at least one study has shown that KL administration to mice leads to degranulation of mast cells in the lungs, which leads to acute respiratory distress.180 The effects of KL on mast cells may have a significant impact on the clinical potential of this molecule for humans.179,181,182 

In contrast to c-kit, flt3 is not expressed on primary mast cells or mast cell lines, and these cells, not surprisingly, do not respond to FL.51,128 This lack of flt3 expression on mast cells is one of the key differences between KL and FL.

Half of c-kit+ murine BM cells coexpress lineage-specific cell surface antigens such as GR-1 and MAC-1 (Lin+), characteristic of cells committed to the myeloid lineage, whereas the remaining half express higher levels of c-kit and are Lin, suggesting that uncommitted progenitor cells might express higher levels of c-kit than those committed to the myeloid lineage.183 Indeed, murine in vitro clonogenic progenitor cells committed to the myeloid lineage and colony-forming units-spleen (CFU-S) progenitors are almost completely depleted in c-kit BM cells, showing that most, if not all, clonogenic myeloid progenitor cells express c-kit.183-188 

Most c-kit+ human BM and fetal liver cells express the progenitor-associated CD34 antigen,172-174 suggesting that overlapping (but not identical) populations each express these two progenitor cell antigens. c-kit+ human BM and fetal liver cells are highly enriched and contain all or most in vitro clonogenic progenitor cells with a myeloid (granulocyte/monocyte), megakaryocytic, and/or erythroid potential.172-174,189 

CD34highCD64+ cells, which are virtually a pure population of human GM progenitor cells, express high levels of c-kit, whereas the more mature CD34lowCD64+ cells express lower levels of c-kit,190 suggesting downregulation of c-kit expression during GM differentiation. Similarly, erythroid progenitor cells (CD34highCD64CD71high and CD34lowCD64CD71high) also express high levels of c-kit.190 Although some studies have suggested that a subclass of mature erythroid progenitor cells (colony-forming units-erythroid [CFU-E]) might not be KL-responsive, c-kit expression has been demonstrated on human CFU-E and erythroblasts.174 The vast majority of human megakaryocyte progenitor cells (burst-forming unit-megakaryocyte [BFU-Mk] as well as colony-forming unit-megakaryocyte [CFU-Mk]) are also c-kit+.191 

Whereas almost 90% of murine BM blast cells express c-kit,162 flt3 expression is restricted to 30% of murine BM blast cells.166 The majority of lineage-restricted murine myeloid and erythroid BM progenitor cells are LinSca-1 and express c-kit.188 However, less than half of these LinSca-1c-kit+progenitors express flt3.166 

More than 60% of flt3+ human BM cells coexpress CD33, a myeloid cell-surface antigen, suggesting that flt3 might be expressed on subsets of myeloid progenitor and/or mature cells.57 Most human CD34+ BM and cord blood cells express flt3, and most GM progenitors express flt3, whereas CD34+flt3+ cells are depleted in erythroid progenitors.176 The majority of CD34+c-kit+ BM and cord blood cells coexpress flt3, but a significant (10% to 25%) population is flt3.

Flt3 appears to be shut off before erythroid differentiation and gradually downregulated during GM differentiation.192 In contrast, c-kit expression is gradually downregulated during both erythroid and GM differentiation.192 Thus, flt3 appears to be expressed on subpopulations of myeloid (GM) progenitor cells, but not on erythroid progenitor cells.

Myeloid-derived dendritic cell (DC) progenitors appear to express c-kit and flt3, because they respond to KL and FL in combination with other cytokines (see DC section for details). However, neither ligand has been shown to have effects on mature DC.193-196 

Besides the mast cell deficiency, the dominating hematopoietic defect resulting from severe mutations in the W or Sl loci is a macrocytic anemia.6,10 KL enhances the in vitro cloning frequency as well as the clonal size of murine79,197 and human33,172,174,198-200 erythroid progenitor cells. KL has its most potent growth promoting effects on early erythroid progenitor cells (BFU-E), whereas more mature progenitors (CFU-E) are less responsive to KL-stimulation.172-174,191,201 

The effects of KL on the growth of BFU-E are predominantly synergistic and require costimulation with erythropoietin (EPO).79,172,174,197-200 However, KL can, in combination with IL-6 and soluble IL-6 receptor, promote EPO-independent growth of human BFU-E in vitro.202 Furthermore, c-kit might activate the EPO receptor by inducing its phosphorylation on tyrosine.203 KL also promotes the adhesion of human BFU-E to fibronectin.204 

In contrast, FL appears to have little or no effect on murine205,206 and human49,50,192,207,208erythropoiesis in vitro. This is in agreement with the observed lack of flt3 expression on normal erythroid progenitor cells166,192as well as erythroleukemic cell lines.123,130 

Although Sl/Sld mice have normal levels of platelets, their BM displays reduced numbers of mature megakaryocytes and megakaryocyte progenitor cells.209-211 Administration of KL to Sl/Sld mice not only reverses the macrocytic anemia, but results in enhanced platelet production.36 In vitro, KL enhances megakaryocyte progenitor cell cloning frequency and growth potential in combination with other cytokines, including GM-CSF, IL-3, IL-6, and IL-11.168,212-215 Whereas some studies have found little or no effect on megakaryocyte maturation and ploidy, others have suggested that KL can promote megakaryocyte maturation and ploidy,216and subsets of early megakaryocytes express c-kit.167 

Thrombopoietin (TPO) is the primary regulator of megakaryocyte and platelet production,217 and KL appears to interact with TPO at two levels in the hematopoietic hierarchy. First, a synergistic interaction is observed on committed megakaryocyte progenitor cells, enhancing megakaryocyte production.217-221 In addition, KL and TPO interact synergistically on candidate murine and human stem cell populations to stimulate multilineage growth in vitro.222-226 Thus, the primary role of KL in platelet production might be through its interaction with TPO.

Unlike W/Wv andSl/Sld mice, flt3 knockout mice have not been reported to have any defects in megakaryocyte and platelet production,227 and FL alone or in combination with IL-3, KL, or TPO has no effect on in vitro growth of murine megakaryocyte progenitor cells.65 Similarly, FL has no effect on megakaryocyte ploidy by itself or in combination with TPO.65 In contrast, FL acts synergistically with TPO to enhance the growth of candidate murine stem cells.223 

Some data suggest that FL might have effects on human megakaryocytopoiesis. Some megakaryocytic leukemic cell lines, as well as primary megakaryoblastic leukemic cells, express flt3, although less frequently than c-kit.65,123,130 In addition, studies of FL effects on primary BM cells have demonstrated effects on megakaryocyte formation.228 Unlike KL, FL has been reported to have no synergistic interaction with TPO on in vitro clonogenic growth of human megakaryocyte progenitor cells.169 Thus, the finding that FL and TPO synergistically promote prolonged megakaryocyte progenitor cell formation in long-term cultures of human CD34+ cord blood cells229 could result from a recruitment of primitive (uncommitted) progenitor cells that might subsequently become responsive to TPO alone.

About 25% of B220+ murine BM cells express c-kit, accounting for more than half of the total c-kit+cells.164 However, no BM cells (or fetal liver cells) expressing cytoplasmic μ coexpress c-kit, suggesting that c-kit expression is restricted to the earliest stages of B-cell progenitors, whereas the pre-B-cell and subsequent stages are c-kit.163,164,230,231 

Flt3 mRNA is expressed in early murine pre-pro and pro-B cells, whereas pre-B cells, as well as immature and mature B cells, are devoid of flt3 expression.171 A similar pattern of flt3 expression is seen at the cell surface of pro-B, pre-B, and mature B cells.166c-kit is also expressed at low levels on subsets of human pro-B cell progenitor cells (CD34+CD19+).173,189,190Twenty-five percent of BM CD34+CD19+ (pro-B cells) express flt3, as do subfractions of CD10+ and CD20+ B-cell precursors.176 

c-kit is expressed at high levels on the most primitive subsets of murine fetal and adult thymocytes, including CD4CD8CD3CD44+CD25+pro-T cells and more primitive CD4loCD8CD3 thymocytes, the latter cells also having the potential to develop into B cells.165,232-235 When thymocytes develop into CD4CD8CD3CD44CD25+pre-T cells, they still express low levels of c-kit, which is lost in later stages of T-cell development.165 

Like c-kit, flt3 expression is restricted to the most immature CD4CD8 murine thymocytes, whereas more mature thymocytes expressing CD4 and/or CD8 are flt3.19 

Because human NK cell progenitor cells respond to KL or FL (see separate section), they most likely express c-kit and flt3. However, there is as yet no direct evidence for c-kit or flt3 expression on NK cell progenitor cells, and the few human NK cell lines examined lack flt3 expression.130,236 

Multipotent lymphoid progenitor cells capable of producing DC express high levels of flt3.237 Because a DC-restricted lymphoid progenitor has not yet been identified, c-kit and flt3 expression on such a CFU-DC remains to be established.

Although no reduction in cells of the B-cell lineage has been reported in adult W mutant mice, embryonic mice deficient in c-kit or KL expression have reduced numbers of B-cell progenitor cells in fetal liver.238 Such a reduction could indicate a direct role of c-kit and its ligand in B lymphopoiesis or, alternatively, an indirect effect of a depleted pool of pluripotent stem cells and/or altered stromal cells in these mice.186 

KL can synergize with IL-7 to promote stroma-independent growth of murine BM pro-B- and pre-B-cell progenitors unresponsive to IL-7 alone, whereas KL lacks proliferative activity on B220++ pre-B cells.33,118,239,240 One study found that KL in combination with IL-7 could promote development of pre-B cells and expression of μ-heavy chain118; other studies have not found KL plus IL-7 sufficient to allow differentiation of pro-B cells into pre-B cells in vitro, even though such pro-B cells coexpress c-kitand IL-7 receptors.231,239,240 Furthermore, a blocking antibody against c-kit inhibits the growth of murine pro-B cells cultured on stromal cells in the presence of IL-7, but has no effect on pre-B-cell differentiation supported by the same stroma cells.163,241,242 Similarly, KL in combination with IL-7 can replace the requirement for stroma to induce pro-B-cell proliferation, but not differentiation into pre-B cells.239In addition to its ability to promote growth of committed pro-B cells, KL in combination with IL-7 can stimulate stroma-independent B-cell progenitor cell development from candidate murine stem cells243-245 or from bipotent macrophage-B-cell progenitor cells.246 

In vivo treatment of mice with a blocking antibody against c-kit results in an almost complete elimination of myeloid and primitive hematopoietic progenitor cells, leaving virtually no mature granulocytes and erythroblasts in the BM.164,183 However, the total number of BM cells are normal, of which the majority are B220+.164,183 A concomitant expansion in the number of pre-B-cell progenitor cells is observed,164,183suggesting that an interaction between c-kit and KL is not required for B-cell development in vivo. In support of this, W/Wstem cells are as efficient as wild-type stem cells at reconstituting BM B cells in RAG-2-deficient mice.247 Thus, unlike the critical role of c-kit/KL interaction in generation of the erythroid, myeloid, and T-cell lineages, c-kit-KL is not required for normal B-cell development in adult mice. The mechanism behind the intriguing observation that a c-kit antibody blocks the production of mature myeloid and erythroid progeny but enhances B-cell development remains unclear, although it appears to result from an indirect rather than a direct effect.

An important and distinct role of FL in early stages of B-cell development is supported by studies of flt3-deficient mice. These animals, unlike c-kit-deficient mice, have reduced numbers of pro-B cells in the BM, although the number of mature B cells is normal.227 These findings have also been confirmed in FL-deficient mice.248 

FL promotes the in vitro growth of early B-cell progenitor cells in a pattern distinct from that of KL. Primitive (CD43+B220lowCD24) B-cell progenitors in murine BM do not respond to either FL or IL-7 individually, but in combination the two cytokines induce a greater proliferative response than IL-7 plus KL.249 In contrast, more differentiated CD43+B220lowCD24+ B-cell progenitors fail to respond to FL, whereas KL enhances IL-7-induced proliferation, indicating that FL activity is restricted to an earlier stage of B-cell development than KL activity. Another important finding is the capacity of FL plus KL to promote the growth of CD43+B220lowCD24B-cell progenitor cells in the absence of IL-7.249 This might help explain why IL-7 receptor-deficient mice have normal levels of these primitive B-cell progenitors, but dramatic reductions in more differentiated B-cell progenitors and mature B cells.250 It could also explain why mice with a combined deficiency in flt3 and c-kit have a more severe reduction in early B-cell progenitors than mice deficient in flt3 only.227 

FL synergizes with IL-7 to enhance the production of B220+cells from B220+ as well as B220 murine BM cells.245 IL-7-independent B220+ cell development occurs in the presence of FL alone, but not KL alone, indicating a primary role of FL over KL in early murine B-cell development. Pro-B cells isolated from murine fetal liver also proliferate in response to either FL or KL in combination with IL-7, maintaining a population of early pro-B cells.251 

Because the B-cell defect in flt3-deficient mice is restricted to a reduction in the most primitive B-cell progenitors, an essential role of flt3/FL might be to promote B-cell development from progenitor/stem cells not yet committed to the B-cell lineage. In support of this, FL and KL can each promote the growth of fetal liver and BM progenitor cells with a combined myeloid and lymphoid potential.251,252 FL and IL-7 synergize to enhance the growth of primitive murine LinSca-1+ BM progenitors, resulting in production of almost exclusively pro-B cells, whereas KL plus IL-7 stimulate formation of 90% myeloid cells.252 

Studies of the early stages of human B-cell growth have been hampered by the lack of optimized in vitro systems. Therefore, the potential roles of KL and FL in human B-cell development remain to be elucidated. A stimulatory effect of KL on committed human B-cell progenitors has been suggested,253 although stromal and IL-7-dependent early B lymphoid growth from BM or cord blood cells in vitro is neither stimulated by KL nor inhibited by a neutralizing anti-KL antibody.254-256 In contrast, FL in combination with IL-7 promotes stromal cell-independent growth of human fetal BM pro-B cells (CD34+CD19+), whereas KL has no effect.256 

Although the precise roles of FL and KL in B lymphopoiesis remain to be determined, the available in vitro, in vivo, and knockout data suggest that flt3 and FL may be more critically involved in early B-cell development than c-kit and KL, perhaps identifying a physiologically important difference between KL and FL.

In mice lacking functional c-kit expression, T-cell numbers in peripheral blood are normal,257 although a deficiency in fetal thymic development has been reported.258 

One purified c-kit+ BM stem cell can reconstitute the thymus in more than 40% of sublethally irradiated mice, whereas c-kit stem cells have little or no such ability.259 Although the BM population can produce myeloid/erythroid as well as T-cell progeny, thymus-derived c-kit+LinThy-1lo cells appear to be lymphoid-restricted.260 Anti-c-kitantibodies completely block T-cell generation from BM, but not thymic cells, suggesting that T-cell generation from these primitive, lymphoid-committed stem cells in the thymus might not require signaling through c-kit.260 

KL has little or no growth-promoting activity alone, but promotes IL-7-stimulated growth of primitive mouse CD4CD8CD3 thymocytes, but not CD4+CD8+ cells or single CD4+and CD8+ cells.234,261 Anti-c-kitantibodies dramatically inhibit in vitro fetal thymic T-cell production and differentiation from fetal liver progenitor cells.234Similarly, anti-c-kit antibodies reduce cell production and differentiation towards CD4+CD8+ cells in a reconstitution assay with fetal thymocytes into fetal thymus.232 This suggests that KL might be involved in promoting the growth and differentiation of immature thymocytes. IL-3 and IL-12 have been shown to synergize with KL to enhance the growth of primitive, but not more mature, thymocyte populations.235 

T-cell numbers in peripheral blood are normal, but a reduction in early T-cell progenitors is seen postnatally in flt3-deficient mice, and flt3-deficient stem cells are impaired in their ability to reconstitute T cells in the thymus and peripheral blood.227 

FL synergizes with IL-7 to stimulate the proliferation of unfractionated murine thymocytes, and a stimulatory effect can be seen in response to FL in the absence of IL-7.49 The most primitive CD4low thymic progenitor cells capable of generating multiple lymphoid lineages are growth stimulated by FL (in combination with IL-3, IL-6, and IL-7) more efficiently than with KL.262 In contrast, pro-T cells are more efficiently expanded with KL than FL, suggesting that FL might be more active than KL at an earlier stage of T-cell growth.262 In agreement with this, FL appears to preferentially promote self- renewal of CD4low cells in fetal thymic organ culture, whereas KL promotes early T-cell differentiation.262 

Studies of cytokine effects on the regulation of human T-cell development have been difficult due to the lack of appropriate in vitro assays. However, KL enhances thymic stromal cell-supported production of human CD4+ and/or CD8+ cells from CD34+CD4CD8 BM progenitor cells,263 whereas FL promotes IL-12-stimulated T-cell production from human CD34+ BM cells on thymic stromal layers.264 

c-kit is constitutively expressed on a small subset of resting human NK cells in peripheral blood characterized by high CD56 expression, but not on the larger fraction of more differentiated NK cells with low CD56 expression.175 These c-kitreceptors are functional because KL suppresses apoptosis, apparently through induction of bcl-2 expression, although it does not promote proliferation, differentiation, or cytotoxicity on its own.152,175 However, KL in combination with IL-2 promotes the growth, but not cytotoxicity, of this population of resting NK cells.175 

KL enhances stroma-independent NK cell development from human BM progenitor cells stimulated by IL-2, IL-7, or IL-15 in vitro.265-267 An important regulatory role of flt3 and its ligand in NK cell development is supported by the finding that FL-deficient mice treated with poly IC or IL-15 are devoid of NK cell activity in the spleen.248 Furthermore, FL in combination with IL-15 promotes the expansion but not differentiation of CD3CD56+ NK cells from human CD34+ progenitor cells.268 

All DC express CD45 and arise from BM progenitor cells; evidence suggests that DC derive from myeloid and lymphoid progenitor cells.269,270 Myeloid-derived DC can be generated in vitro from progenitor cells isolated from BM, mobilized peripheral blood, or cord blood; GM-CSF appears to play a primary role in promoting their production.269,270 A number of cytokines, including tumor necrosis factor-α (TNF-α), IL-4, and KL, can enhance DC formation induced by GM-CSF.269,270 KL stimulates DC formation from human CD34+ BM and cord blood progenitor cells in combination with GM-CSF and TNF-α without affecting DC differentiation.193-195 

FL increases the production of DC from CD34+ BM progenitor cells in combination with GM-CSF plus TNF plus IL-4.196This enhanced DC production is similar to that observed in response to KL, and when these two cytokines are combined, the effect is additive.196 As with KL, FL does not appear to affect the differentiation, but rather the production, of DC.196Production of DC from mobilized CD34+ peripheral blood progenitor cells (PBPC) by GM-CSF and TNF-α is enhanced by KL and FL individually; combining them results in an additive response.271 

KL or FL (in combination with other cytokines) promotes DC formation from uncommitted thymic precursors,272 but the identity and responsiveness to KL or FL of committed lymphoid-derived CFU-DC remains to be determined.

In vivo treatment of mice with FL results in a dramatic increase in the number of myeloid- and lymphoid-derived functional DC in BM, spleen, thymus, peripheral blood, gastrointestinal lymphoid tissues, and other tissues, indicating an absolute increase in functionally mature DC rather than a redistribution.273 In contrast, administration of KL, GM-CSF, or IL-4 to mice does not expand the number of DC in the spleen. A key role of FL in DC generation is further supported by reduced numbers of DC in FL-deficient mice.248 

Many studies have suggested that most, if not all, pluripotent long-term reconstituting murine stem cells (LTRC; purified by various methods from BM, fetal liver, and the intra-embryonic aorta-gonad-mesonephros) express c-kit.184-188,274-276 Particularly noteworthy was a study in which a single LinSca-1+CD34low/-c-kit+stem cell efficiently long-term reconstituted as much as one of five transplanted mice.277 In addition, cells with the same phenotype isolated from primary recipients were able to reconstitute secondary recipients.277 The corresponding c-kit population was not investigated. Although these studies have clearly established that a large fraction and probably most LTRC are c-kit+, they do not necessarily rule out the possibility of a coexisting, and probably less frequent c-kit LTRC, because the reconstitution assays might not have been optimal for detecting the LTRC activity of a (putative) c-kit stem cell population.

In support of the potential existence of c-kit stem cells, c-kit murine BM cells without detectable c-kit expression but with LTRC, but no short-term reconstitution activity, have been identified.278 One study identified a minor but efficient c-kit LTRC population (0.005% of BM cells).279 The absence of c-kit expression was verified at the cell surface as well as by RT-PCR. As few as 10 of these cells efficiently generated all blood cell lineages for the life span of the mice and showed extensive in vivo self-renewal ability, as assessed through serial transplantation. In contrast, as many as 1,000 of these cells showed no ability to promote radioprotection.279 This is in contrast to most c-kit+ LTRC (with the exception of CD34−/low c-kit+ stem cells277), which in general have been found to also be enriched in short-term reconstituting and radioprotective ability.184-186,188 

The existence of an LTRC population with little or no c-kitexpression is also supported by another study280 in which candidate stem cells were subfractionated into c-kitlow and c-kit<low (no detectable cell surface expression but positive for c-kit mRNA) populations, representing 0.006% and 0.008% of the BM cells, respectively. These two populations did not differ in their capacity to provide donor long-term multilineage reconstitution in primary irradiated recipients. However, when BM from primary recipients was transplanted into secondary recipients, multilineage donor reconstitution could only be obtained from cells whose origin was c-kit<low stem cells.280 Tertiary recipients receiving cells derived from c-kit<lowstem cells were also efficiently reconstituted.280 

Other investigators have subfractionated murine BM progenitor/stem cells based on different levels of c-kit expression. In one study, murine BM stem cells were isolated by counterflow centrifugal elutriation; subsequently fractionated into c-kitneg, c-kitdull, and c-kitbright subpopulations; and administered to unirradiated W/Wvrecipients.187 One hundred c-kitbrightcells were sufficient to repopulate lympho-hematopoiesis inW/Wv recipients, whereas as many as 2.5 × 104 c-kitdull or 5 × 105 c-kitneg cells had no LTRC activity.

Whereas the majority of BM colony-forming cells in normal mice are c-kitbright, most progenitors from 5-FU-treated mice are c-kitdull.281 Cells resistant to 5-FU represent predominantly dormant progenitor cells; moreover, c-kitdull progenitor cells, unlike c-kitbright progenitor cells, require multiple cytokines to be recruited to proliferate and develop in culture into c-kitbright progenitor cells. This suggests that the most primitive murine progenitors might be c-kitdull.281 

The different conclusions reached in these studies might simply reflect that LTRC are heterogeneous with regard to c-kit expression and that differences in purification strategies and reconstitution assays might result in enrichment and detection of different subpopulations of stem cells. For instance, it is possible that the in vitro (cytokine stimulation) and in vivo (5-FU treatment) manipulation of these cells might modulate (up or down) the expression of c-kit. Thus, although a certain level of c-kit expression might prove useful for purification and characterization of LTRC by one specific procedure, it is not necessarily transferable to other methods.

Collectively, these studies suggest that, although most murine LTRC express low or high levels of cell-surface c-kit, they coexist with less frequent subpopulations of LTRC with undetectable c-kit expression. However, cells found to be c-kit by flow cytometry are not necessarily devoid of cell-surface c-kit expression, because the limit of detection of this method is around 500 molecules per cell. In addition, the finding of c-kit mRNA expression using the much more sensitive RT-PCR method might be due to a minor contaminating c-kit+cell population and does not necessarily reflect cell-surface expression of c-kit. Thus, currently it appears most correct to define apparently c-kit stem cells as c-kit<low.280 Because these c-kit<low stem cells appear to represent highly quiescent LTRC, they might exclusively promote late, rather than early, engraftment and have a higher self-renewal capacity than most c-kit+ stem cells, as shown through stringent serial transplantation assays.279,280 The inability of c-kit/c-kit<low murine BM cells to provide long-term reconstitution in other studies might be a direct consequence of such stem cells being present in low numbers and/or not activated when transplanted after standardized myeloablative or nonablative regimens.

In the stem and progenitor cell compartment in mice, the flt3 receptor has been found in LinSca-1+AA4+fetal liver cells,19,166LinSca-1+ BM cells,19,166 and WGA+15-1.1Rh123 bright and dull cells.282 

Virtually all AA4+CD34+ fetal liver cells express c-kit. These, as well as LinSca-1+c-kit+ BM cells, contain distinct flt3+ and flt3subpopulations, and the long-term repopulating activity appears to be predominantly found in the flt3subfraction.45 Thus, most murine LTRC appear to be c-kit+ but flt3/flt3<low. This observation, combined with flt3+ stem cell populations having a lower fraction of cells residing in G0 than flt3 stem cells, has led to the proposal that flt3+ repopulating cells might represent an activated subset of stem cells.45,187 However, note that subpopulations of flt3+ stem cells are quiescent and capable of promoting long-term reconstitution.45Additional long-term serial transplant reconstitution studies using flt3 and flt3+ stem cell populations could provide more definite information regarding the self-renewal capacity of flt3 and flt3+ stem cell populations.

A characteristic of the most primitive hematopoietic progenitor/stem cells is the requirement for simultaneous activation through multiple cytokine receptors to allow recruitment into active cell cycling.2,4 

Based on different patterns of growth-promoting activities on candidate stem cells and their ability to synergistically interact with other factors, cytokines can be grouped into different classes (Table 4). Synergy appears to be most pronounced when cytokines from different classes are combined.2 KL and FL are the only identified members of a distinct group of early acting stem cell factors with unique and potent activities on a variety of candidate murine stem cell populations. Although they have little or no in vitro growth-promoting activity when acting alone, both KL162,197,222,223,281,283-292 and FL45,48,49,166,205,206,223,245,293 can act in combination with most, if not all, other cytokines from the two groups of early acting cytokines to enhance growth of primitive murine progenitor/stem cells through enhanced recruitment of otherwise quiescent progenitor cells and enhanced proliferative activity.

Table 4.

Classification of Early Acting Cytokines

Class Members
I. Stem cell factors  KL, FL 
II.  Colony-stimulating factors  G-CSF, M-CSF, IL-3, TPO  
III.  Purely synergistic factors  IL-1, IL-4, IL-6, IL-11, IL-12, LIF 
Class Members
I. Stem cell factors  KL, FL 
II.  Colony-stimulating factors  G-CSF, M-CSF, IL-3, TPO  
III.  Purely synergistic factors  IL-1, IL-4, IL-6, IL-11, IL-12, LIF 

Classification is based exclusively on the functional ability of various cytokines to promote growth of primitive murine hematopoietic progenitor cells and candidate stem cells in vitro. In principle, this classification holds true for primitive human progenitor cells as well. Recruitment of primitive hematopoietic progenitor/stem cells generally can only occur through combined activation of at least two cytokine receptors, whereas optimal growth usually requires the synergistic interaction between multiple cytokines.2,4 Although usually having little or no growth-promoting activity alone, FL and/or KL can, with few exceptions, synergistically interact with any of the colony-stimulating factors and/or purely synergistic cytokines to enhance growth. Although synergy can occur between cytokines within one class, the most efficient growth-stimulation is obtained by combining cytokines from different classes.2,4 

Several studies involving single-cell cloning and delayed addition of cytokines have shown that the effects of KL and FL are mediated directly on the primitive progenitor cells, ruling out indirect effects mediated by other cells. However, the extent of synergy exhibited by KL and FL, both with regard to recruitment and enhanced proliferation, varies considerably, depending in part on the interacting cytokine(s) and the specific target population investigated. Although the magnitude of synergy a specific cytokine exhibits in combination with KL and FL is likely to result from interactions of the distinct signaling pathways involved, it might also be a reflection of the heterogeneity in expression of other cytokine receptors on primary hematopoietic cell populations.2,4 When directly compared and combined with the same cytokine(s), KL often recruits a slightly higher number of primitive murine myeloid progenitor/stem cells into in vitro proliferation than FL does.45,48,49,166,205,206,223,245,293-297 This occurs independently of which cytokine is used as the synergistic factor. In addition, the average size of the resulting colonies is usually significantly larger in KL- than in FL-supplemented cultures. Finally, the progeny of primitive murine progenitor cells usually remain more undifferentiated in FL- than in KL-supported cultures.166,205,206,245 

As already described in detail, the expression of flt3 appears more confined to primitive progenitor cells than c-kit, which is also highly expressed on various populations of more committed myeloid progenitor cells (Fig 2). Thus, the smaller clone size and less differentiated progeny observed in FL-supplemented cultures could result from the loss of flt3 expression at an earlier stage than c-kit. In addition, c-kit is expressed on a higher percentage of primitive progenitor/stem cells than flt3,45,166 which may explain the lower cloning frequency of primitive murine progenitor cells cultured/supplemented with FL rather than KL.

The activities of FL on primitive murine progenitor cells may overlap and be redundant with those of KL, as suggested for a number of other cytokines with activity on primitive hematopoietic progenitors.2,4 However, although KL and FL have largely overlapping activities, they can also synergize with each other to promote in vitro growth of primitive murine progenitor/stem cells.205,206,245 This synergistic interaction might help to explain why mice with a combined c-kit and flt3 deficiency have a more severe stem cell defect than mice with a single deficiency in c-kit or flt3.227 

Because no routine and optimal reconstitution assay exists for human LTRC, its status with regard to c-kit and flt3 expression has yet to be established. However, much has been learned from studies of candidate human stem cells in various surrogate assays. c-kitis highly expressed in the CD38 subfraction of CD34+ BM cells,190,298 which, although representing only 0.05% to 0.1% of MNC, contains most, if not all, cells capable of long-term multilineage reconstitution of preimmune fetal sheep and immune-deficient mice.299,300 c-kitis also expressed on all cells in a population of purified quiescent human stem cells that is devoid of progenitors responsive to defined cytokines in vitro but highly enriched in long-term culture-initiating cells (LTC-IC).301 Other studies have shown that most, if not all, LTC-IC are c-kit+.189,191 

In one study, CD34+c-kit cells produced no colony-forming cells (CFC), although more CFC were formed by CD34+c-kitlow than CD34+c-kithigh cells after 9 weeks of culture. In addition, c-kithigh cells emerged from c-kitlow cells after 4 weeks of culture.302 

Enrichment of primitive human progenitor cells in the CD34+c-kitlow fraction as compared with the CD34+c-kithigh fraction of BM cells was recently confirmed in long-term engraftment studies in preimmune fetal sheep.303 Although few animals were transplanted in this study, the findings clearly support that CD34+ human BM cells expressing low levels of c-kit are enriched in cells with an ability to provide long-term multilineage reconstitution. In contrast, cells with no or high c-kit expression have less long-term reconstituting ability.303 

Subfractionation of CD34+ cord blood into c-kit, c-kitlow, and c-kithigh populations shows a pattern similar to BM in that c-kitlow cells appear to contain more quiescent and blast cell progenitors.304 

There is no evidence yet for a population of c-kit/c-kit<lowlong-term repopulating human stem cells. However, such a stem cell population is likely to be present at a very low frequency, and current in vivo (and in vitro) reconstitution assays for human cells may be inadequate for detection of such a highly quiescent stem cell population. Therefore, the status of c-kit expression on the earliest human hematopoietic stem cells remains to be elucidated in more detail.

One study has suggested that virtually all BM cells expressing high levels of CD34 and low levels of c-kit are flt3.57 Because the most primitive human stem cells have been suggested to express low levels of c-kitand high levels of CD34,302,303 this finding would suggest that the earliest human stem cells might not express detectable levels of flt3. However, in another recent study,176 most c-kitlow cells as well as CD34+CD38 cells were found to coexpress flt3 at low levels, and primitive cobblestone area-forming cells appeared to be flt3+ as well as flt3. However, the flt3 status of human LTRC remains to be investigated.

Our current knowledge regarding c-kit and flt3 expression on hematopoietic stem cells is summarized in Fig 2. Most long-term reconstituting stem cells identified to date in murine reconstitution assays express c-kit.184-188,274-276 The few studies investigating flt3 expression on LTRC suggest that most are flt3 and that these might be more primitive/quiescent than flt3+ LTRC.45,187 However, further studies will be required to dissect the expression of flt3 on the earliest stem cells.

The existence of c-kit<low LTRC has been shown as well278-280 and, depending on the long-term reconstitution assay and stem cell population used, LTRC may predominantly express high, low, or undetectable levels of c-kit.187,278-281,303 

It is unclear whether such distinct patterns of c-kit and flt3 expression might help identify subpopulations of LTRC within a hematopoietic hierarchy, although available data indicate the existence of such a hierarchy (Fig 2). The most primitive stem cell is likely to be less frequently and more deeply quiescent than stem cells further down in the hierarchy. These characteristics might make it difficult to purify and subsequently activate this stem cell population in standard reconstitution assays, in which more activated stem cells might have a repopulating advantage. Thus, a minor population of c-kit<low (potentially c-kit) stem cells that efficiently and exclusively provides long-term reconstitution and has a high self-renewal potential278-280 is likely to represent a highly quiescent stem cell population. The status of flt3 expression on this stem cell population remains to be determined, but some studies indicate that flt3 is predominantly expressed on activated stem cells45,187; thus, the earliest stem cells might also be flt3. Such c-kit<low/−flt3<low/− stem cells might, upon activation, give rise to long-term repopulating stem cells expressing detectable but low levels of cell-surface c-kit but not flt3.187,281,303 We propose that this stem cell population could next give rise to c-kithighflt3<low stem cells.187,281,302,303 There is also evidence for an activated stem cell population with more restricted long-term repopulating activity that expresses high levels of c-kit as well as flt3.45 

It is important to emphasize that this represents a proposed and simplified stem cell hierarchy, exclusively based on expression of c-kit and flt3 and predominantly based on studies in mice. In addition, the information regarding flt3 expression on LTRC is much more limited than for c-kit (in particular for human stem cells). Furthermore, heterogeneity would be expected within each level of the hierarchy based on variable expression of other, potentially important stem cell molecules. Thus, additional studies will be required to confirm or redefine the proposed stem cell hierarchy.

A similar pattern of growth-promoting activities of KL172,191,199,200,224,226,254,302,304-310 and FL48-50,192,207,208,224,293,311,312 is observed on primitive human hematopoietic progenitor cells, as described above for murine progenitors. When stimulated by KL or FL alone, primitive human progenitor cells isolated from fetal liver, cord blood, or BM show little or no growth response, but both ligands in combination with other early acting cytokines synergistically enhance growth in a direct manner. Whereas multiple studies on different populations of primitive murine progenitor cells have found KL more efficient than FL at recruiting primitive progenitor cells into proliferation, several studies on enriched primitive human progenitor cells indicate that FL is at least as efficient as KL at recruiting human cells.192,207,313-315 FL also appears to be more efficient than KL at maintaining primitive human progenitor cells in a less differentiated state.313-316 Again, this might result from the more restricted expression of flt3 on more committed progenitor cells.

In the mouse, LTRC can be quantified by a competitive repopulation assay; an equivalent assay for human stem cells does not currently exist. Accordingly, the ability of candidate human stem cells to produce committed progenitors over extended periods of culture (minimum of 5 weeks) on established stromal cell layers has been used as a surrogate human stem cell assay, although this should not be considered to represent a true stem cell assay.313,314,317,318 

Murine LTC-IC express c-kit and, although their optimal growth and differentiation in stroma-dependent cultures is enhanced by KL, their formation and maintenance appear to be KL-independent.275,319,320 Furthermore, no difference in KL expression is observed between cell clones capable and incapable of maintaining long-term repopulating cells, and the addition of exogenous KL does not reverse the inability of certain clones to support long-term hematopoiesis.320 Similarly, the ability of several stromal cell lines to conserve long-term marrow repopulating stem cells is unaffected by c-kit blocking antibodies, whereas their ability to promote myelopoiesis is virtually eliminated by the same antibody.275,320 Finally, LTC-IC numbers are only marginally reduced in W mutant mice.319 

Human LTC-IC, like those of mice, express c-kit but do not depend on c-kit activation for survival; but the addition of c-kit blocking antibodies to long-term cultures inhibits production of mature myeloid and erythroid progenitor cells from human stem cells.189,302,321,322 Although Sl/Slfibroblasts are as efficient as normal murine fibroblasts or irradiated human marrow feeder layers at supporting maintenance and clonogenic cell output of LTC-IC, KL in the absence of feeder layers can also efficiently maintain LTC-IC.322 This suggests that KL, although not required, can support these primitive cells. The superior ability of BM stromal cells to promote long-term hematopoiesis compared with umbilical cord vein endothelial cells or human fibroblasts does not appear to be mediated through c-kit, because these stromal cells do not differ in their expression of soluble or membrane-bound KL.323 

Although less is known about the expression and function of flt3 on LTC-IC, several lines of data suggest that LTC-IC (at least in part) express flt3 and that FL, like KL, can enhance their growth and differentiation.17,313,314 Antisense oligonucleotides against flt3 almost completely block the ability of human LTC-IC to form mature myeloid progenitor cells in BM stromal cultures.17 Furthermore, FL on its own has the unique ability to expand human LTC-IC which are reduced in cultures containing KL alone314 and in combination with TPO it maintains LTC-IC over prolonged culture.229 

A critical role in hematopoiesis has been implicated for the very late antigen (VLA) family of integrins.324-328 KL is a potent stimulator of the adhesion of mast cells, hematopoietic progenitor cell lines, and CD34+ BM progenitor cells to fibronectin and vascular cell adhesion molecule-1 (VCAM-1) through activation of VLA-4 and VLA-5.329-332 Only one hundredth of the amount of KL is required to induce adhesion compared with the amount needed to induce proliferation.331 

The ability of KL to promote adhesion may have physiologic and potential clinical significance, because adhesion molecules are thought (1) to be important regulators of anchoring, migration, and mobilization of stem cells; (2) to affect cell growth and differentiation; and (3) to improve gene transfer into candidate hematopoietic stem cells.333-335 

Membrane-bound KL is likely to function in part as an adhesion molecule for mast cells and hematopoietic progenitor cells.336-340The ability of KL to promote adhesion of c-kit+hematopoietic progenitors might explain why progenitor cells exposed to blocking c-kit antibodies show reduced homing efficiency.341 The effect of KL on homing and migration might also result from its chemotactic effect on mast cells and hematopoietic progenitor cells.342-344 Studies have not yet been performed to determine whether FL has a similar ability as KL to promote adhesion of hematopoietic cells.

Although the primary function of KL and FL in early hematopoiesis might be to induce the growth of quiescent progenitor/stem cells through synergistic interactions with other early acting cytokines, there is also ample evidence that KL345-350 and FL,166,311,351,352 in the absence of other cytokines, selectively promote viability rather than proliferation of primitive murine and human progenitor cells, including the LTRC in the case of KL.345,347,348 

Although the physiologic significance of growth inhibitory cytokines in steady-state hematopoiesis remains to be established, the interactions of transforming growth factor-β (TGF-β) and tumor necrosis factor-α (TNF-α) with KL and FL on primitive hematopoietic progenitor cells are worth mentioning. TGF-β, a potent inhibitor of primitive hematopoietic progenitor cell growth,353 hinders the viability and growth-stimulatory effects of KL and FL on primitive murine and human hematopoietic progenitor cells.224,295,351,354-356 TNF-α, a cytokine that can directly stimulate or inhibit the growth of primitive and committed hematopoietic progenitor cells,357 inhibits KL- and FL-stimulated growth, viability, and expansion of normal primitive murine and human progenitor cells.296,314,358-360 

As described above, KL and FL are produced in membrane-bound as well as in soluble forms. In addition to potentially functioning as adhesion molecules by binding to their respective receptors, membrane-bound KL has activities distinct from those of soluble KL.Sl/Sld mutant mice that only produce the secreted form of KL have the same hematopoietic defects characteristic of Sl/Sl mutant mice, suggesting that there is an essential role for membrane-bound KL.88,92 When cDNAs encoding soluble or membrane-bound isoforms of human KL are transfected into stromal cells derived from Sl/Sl mice, membrane-bound KL maintains human hematopoiesis longer than secreted KL.89Membrane-bound KL (or immobilized anti-kit antibodies), when compared with soluble KL, induces (1) more c-kit kinase activity, (2) less rapid downregulation of cell surface c-kitexpression, and (3) enhanced stability of c-kit.361,362 Thus, the difference in activity between soluble and membrane-bound KL might result from the soluble c-kit/KL complex being rapidly internalized and degraded, resulting in transient tyrosine kinase activation of c-kit. In contrast, if the membrane-bound c-kit/KL complex is not internalized and degraded, it could result in a sustained period of enhanced c-kit kinase activity.

Mutations in the W or Sl loci result in reductions of various primitive hematopoietic progenitor cells,10 but except for erythrocytes, the numbers of other mature blood cells appear normal under steady state conditions. Sl/Sld mice, although severely anemic, survive to adulthood; administration of KL improves their anemia, which reappears when KL treatment is discontinued.36 KL treatment also increases their platelets, granulocytes, monocytes, and lymphocytes above the levels seen in wild-type mice36 and increases CFU-S numbers in their BM and spleen.345 

Sl/Sld mice display a dysfunctional regulation of platelet production in response to cytotoxin-induced thrombocytopenia; they do not undergo the rebound thrombocytosis observed in wild-type mice after 5-FU treatment.167 However,Sl/Sld mice treated with 5-FU have a rebound thrombocytotic response after the administration of KL.167Enhanced KL mRNA expression in response to 5-FU-induced thrombocytopenia in the BM of normal mice and c-kit expression on immature megakaryocytes further substantiate the role KL plays in promoting platelet recovery after BM suppression.167 KL also increases the number of megakaryocytes and platelets in normal mice.167 

The role of KL in promoting platelet production after hematopoietic injury might be due to its ability to synergize with TPO to enhance megakaryocyte progenitor cell growth.217 Although TPO is the primary regulator of megakaryocytopoiesis and platelet production,217,363 mice deficient in TPO or c-mpl(the TPO receptor) expression do produce functionally mature platelets, albeit at dramatically reduced levels.363 In addition, KL administration to TPO-deficient mice increases platelet counts.364 Thus, it appears that there are TPO-independent mechanisms for platelet production in which KL might also play a role.

Sl/Sl mice lacking functional KL die at day 15 or 16 of gestation.29 However, the total number of fetal liver cells in normal or Sl/Sl mice increase by more than 10-fold between day 13 and 15 of gestation and, although the fetal liver cellularity in the KL-deficient mice is only 20% to 25% of wild-type fetal liver, the increase in fetal liver cells is similar.186 More importantly, the number of cells with a stem cell phenotype (LinSca-1+Thy-1lo) and CFU-S activity also increases in Sl/Sl mice from day 13 to 15.186 This suggests that KL might not be essential for early hematopoietic development in mouse embryos and that fetal hematopoietic progenitor/stem cells can expand/self-renew in the absence of KL.

In mice with viable W mutations, disruption of hematopoiesis appears largely restricted to erythropoiesis and mast cell generation. Specifically, in BM of W41/W41mice (with a partial c-kit signaling deficiency), the number of erythroid, myeloid, pre-B, and multipotent progenitor cells, as well as LinSca-1+ candidate stem cells and LTC-IC, are at near-normal levels.319 However, long-term repopulating units in W41/W41BM are reduced 17-fold.319 Furthermore,W41/W41 fetal liver cells are qualitatively and quantitatively close to normal in their short-term reconstituting ability but promote less long-term reconstitution.365,W42 mutant fetal liver cells (completely silent c-kit receptor) show an even more pronounced inability to provide long-term reconstitution. Thus, although c-kit/KL interaction might not be critical for stem cell generation and expansion during early ontogeny, their sustained self-renewal might in fact be KL-dependent. An important role for KL in promoting reconstitution by LTRC is also supported by enhanced expression of KL following myeloablative treatment167,366and the ability of endogenous and exogenous KL to promote survival and hematopoietic reconstitution of mice and dogs after myeloablation.366-370 

Other findings indicate that KL plays an important role in steady-state adult hematopoiesis. As early as 2 days after injection of normal mice with c-kit antibodies, most myeloid and erythroid cells disappear, although the BM cellularity remains normal.183The content of in vitro clonogenic myeloid progenitor cells and CFU-S in the BM declines rapidly, whereas a concomitant increase in B-cell precursors is observed.183 

KL administration in vivo to normal mice results in an increase in peripheral white blood cells (WBC), predominantly neutrophilic granulocytes, and also a slight increase in lymphocytes.371BM cellularity is not affected, and its content of in vitro clonogenic myeloid progenitor cells and day-8 CFU-S is only slightly enhanced.371 In contrast, the number of myeloid progenitors and CFU-S in the spleen increases dramatically, and KL induces a more rapid and pronounced leukocytosis in splenectomized mice.371 

KL administration to mice for 7 days results in depletion of candidate BM stem cells (LinSca-1+Thylo) and a corresponding reduction in radioprotective ability.372 A concomitant increase in both these hematopoietic parameters, as well as multilineage long-term reconstituting activity, is observed in spleen and peripheral blood.372 Because the total number of LinSca-1+Thylo did not significantly change, it was postulated that administration of KL does not result in a net expansion of long-term reconstituting stem cells, but rather redistributes existing stem cell activity to peripheral sites.

The progenitor/stem cell mobilizing ability of KL has been investigated extensively in various animal models. Low doses (25 μg/kg/d) of KL have little or no effect on the number of PBPC in splenectomized mice, but KL synergistically enhances WBC counts and mobilization of PBPC in combination with an optimal dose of G-CSF (200 μg/kg/d).373 The increase includes cells with both short-term and long-term repopulating activity.374Administration of KL to normal mice results in a threefold increase in LTRC that are predominantly redistributed to peripheral blood and the spleen.375 KL in combination with G-CSF also mobilizes progenitor/stem cells to the blood that are capable of engrafting lethally irradiated dogs and baboons.376-379 Although the ability of KL plus G-CSF–mobilized progenitor cells to long-term engraft baboons and dogs remains to be established, it appears that blood count recovery occurs earlier with grafts mobilized with KL plus G-CSF than with G-CSF alone.376-378 

In humans, daily administration of KL at dosages of up to 50 μg/kg for 14 days does not increase the number of peripheral blood CD34+ cells, but does increase the absolute number of CD34+ cells and assayable primitive and committed myeloid progenitor cells in BM.380 In a phase I/II study in patients with high-risk breast cancer, mobilization of progenitor cells to peripheral blood by KL plus G-CSF was superior to G-CSF alone.381 

The administration of KL plus G-CSF in mice has shown interesting kinetic aspects of distribution/expansion of stem cells.382The most dramatic increase in repopulating ability of peripheral blood stem cells is observed immediately after cytokine treatment, concomitant with a reduction in reconstituting ability of the BM. Subsequently, the repopulating activity of peripheral blood stem cells declines to normal levels within 6 weeks of termination of cytokine treatment, whereas the repopulating activity of BM cells increases by day 14 to levels 10-fold higher than BM cells from untreated mice. The mechanism for this large yet temporary increase in the repopulating activity of BM stem cells after administration of KL and G-CSF is unclear. Increased numbers of primitive (CD34+CD38) cells are also seen in the BM of rhesus monkeys as long as 2 to 3 weeks after administration of KL and G-CSF.383 

In vivo daily administration of recombinant human FL (500 μg/kg/d) to normal mice stimulates an increase in WBC.384 The increase in WBC counts is reflected in an increase in the number of lymphocytes, granulocytes, and especially monocytes.384 A small decrease in hematocrit after 10 days of treatment is reversed upon cessation of treatment. BM cellularity is not affected by FL treatment. The number of CD4+ and CD8+ T cells in the BM is reduced, as are mature (B220+IgM+) B cells.384 In contrast, FL treatment increases the number of immature (B220+IgM) B cells. The number of monocytes and granulocytes increases as well, as do DC, whereas the number of immature erythroid cells is reduced by 90%.384This decrease may result from the mobilization of erythroid precursors from BM and/or an altered differentiation pathway for progenitors of these erythroid precursors; the exact cause is not known.

Splenic cellularity increases after 10 days of FL treatment, with little effect on CD4+ and CD8+ T cells, but with an increase in NK cells and DC. Most striking is the ninefold increase in B220+IgM B-cell progenitors, with only a marginal effect on splenic mature B220+IgM+ B cells. As in BM, the number of splenic myeloid cells increases as much as 10-fold. Splenic primitive erythroid cells also increase, although these cells decrease in BM.384 

The number of BM GM progenitor cells increases fivefold after 3 days of FL treatment. The number of these cells subsequently decline during the next 12 days of treatment, and decrease to 50% below control levels 1 week after cessation of FL treatment.384 BFU-E numbers in BM increase slightly after 3 days of FL treatment, but decrease subsequently. Colony-forming unit granulocyte, erythrocyte, monocyte, megakaryocyte (CFU-GEMM) numbers also peak early in BM and subsequently return to control values. CFU-GM, BFU-E, and CFU-GEMM increase 123-fold, ninefold, and 108-fold, respectively, in spleen. Maximum levels are seen after 8 to 10 days of treatment, and these numbers return to control levels 1 week after treatment. In peripheral blood, a 537-fold, 113-fold, and 585-fold increase in CFU-GM, BFU-E, and CFU-GEMM, respectively, is observed after 10 days of FL treatment.384 FL also mobilizes primitive, day-13 CFU-S into peripheral blood. Finally, an increase in cells with a stem cell phenotype (LinSca-1+kit+) is observed in the BM, spleen, and peripheral blood of FL-treated mice.384 

Cells mobilized to peripheral blood with FL have been shown to have long-term (6 months) reconstituting ability.385 FL also mobilizes progenitor/stem cells into the peripheral blood of nonhuman primates and shows synergy with either G-CSF or GM-CSF with regard to mobilizing ability.385,386 

Preliminary results from human clinical trials show that the administration of FL to normal, healthy volunteers is safe and effectively elevates the numbers of CD34+ cells and DC in peripheral blood (Mel Lebsack and Eugene Maraskovsky, Immunex; personal communication). The in vivo hematologic/hematopoietic effects of FL and KL are summarized in Table 5.

Table 5.

In Vivo Hematopoietic Effects of KL and FL

Cell Type Response KL FL
LTRC  Expansion  +  
 Mobilization  +  
Primitive/committed progenitors  Expansion Mobilization + +  + +  
Red blood cells  Reticulocytes  +/NE ND  
 Hematocrit  +/NE  Reduced  
Platelets Megakaryocytes  +/NE  ND  
 Platelets  +/NE  ND 
White blood cells  Total number  +  +  
 Granulocytes  +  +  
 Monocytes  +/NE  +  
 Lymphocytes  +/NE  +  
Mast cells  Number  +  NE 
 Activation  +  NE  
Dendritic cells  Number  NE 
Cell Type Response KL FL
LTRC  Expansion  +  
 Mobilization  +  
Primitive/committed progenitors  Expansion Mobilization + +  + +  
Red blood cells  Reticulocytes  +/NE ND  
 Hematocrit  +/NE  Reduced  
Platelets Megakaryocytes  +/NE  ND  
 Platelets  +/NE  ND 
White blood cells  Total number  +  +  
 Granulocytes  +  +  
 Monocytes  +/NE  +  
 Lymphocytes  +/NE  +  
Mast cells  Number  +  NE 
 Activation  +  NE  
Dendritic cells  Number  NE 

The table is based on the effects of in vivo administration of KL or FL as single agents to normal subjects. The effects of KL are based on studies in mouse, rat, dogs, nonhuman primates, and humans, whereas the effects of FL are predominantly based on studies in mouse and nonhuman primates.

Abbreviations: +, increase; NE, no effect; +/NE, effect found in some but not all species investigated; ND, not determined.

Whether flt3 or FL are required for normal hematopoiesis has been addressed by creating mice that carry a homozygous deletion of most of the gene encoding the flt3 receptor227 or FL.248 Mice in which either the flt3 receptor or ligand have been knocked out are generally healthy, which is in marked contrast to the lethality observed in mice homozygous for the deletion of the gene encoding the c-kit receptor or KL protein.24 The flt3 knockout mice have normal levels of peripheral blood cells.227 However, the loss of a functional flt3 receptor results in a reduced number of early B-cell precursors and a defect in primitive stem cells, as measured in a long-term competitive repopulation assay. Upon adoptive transfer to irradiated secondary recipients, stem cells from flt3 deficient−/− mice have an impaired ability to repopulate myeloid, T-, and B-lymphoid lineages.

Mice bearing targeted disruptions in the flt3 receptor were bred with mice carrying mutations in the c-kit receptor to generate animals of the genotype flt3/flt3W/Wv. Offspring had severely reduced numbers of hematopoietic cells and died between 20 and 50 days of age.227 These experiments demonstrated a requirement for both flt3 and c-kit receptors in the development of a normal, functional hematopoietic system.

There is no evidence that FL binds to any other protein in addition to the flt3 receptor. Similarly, no other ligands are known that bind to the flt3 receptor. Thus, one would predict that mice homozygous for a targeted disruption of the FL gene would have an identical phenotype to flt3 receptor knockout mice. FL knockout mice, like the flt3 receptor knockout mice, have a normal, healthy appearance.248 They have a defect in early B-cell development, as do the flt3 receptor knockout mice. However, a couple of significant observations have been made in analyzing the FL knockout mice that were not reported with the flt3 receptor knockout mice. There is a significant reduction in the cellularity in the peripheral blood, spleen, and BM of FL knockout mice, whereas no change in cellularity was reported in the flt3 receptor knockout mice. DC in the spleens of these animals are also significantly reduced. Most notable is a lack of NK cell activity in the spleens of mice treated with either poly IC or IL-15. It is unclear if these unique observations in the FL knockout mice reflect a truly different phenotype or whether strain variations or the depth of analysis account for the observed differences.

Levels of KL in human serum from normal individuals are usually found in the range of 2 to 5 ng/mL.387 KL serum levels have also been examined in a wide variety of patients with hematopoietic disorders, and they do not vary much or appear to be of clinical significance.388 

In contrast to the relatively high levels seen with KL, serum levels of FL in normal individuals average less than 100 pg/mL, which is the limit of detection of the enzyme-linked immunosorbent assay.389 FL levels are not elevated in a variety of anemias that predominantly affect only the erythroid lineage389 or in patients with rheumatoid arthritis, systemic lupus erythematosus, AML, ALL, or human immunodeficiency virus (Lyman et al, unpublished observations).

In contrast, serum levels of FL are highly elevated in patients with hematopoietic disorders that specifically affect the stem cell compartment. Thus, a majority of patients with anemias affecting multiple hematopoietic lineages (eg, Fanconi anemia, acquired aplastic anemia) have highly elevated levels of FL (up to 10 ng/mL).389 Cancer patients treated with chemotherapy and/or radiation also have highly elevated levels of FL.390 

The simplest interpretation of these data is that the loss of functional stem/progenitor cells leads to the loss of a negative regulator of FL production made by the stem/progenitor cells. FL concentrations in blood then become elevated (to a physiologically relevant level) as part of a compensatory hematopoietic response to drive the proliferation of the remaining stem/progenitor cells.

Serum levels of FL returned to normal in a Fanconi anemia patient after a cord blood transplant that cured the pancytopenia.389Similarly, successful treatment of acquired aplastic anemia patients with either BM transplants or immunosuppressive therapy also led to a return to normal of FL serum levels.390 These data suggest that restoration of stem cells in these patients is associated with a return of FL serum levels to those measured in normal, healthy individuals and that FL serum levels may be a surrogate marker for stem cell activity or content in BM.

However, the hypothesis cited above does not explain why about 50% of patients with refractory anemia (RA) have elevated levels of FL,391 because RA is not considered a disease of either stem cell number or activity. FL serum levels are not elevated in any of the other FAB subclasses of myelodysplasia,391 and the reason only some RA patients have elevated serum levels is unknown.

Because both KL and FL have potent effects on primitive hematopoietic cells, the majority of clinical uses envisioned are designed to exploit this activity (Table 6). Both proteins synergize with a wide range of cytokines, and it is possible that they could enhance the effects of other cytokines that function on primitive as well as more differentiated hematopoietic cells.

Table 6.

Some Potential Clinical Uses of KL and FL

Comments
Likely applications 
 Ex vivo expansion/purging of progenitor/stem cell grafts  In combination with other early acting (stem cells) and lineage-selective cytokines (progenitors) to improve reconstitution and to purge tumor-contaminated progenitor/stem cell grafts. 
 Progenitor/stem cell mobilization  In combinations with other cytokines (GM-CSF, G-CSF, TPO, or others) to improve mobilization of progenitor/stem cells to peripheral blood to be used in transplantation.  
 Gene therapy  (1) In combination with other early acting cytokines to improve gene transfer to stem cells in vitro. (2) Mobilize/expand stem cells in vivo (see above) that might prove better targets for gene transfer.  
 Immunotherapy (1) Ex vivo (KL and FL) and in vivo (only FL) expansion of DC for use as vaccine adjuvant. (2) In vivo antitumor activity of FL (via effects on DC and NK cells).  
Additional potential applications 
 Stem cell deficiencies  Potential diseases include aplastic anemia and myelodysplastic syndromes. 
 Pure erythroid aplasia (Diamond-Blackfan anemia)  KL might prove more efficient than FL due to the wide expression of c-kit and lack of flt3 on primitive erythroid progenitors. 
 Cytopenias after chemotherapy/bone marrow transplantation G-CSF/GM-CSF are efficient at promoting neutrophil recovery, and TPO may prove efficient at enhancing platelet recovery. However, KL and FL might, in combination with G-CSF and/or TPO, be of benefit when primitive progenitor/stem cells are severely compromised. 
 Immunodeficiencies (HIV)  Adjuvant treatment of cytopenias. 
Comments
Likely applications 
 Ex vivo expansion/purging of progenitor/stem cell grafts  In combination with other early acting (stem cells) and lineage-selective cytokines (progenitors) to improve reconstitution and to purge tumor-contaminated progenitor/stem cell grafts. 
 Progenitor/stem cell mobilization  In combinations with other cytokines (GM-CSF, G-CSF, TPO, or others) to improve mobilization of progenitor/stem cells to peripheral blood to be used in transplantation.  
 Gene therapy  (1) In combination with other early acting cytokines to improve gene transfer to stem cells in vitro. (2) Mobilize/expand stem cells in vivo (see above) that might prove better targets for gene transfer.  
 Immunotherapy (1) Ex vivo (KL and FL) and in vivo (only FL) expansion of DC for use as vaccine adjuvant. (2) In vivo antitumor activity of FL (via effects on DC and NK cells).  
Additional potential applications 
 Stem cell deficiencies  Potential diseases include aplastic anemia and myelodysplastic syndromes. 
 Pure erythroid aplasia (Diamond-Blackfan anemia)  KL might prove more efficient than FL due to the wide expression of c-kit and lack of flt3 on primitive erythroid progenitors. 
 Cytopenias after chemotherapy/bone marrow transplantation G-CSF/GM-CSF are efficient at promoting neutrophil recovery, and TPO may prove efficient at enhancing platelet recovery. However, KL and FL might, in combination with G-CSF and/or TPO, be of benefit when primitive progenitor/stem cells are severely compromised. 
 Immunodeficiencies (HIV)  Adjuvant treatment of cytopenias. 

Adverse events associated with KL administration in humans in phase I and phase II trials have been primarily dermatologic reactions (eg, pruitic wheals with erythema at the site of injection) and, more rarely, multisymptom systemic anaphalactoid reactions.8,179,181,182 The most likely cause of these effects is mast cell hyperplasia, activation, and mediator release; as a result, prophylactic antihistamine treatment has been incorporated into clinical protocols.8 

Limited data on the hematologic effects of FL in humans have been reported392 and indicate that FL appears to have a good safety profile. This is consistent with the observation that no overt toxicities were seen when short courses of FL were administered to animals in vivo.384,386,393 

Stem cell mobilization.

As described above, KL and FL may prove useful for mobilizing or expanding BM stem cells in vivo. These stem cells can be used in various transplantation settings, in particular autologous and allogeneic stem cell transplantation of cancer patients after high-dose chemotherapy. In addition, mobilized stem cells might be excellent targets for gene therapy383,394-397 (see below). The use of KL and/or FL along with a second cytokine, such as G-CSF or GM-CSF, appears to increase the number of stem cells mobilized (see above). Stem cells mobilized/expanded in vivo by KL plus G-CSF might be better targets for gene therapy than those mobilized with G-CSF alone.366,374,382,383,394 However, qualitative differences in stem cell populations mobilized by different cytokine treatments have not yet been examined in sufficient detail and therefore require further study.

Ex vivo stem/progenitor cell expansion.

Ex vivo expansion of hematopoietic progenitor/stem cells is an area of intense study due to its clinical potential. However, a number of obstacles must be overcome before it can be established whether or not ex vivo-expanded progenitor/stem cells represent an improved therapeutic modality in various settings (for detailed reviews see Williams,398 Lange et al,399 and Emerson400).

Ex vivo–expanded progenitor/stem cells could reduce the need for extensive BM harvests or leukaphereses and enable repetitive cycles of high-dose chemotherapy. Because contaminating tumor cells in autologous stem/progenitor cell grafts can contribute to relapse,401,402 selective ex vivo expansion of progenitor/stem cells may also reduce or eliminate such tumor cells.399,400 

Murine in vitro clonogenic progenitor cells as well as CFU-S efficiently expand when stimulated by KL or FL in combination with cytokines such as IL-1, IL-3, IL-6, IL-11, TPO, and G-CSF.205,206,222,287,345,403 Importantly, KL in combination with IL-1, IL-6, or IL-11 promotes efficient expansion of murine (short-term repopulating) progenitor cells without loss of long-term reconstituting ability in the expanded graft.403-406 

Because IL-3 has been used extensively in ex vivo expansion protocols, it is noteworthy that IL-3 appears to compromise the long-term reconstituting ability of murine grafts expanded in either KL or FL in combination with other early acting cytokines.404,407 

Optimal expansion of human progenitor cells requires the interaction of KL with multiple cytokines, including IL-1, IL-3, IL-6, GM-CSF, G-CSF, and EPO.306-308,408-410 As discussed above, the membrane-bound form of KL is more efficient than the soluble form at maintaining progenitor cell production in stromal cell cultures,89 indicating that membrane-bound KL might be beneficial for maintaining primitive progenitor/stem cells. FL also expands human myeloid progenitor cells in combination with other cytokines.192,208,224,297,311,313,315,316,411 

Although KL and FL are efficient at stimulating production of multipotent and lineage-restricted myeloid progenitor cells from candidate human stem cells, the key question of whether ex vivo expansion protocols for human progenitor/stem cells maintain sufficient pluripotent long-term repopulating stem cells remains unanswered. Currently in patients receiving high-dose chemotherapy, the predominant function of progenitor/stem cell grafts might be to provide efficient short-term reconstitution, whereas long-term reconstitution might be provided equally well by endogenous stem cells surviving the high-dose treatment. However, if high-dose chemotherapy is further intensified, it might become crucial to ensure that transplants also contain sufficient LTRC.398-400 In the case of gene therapy, in which the ultimate goal is the introduction of therapeutic genes into LTRC, it is already paramount that such grafts contain LTRC412 (see below). Thus, it will be important to investigate the effects in ex vivo-expansion cultures on the earliest human stem cells using techniques such as gene marking.413 

Although not conclusive with regard to LTRC, some recent studies cast light on the ability of FL and KL to maintain/expand candidate human stem cells. In one study, FL alone had the unique ability to slightly expand the number of primitive LTC-IC in CD34+CD38 BM cells, whereas LTC-IC were depleted in cultures containing KL alone.314 Furthermore, in a detailed study of 16 different cytokines, a combination of FL, KL, and IL-3 was both necessary and sufficient to obtain a 30-fold expansion of 6-week LTC-IC.314 In other studies, FL and KL were found to be equally efficient at stimulating the production of progenitor cells for 30 days from CD34+CD38progenitor cells cultured on stroma,313 whereas progenitor cell output beyond 56 days was significantly higher in FL- than in KL-supplemented cultures.313 In addition, human CD34+ BM cells expanded under stroma-free conditions in KL plus IL-3 plus IL-6 in the presence (but not in the absence) of FL provided long-term reconstitution of immune-deficient mice.316 Other groups have found FL more efficient than KL at expanding human LTC-IC.414 Another promising combination of factors for the ex vivo expansion of stem/progenitor cells from cord blood was the combination of FL and TPO, which allowed continuous expansion of these cells for as much as 5 months.229 

Gene therapy.

Hematopoietic stem cells are considered optimal targets for gene therapy, because they display extensive capacity to self-renew and to produce large numbers of progeny that are widely distributed throughout the body. In addition, stem cells can be readily obtained from BM, mobilized peripheral blood, or cord blood and can therefore be easily manipulated in vitro.412,415,416 

Gene transfer into mouse long-term repopulating stem cells can be performed with high efficiency and success.417-421 In contrast, gene transfer into stem cells in larger animal models (including studies in humans) has been disappointing.412,415,416 

Currently, mouse retroviruses are the only vectors shown to integrate permanently into host DNA, and most gene therapy protocols targeting stem cells use these vectors. One of the caveats with such retroviruses is that they cannot efficiently transduce and integrate into quiescent cells.412,415,416 Therefore, stem cells that normally are highly quiescent must be recruited into active cell cycle to enable successful transduction with such vectors, and FL and KL may be of use through their ability to efficiently trigger cell cycling of candidate stem cells. In addition, it is possible that these early acting cytokines might have a more beneficial effect on preserving the self-renewal, pluripotentiality, and engrafting potential of targeted stem cells than later-acting cytokines.

KL in combination with IL-3 and IL-6 efficiently promotes transduction of mouse stem cells while maintaining their long-term reconstituting ability.419,421 KL plus IL-3 plus IL-6 is also the combination predominantly used to achieve retroviral transduction of human hematopoietic progenitor cells, resulting in high gene transfer efficiency to committed as well as more primitive human progenitor cells (LTC-IC).422-426 

Recent studies suggest that FL might be more efficient than KL at promoting gene transfer into human hematopoietic progenitor cells. Specifically, when combined with IL-3, FL is superior to KL at promoting retroviral gene transfer to committed myeloid progenitor cells, and the addition of KL (and other cytokines) to FL plus IL-3 significantly reduces the gene transfer efficiency.315 In the absence of stroma or fibronectin, the combination of IL-3, IL-6, and KL is unable to preserve the capacity of retrovirally transduced human BM CD34+ progenitor cells to sustain long-term hematopoiesis in immune-deficient mice in vivo.316 However, when FL is added to this cytokine combination, the transfected cells support long-term reconstitution of immunodeficient mice,316 although FL cannot fully replace the effect of stromal cells.316 The ability of FL to preserve the capacity of putative human stem cells to sustain long-term hematopoiesis in immune-deficient mice does not necessarily imply that FL enhances gene transfer to long-term repopulating stem cells. It is also possible that FL might have a positive effect on the self-renewal and/or engrafting potential of these cells.

KL and FL might also be used to enhance gene transfer into hematopoietic stem cells through their ability to mobilize stem cells to peripheral sites (described in detail above). Long-term reconstituting mouse stem cells mobilized to peripheral sites in response to administration of KL alone can be as efficiently transduced with retroviral vectors as mice treated with 5-FU.375 In mice treated with a combination of G-CSF and KL, mobilized long-term repopulating stem cells are expanded and transduced 2 to 3 times as efficiently as BM from 5-FU-treated mice, making such cells particularly attractive for gene therapy applications.394 

The number of LTRC in the BM of mice and rhesus monkeys is expanded and shows improved gene transfer 1 to 2 weeks after treatment with KL and G-CSF.383 Similar studies of the efficiency of retroviral gene transfer to stem cells mobilized by FL in combination with G-CSF in primates also show an increased efficiency of gene transfer (Harry Malech, NIH, Bethesda, MD; personal communication).

Efficient gene transfer of human c-kit+hematopoietic cell lines has been achieved through targeting of c-kit with a molecular conjugate vector coupled to KL.427 However, whether a similar approach will be successful in normal hematopoietic progenitor/stem cells and whether permanent gene expression can be achieved remains unanswered.

Although these studies imply a role for KL and/or FL in human gene therapy in hematopoietic stem cells, most of these findings have been made in vitro or in immune-deficient mice and do not necessarily reflect true human stem cells. Thus, reproduction of such findings in nonhuman primates and eventually humans is essential.

Immunotherapy.

Immune DC, which may be thought of as professional antigen-presenting cells, have been proposed as cellular vectors for either antitumor or infectious disease vaccines, or as inducers of transplantation tolerance.428-430 The feasibility of using DC as immunotherapy vectors in the clinic has been limited by the small number of DC that can be isolated from the peripheral blood of normal individuals.

Although both KL193,194,431 and FL196,271stimulate the production of DC in vitro (see above), to date only FL has been shown to stimulate DC generation in vivo.273 These DC appear to be both myeloid and lymphoid derived.273Therefore, FL could possibly be used as a vaccine adjuvant: DC subsets would be expanded in vivo by treating individuals with FL, and then antigen-based vaccines would be injected. The goal would be to enhance the magnitude and quality of the immune response generated without the need for chemical adjuvants. Alternatively, larger numbers of circulating DC from FL-treated individuals could be isolated via apheresis for ex vivo manipulation (eg, vaccine or tolerogen exposure), followed by reinfusion of these DC.

Finally, and perhaps most promising, FL may have antitumor effects in vivo that are immune-system mediated. FL administration to mice has been shown to inhibit the growth of a fibrosarcoma cell line in vivo in a dose-dependent manner.432 Administration of FL to mice injected with a breast cancer cell line leads to rejection of these cells in syngeneic mice,433 as does ectopic expression of FL by these breast cancer cells.434 FL may stimulate DC production, which in turn presents tumor antigen(s) to T cells, leading to rejection of the tumors. NK cells are also likely to have a role in this process.

KL and FL, acting through their respective tyrosine kinase receptors c-kit and flt3, have pleiotropic and potent effects on hematopoiesis in vitro and in vivo. Based on studies of the expression and function of the two receptors, it is now evident that the hematologic actions of these two cytokines are predominantly restricted to the progenitor/stem cell compartment. One important exception is the functional expression of c-kit, but not flt3, on mast cells, which helps explain the adverse events associated with KL administration in humans. The physiologic importance (if any) of the residual expression of c-kit and flt3 on other mature cell types remains unknown.

In the (long-term reconstituting) stem cell compartment, c-kitappears to be expressed on more stem cells than flt3, and, although not yet conclusively documented, c-kit might be expressed on earlier stem cells than flt3. Although recent data suggest that the earliest stem cells might express no or very low levels of c-kit and flt3, the status of c-kit and flt3 expression and function on hematopoietic stem cells needs to be studied in more depth, particularly in the human system.

Most of the hematopoietic activities of KL and FL appear to require a synergistic interaction with other early acting or lineage-selective cytokines. c-kit/KL might be critical for maintenance and self-renewal of long-term reconstituting stem cells, particularly in adult hematopoiesis. In addition, these two ligands appear to be essential for optimal production of mature hematopoietic cells from stem cells. Accordingly, stem cells deficient in c-kit or flt3 expression are defective in their ability to reconstitute hematopoiesis in myeloablated animals.

Interestingly, FL appears more critical for generation of lymphoid progeny than KL. In contrast, multiple lines of data suggest that KL inhibits B-cell development in mice.

The finding that FL plays a less crucial role than KL in the regulation of myelopoiesis and erythropoiesis is not surprising, because flt3 is generally expressed on less myeloid progenitor cells and is not found on erythroid progenitor cells. Thus, both KL and FL appear to have a dual function in hematopoiesis in that they both have activity on stem cells and appear to act as critical early regulators of myelopoiesis/erythropoiesis and lymphopoiesis, respectively.

The activities of FL and KL are distinct, although in some instances they may be complimentary to, synergistic with, or antagonistic to each other. It will be important to further dissect the distinct biological activities of the membrane-bound and soluble forms of KL and to determine whether membrane-bound FL functions differently from soluble FL. Whether these key hematopoietic regulators are involved in diseases or potentially could be used therapeutically remains to be further investigated. In that regard, combination therapy with other cytokines will be of particular interest.

The authors acknowledge the extensive and important contributions of colleagues at Immunex, especially Hilary McKenna, Ken Brasel, and Eugene Maraskovsky, and also Doug Williams, Bali Pulendran, Subhashini Srinivasan, Claudia Jochheim, and Dave Lynch for thoughtful discussions and reviewing the manuscript. We also thank members of the Stem Cell Laboratory, University of Lund including Ole Johan Borge, Veslemøy Ramsfjell, Cui Li, and Ole Peter Veiby for valuable input and reviewing the manuscript. We thank Hal Broxmeyer, Hans Drexler, Stefan Karlsson, Jonathan R. Keller, Makio Ogawa, Francis W. Ruscetti, and Alexandra Wodnar-Filipowicz for their critical review of the manuscript. Finally, we thank Anne Bannister and Christine Jones for expert editorial assistance.

Address reprint requests to Stewart D. Lyman, PhD, Department of Molecular Genetics, Immunex Corp, 51 University St, Seattle, WA 98101; or Sten Eirik W. Jacobsen, MD, PhD, Stem Cell Laboratory, Department of Internal Medicine, University Hospital of Lund, S-221 85 Lund, Sweden.

1
Spangrude
GJ
Smith
L
Uchida
N
Ikuta
K
Heimfeld
S
Friedman
J
Weissman
IL
Mouse hematopoietic stem cells.
Blood
78
1991
1395
2
Ogawa
M
Differentiation and proliferation of hematopoietic stem cells.
Blood
81
1993
2844
3
Moore
MA
Review: Stratton Lecture 1990. Clinical implications of positive and negative hematopoietic stem cell regulators.
Blood
78
1991
1
4
Metcalf
D
Hematopoietic regulators: Redundancy or subtlety?
Blood
82
1993
3515
5
Broudy
VC
Stem cell factor and hematopoiesis.
Blood
90
1997
1345
6
Galli
SJ
Zsebo
KM
Geissler
EN
The kit ligand, stem cell factor.
Adv Immunol
55
1994
1
7
Namikawa
R
Muench
MO
Roncarolo
MG
Regulatory roles of the ligand for flk2/flt3 tyrosine kinase receptor on human hematopoiesis.
Stem Cells
14
1996
388
8
McNiece
IK
Briddell
RA
Stem cell factor.
J Leukoc Biol
58
1995
14
9
Silvers WK: Dominant Spotting, Patch, and Rump-White, in Silvers WK (eds): The Coat Colors of Mice: A Model for Mammalian Gene Action and Interaction. New York, NY, Springer-Verlag, 1979, p 206
10
Russell
ES
Hereditary anemias of the mouse: A review for geneticists.
Adv Genet
20
1979
357
11
Chabot
B
Stephenson
DA
Chapman
VM
Besmer
P
Bernstein
A
The proto-oncogene c-kit encoding a transmembrane tyrosine kinase receptor maps to the mouse W locus.
Nature
335
1988
88
12
Geissler
EN
Ryan
MA
Houseman
DE
The dominant-white spotting (W) locus of the mouse encodes the c-kit proto-oncogene.
Cell
55
1988
185
13
Coussens
L
Van Beveren
C
Smith
D
Chen
E
Mitchell
RL
Isacke
CM
Verma
IM
Ullrich
A
Structural alteration of viral homologue of receptor proto-oncogene fms at carboxyl terminus.
Nature
320
1986
277
14
Woolford
J
McAuliffe
A
Rohrschneider
LR
Activation of the feline c-fms proto-oncogene: Multiple alterations are required to generate a fully transformed phenotype.
Cell
55
1988
965
15
Rothwell
VM
Rohrschneider
LR
Murine c-fms cDNA: Cloning, sequence analysis and retroviral expression.
Oncogene Res
1
1987
311
16
Rosnet
O
Schiff
C
Pébusque
M-J
Marchetto
S
Tonnelle
C
Toiron
Y
Birg
F
Birnbaum
D
Human FLT3/FLK2 gene: cDNA cloning and expression in hematopoietic cells.
Blood
82
1993
1110
17
Small
D
Levenstein
M
Kim
E
Carow
C
Amin
S
Rockwell
P
Witte
L
Burrow
C
Ratajczak
MZ
Gewirtz
AM
Civin
CI
STK-1, the human homolog of Flk-2/Flt-3, is selectively expressed in CD34+ human bone marrow cells and is involved in the proliferation of early progenitor/stem cells.
Proc Natl Acad Sci USA
91
1994
459
18
Rosnet
O
Marchetto
S
deLapeyriere
O
Birnbaum
D
Murine Flt3, a gene encoding a novel tyrosine kinase receptor of the PDGFR/CSF1R family.
Oncogene
6
1991
1641
19
Matthews
W
Jordan
CT
Wiegand
GW
Pardoll
D
Lemischka
IR
A receptor tyrosine kinase specific to hematopoietic stem and progenitor cell-enriched populations.
Cell
65
1991
1143
20
Yarden
Y
Escobedo
JA
Kuang
WJ
Yang-Feng
TL
Daniel
TO
Tremble
PM
Chen
EY
Ando
ME
Harkins
RN
Francke
U
Fried
VA
Ullrich
A
Williams
LT
Structure of the receptor for platelet-derived growth factor helps define a family of closely related growth factor receptors.
Nature
323
1986
226
21
Gronwald
RG
Grant
FJ
Haldeman
BA
Hart
CE
O'Hara
PJ
Hagen
FS
Ross
R
Bowen-Pope
DF
Murray
MJ
Cloning and expression of a cDNA coding for the human platelet-derived growth factor receptor: Evidence for more than one receptor class.
Proc Natl Acad Sci USA
85
1988
3435
22
Claesson-Welsh
L
Eriksson
A
Moren
A
Severinsson
L
Ek
B
Ostman
A
Betsholtz
C
Heldin
CH
cDNA cloning and expression of a human platelet-derived growth factor (PDGF) receptor specific for B-chain-containing PDGF molecules.
Mol Cell Biol
8
1988
3476
23
Matsui
T
Heidaran
M
Miki
T
Popescu
N
La Rochelle
W
Kraus
M
Pierce
J
Aaronson
S
Isolation of a novel receptor cDNA establishes the existence of two PDGF receptor genes.
Science
243
1989
800
24
Bernstein
A
Forrester
L
Reith
AD
Dubreuil
P
Rottapel
R
The murine W/c-kit and Steel loci and the control of hematopoiesis.
Semin Hematol
28
1991
138
25
Herbst
R
Shearman
MS
Obermeier
A
Schlessinger
J
Ullrich
A
Differential effects of W mutations on p145c-kit tyrosine kinase activity and substrate interaction.
J Biol Chem
267
1992
13210
26
Nocka
K
Tan
JC
Chiu
E
Chu
TY
Ray
P
Traktman
P
Besmer
P
Molecular bases of dominant negative and loss of function mutations at the murine c-kit/white spotting locus: W37, Wv, W41 and W.
EMBO J
9
1990
1805
27
Reith
AD
Rottapel
R
Giddens
E
Brady
C
Forrester
L
Bernstein
A
W mutant mice with mild or severe developmental defects contain distinct point mutations in the kinase domain of the c-kit receptor.
Genes Dev
4
1990
390
28
Tan
JC
Nocka
K
Ray
P
Traktman
P
Besmer
P
The dominant W42 spotting phenotype results from a missense mutation in the c-kit receptor kinase.
Science
247
1990
209
29
Sarvella
PA
Russell
LB
Steel, a new dominant gene in the house mouse.
J Hered
47
1956
123
30
Silvers WK: Steel, Flexed-Tailed, Splotch, and Varitint-Waddler, in Silvers WK (eds): The Coat Colors of Mice: A Model for Mammalian Gene Action and Interaction. New York, NY, Springer-Verlag, 1979, p 242
31
Stubbs L, Poustka A, Rohme D, Russell LB, Lehrach H: Approaching the mouse Steel locus from closely linked molecular markers, in Clarke A, Compans RW, Cooper M, Eisen H, Goebel W, Koprowsi H, Melchers F, Oldstone M, Vogt PK, Wagner H, Wilson I (eds): Current Topics in Microbiology and Immunology. Berlin, Germany, Springer-Verlag, 1988, p 47
32
Huang
E
Nocka
K
Beier
DR
Chu
T-Y
Buck
J
Lahm
HW
Wellner
D
Leder
P
Besmer
P
The hematopoietic growth factor KL is encoded by the Sl locus and is the ligand of the c-kit receptor, the gene product of the W locus.
Cell
63
1990
225
33
Martin
FH
Suggs
SV
Langley
KE
Lu
HS
Ting
J
Okino
KH
Morris
CF
McNiece
IK
Jacobsen
FW
Mendiaz
EA
Birkett
NC
Smith
KA
Johnson
MJ
Parker
VP
Flores
JC
Patel
AC
Fisher
EF
Erjavec
HO
Herrera
CJ
Wypych
J
Sachdev
RK
Pope
JA
Leslie
I
Wen
D
Lin
C
Cupples
RL
Zsebo
KM
Primary structure and functional expression of rat and human stem cell factor DNAs.
Cell
63
1990
203
34
Williams
DE
Eisenman
J
Baird
A
Rauch
C
Van Ness
K
March
CJ
Park
LS
Martin
U
Mochizuki
DY
Boswell
HS
Burgess
GS
Cosman
D
Lyman
SD
Identification of a ligand for the c-kit proto-oncogene.
Cell
63
1990
167
35
Copeland
NG
Gilbert
DJ
Cho
BC
Donovan
PJ
Jenkins
NA
Cosman
D
Anderson
D
Lyman
SD
Williams
DE
Mast cell growth factor maps near the Steel locus on mouse chromosome 10 and is deleted in a number of Steel alleles.
Cell
63
1990
175
36
Zsebo
KM
Williams
DA
Geissler
EN
Broudy
VC
Martin
FH
Atkins
HL
Hsu
RY
Birkett
NC
Okino
KH
Murdock
DC
Jacobsen
FW
Langley
KE
Smith
KA
Takeishi
T
Cattanach
BM
Galli
SJ
Suggs
SV
Stem cell factor is encoded at the Sl locus of the mouse and is the ligand for the c-kit tyrosine kinase receptor.
Cell
63
1990
213
37
Zhou
J-H
Ohtaki
M
Sakurai
M
Sequence of a cDNA encoding chicken stem cell factor.
Gene
127
1993
269
38
Petitte
JN
Kulik
MJ
Cloning and characterization of cDNAs encoding two forms of avian stem cell factor.
Biochim Biophys Acta
1307
1996
149
39
Shull
RM
Suggs
SV
Langley
KE
Okino
KH
Jacobsen
FW
Martin
FH
Canine stem cell factor (c-kit ligand) supports the survival of hematopoietic progenitors in long-term canine marrow culture.
Exp Hematol
20
1992
1118
40
Anderson
DM
Williams
DE
Tushinski
R
Gimpel
S
Eisenman
J
Cannizzaro
LA
Aronson
M
Croce
CM
Huebner
K
Cosman
D
Lyman
SD
Alternate splicing of mRNAs encoding human mast cell growth factor and localization of the gene to chromosome 12q22-q24.
Cell Growth Differ
2
1991
373
41
Rosnet
O
Mattei
MG
Marchetto
S
Birnbaum
D
Isolation and chromosomal localization of a novel FMS-like tyrosine kinase gene.
Genomics
9
1991
380
42
Van Zant
G
Eldridge
PW
Behringer
RR
Dewey
MJ
Genetic control of hematopoietic kinetics revealed by analyses of allophenic mice and stem cell suicide.
Cell
35
1983
639
43
Iwama
A
Okano
K
Sudo
T
Matsuda
Y
Suda
T
Molecular cloning of a novel receptor tyrosine kinase gene, STK, derived from enriched hematopoietic stem cells.
Blood
83
1994
3160
44
Lyman
SD
James
L
Zappone
J
Sleath
PR
Beckmann
MP
Bird
T
Characterization of the protein encoded by the flt3 (flk2) receptor-like tyrosine kinase gene.
Oncogene
8
1993
815
45
Zeigler
FC
Bennett
BD
Jordan
CT
Spencer
SD
Baumhueter
S
Carroll
KJ
Hooley
J
Bauer
K
Matthews
W
Cellular and molecular characterization of the role of the FLK-2/FLT-3 receptor tyrosine kinase in hematopoietic stem cells.
Blood
84
1994
2422
46
Rossner
MT
McArthur
GA
Allen
JD
Metcalf
D
Fms-like tyrosine kinase 3 catalytic domain can transduce a proliferative signal in FDC-P1 cells that is qualitatively similar to the signal delivered by c-Fms.
Cell Growth Differ
5
1994
549
47
Dosil
M
Wang
S
Lemischka
IR
Mitogenic signalling and substrate specificity of the Flk2/Flt3 receptor tyrosine kinase in fibroblasts and interleukin 3-dependent hematopoietic cells.
Mol Cell Biol
13
1993
6572
48
Lyman
SD
James
L
Vanden Bos
T
de Vries
P
Brasel
K
Gliniak
B
Hollingsworth
LT
Picha
KS
McKenna
HJ
Splett
RR
Çletcher
FF
Maraskovsky
E
Farrah
T
Foxworthe
D
Williams
DE
Beckmann
MP
Molecular cloning of a ligand for the flt3/flk-2 tyrosine kinase receptor: A proliferative factor for primitive hematopoietic cells.
Cell
75
1993
1157
49
Hannum
C
Culpepper
J
Campbell
D
McClanahan
T
Zurawski
S
Bazan
JF
Kastelein
R
Hudak
S
Wagner
J
Mattson
J
Luh
J
Duda
G
Martina
N
Peterson
D
Menon
S
Shanafelt
A
Muench
M
Kelner
G
Namikawa
R
Rennick
D
Roncarolo
M-G
Zlotnick
A
Rosnet
O
Dubreuil
P
Birnbaum
D
Lee
F
Ligand for FLT3/FLK2 receptor tyrosine kinase regulates growth of haematopoietic stem cells and is encoded by variant RNAs.
Nature
368
1994
643
50
Lyman
SD
James
L
Johnson
L
Brasel
K
de Vries
P
Escobar
SS
Downey
H
Splett
RR
Beckmann
MP
McKenna
HJ
Cloning of the human homologue of the murine flt3 ligand: a growth factor for early hematopoietic progenitor cells.
Blood
83
1994
2795
51
Lyman
SD
Brasel
K
Rousseau
AM
Williams
DE
The flt3 ligand: A hematopoietic stem cell factor whose activities are distinct from steel factor.
Stem Cells
12
1994
99
52
Matous
JV
Langley
K
Kaushansky
K
Structure-function relationships of stem cell factor: An analysis based on a series of human-murine stem cell factor chimera and the mapping of a neutralizing monoclonal antibody.
Blood
88
1996
437
53
Qiu
F
Ray
P
Brown
K
Barker
PE
Jhanwar
S
Ruddle
FH
Besmer
P
Primary structure of c-kit: Relationship with the CSF-1/PDGF receptor kinase family-oncogenic activation of v-kit involves deletion of extracellular domain and C terminus.
EMBO J
7
1988
1003
54
Yarden
Y
Kuang
W-J
Yang-Feng
T
Coussens
L
Munemitsu
S
Dull
TJ
Chen
E
Schlessinger
J
Francke
U
Ullrich
A
Human proto-oncogene c-kit: A new cell surface receptor tyrosine kinase for an unidentified ligand.
EMBO J
6
1987
3341
55
Majumder
S
Brown
K
Qiu
F-H
Besmer
P
c-kit protein, a transmembrane kinase: Identification in tissues and characterization.
Mol Cell Biol
8
1988
4896
56
Blume-Jensen
P
Claesson-Welsh
L
Siegbahn
A
Zsebo
KM
Westermark
B
Heldin
C-H
Activation of the human c-kit product by ligand-induced dimerization mediates circular actin reorganization and chemotaxis.
EMBO J
10
1991
4121
57
Rosnet
O
Bühring
H-J
Marchetto
S
Rappold
I
Lavagna
C
Sainty
D
Arnoulet
C
Chabannon
C
Kanz
L
Hannum
C
Birnbaum
D
Human FLT3/FLK2 receptor tyrosine kinase is expressed at the surface of normal and malignant hematoietic cells.
Leukemia
10
1996
238
58
Rose
C
Rockwell
P
Yang
JQ
Pytowski
B
Goldstein
NI
Isolation and characterization of a monoclonal antibody binding to the extracellular domain of the flk-2 tyrosine kinase receptor.
Hybridoma
14
1995
453
59
Maroc
N
Rottapel
R
Rosnet
O
Marchetto
S
Lavezzi
C
Mannoni
P
Birnbaum
D
Dubreuil
P
Biochemical characterization and analysis of the transforming potential of the FLT3/FLK2 receptor tyrosine kinase.
Oncogene
8
1993
909
60
Broudy
VC
Kovach
NL
Bennett
LG
Lin
N
Jacobsen
FW
Kidd
PG
Human umbilical vein endothelial cells display high-affinity c-kit receptors and produce a soluble form of the c-kit receptor.
Blood
83
1994
2145
61
Broudy
VC
Smith
FO
Lin
N
Zsebo
KM
Egrie
J
Bernstein
ID
Blasts from patients with acute myelogenous leukemia express functional receptors for stem cell factor.
Blood
80
1992
60
62
Lev
S
Yarden
Y
Givol
D
Dimerization and activation of the kit receptor by monovalent and bivalent binding of the stem cell factor.
J Biol Chem
267
1992
15970
63
Turner
AM
Zsebo
KM
Martin
F
Jacobsen
FW
Bennett
LG
Broudy
VC
Nonhematopoietic tumor cell lines express stem cell factor and display c-kit receptors.
Blood
80
1992
374
64
Turner
AM
Bennett
LG
Lin
NL
Wypych
J
Bartley
TD
Hunt
RW
Atkins
HL
Langley
KE
Parker
V
Martin
F
Broudy
VC
Identification and characterization of a soluble c-kit receptor produced by human hematopoietic cell lines.
Blood
85
1995
2052
65
Turner
AM
Lin
NL
Issarachai
S
Lyman
SD
Broudy
VC
FLT3 receptor expression on the surface of normal and malignant human hematopoietic cells.
Blood
88
1996
3383
66
Blechman
JM
Lev
S
Barg
J
Eisenstein
M
Vaks
B
Vogel
Z
Givol
D
Yarden
Y
The fourth immunoglobulin domain of the stem cell factor receptor couples ligand binding to signal transduction.
Cell
80
1995
103
67
Lemmon
MA
Pinchasi
D
Zhous
M
Lax
I
Schlessinger
J
Kit receptor dimerization is driven by bivalent binding of stem cell factor.
J Biol Chem
272
1997
6311
68
Blechman
JM
Lev
S
Brizzi
MF
Leitner
O
Pegoraro
L
Givol
D
Yarden
Y
Soluble c-kit proteins and antireceptor monoclonal antibodies confine the binding site of the stem cell factor.
J Biol Chem
268
1993
4399
69
Hsu
Y
Wu
G
Mendiaz
EA
Syed
R
Wypych
J
Toso
R
Mann
MB
Boone
TC
Narhi
LO
Lu
HS
Langley
KE
The majority of stem cell factor exists as monomer under physiological conditions.
J Biol Chem
272
1997
6406
70
Lev
S
Yarden
Y
Givol
D
A recombinant ectodomain of the receptor for the stem cell factor (SCF) retains ligand-induced receptor dimerization and antagonizes SCF-stimulated cellular responses.
J Biol Chem
267
1992
10866
71
Lev
S
Givol
D
Yarden
Y
Interkinase domain of kit contains the binding site for phosphatidylinositol 3′ kinase.
Proc Natl Acad Sci USA
89
1992
678
72
Reith
AD
Ellis
C
Lyman
SD
Anderson
DM
Williams
DE
Bernstein
A
Pawson
T
Signal transduction by normal isoforms and W mutant variants of the Kit receptor tyrosine kinase.
EMBO J
10
1991
2451
73
Hayashi
S-I
Kunisada
T
Ogawa
M
Yamaguchi
K
Nishikawa
S-I
Exon skipping by mutation of an authentic splice site of c-kit gene in W/W mouse.
Nucleic Acids Res
19
1991
1267
74
Crosier
PS
Ricciardi
ST
Hall
LR
Vitas
MR
Clark
SC
Crosier
KE
Expression of isoforms of the human receptor tyrosine kinase c-kit in leukemic cell lines and acute myeloid leukemia.
Blood
82
1993
1151
75
Piao
X
Curtis
JE
Minkin
S
Minden
MD
Bernstein
A
Expression of the Kit and KitA receptor isoforms in human acute myelogenous leukemia.
Blood
83
1994
476
76
Wypych
J
Bennett
LG
Schwartz
MG
Clogston
CL
Lu
HS
Broudy
VC
Bartley
TD
Parker
VP
Langley
KE
Soluble kit receptor in human serum.
Blood
85
1995
66
77
Lavagna
C
Marchetto
S
Birnbaum
D
Rosnet
O
Identification and characterization of a functional murine FLT3 isoform produced by exon skipping.
J Biol Chem
270
1995
3165
78
Bazan
JF
Genetic and structural homology of stem cell factor and macrophage colony-stimulating factor.
Cell
65
1991
9
79
Anderson
DM
Lyman
SD
Baird
A
Wignall
JM
Eisenman
J
Rauch
C
March
CJ
Boswell
HS
Gimpel
SD
Cosman
D
Williams
DE
Molecular cloning of mast cell growth factor, a hematopoietin that is active in both membrane bound and soluble forms.
Cell
63
1990
235
80
Arakawa
T
Yphantis
DA
Lary
JW
Narhi
LO
Lu
HS
Prestrelski
SJ
Clogston
CL
Zsebo
KM
Mendiaz
EA
Wypych
J
Langley
KE
Glycosylated and unglycosylated recombinant-derived human stem cell factors are dimeric and have extensive regular secondary structure.
J Biol Chem
266
1991
18942
81
Lu
HS
Clogston
CL
Wypych
J
Fausset
PR
Lauren
S
Mendiaz
EA
Zsebo
KM
Langley
KE
Amino acid sequence and post-translational modification of stem cell factor isolated from buffalo rat liver cell-conditioned medium.
J Biol Chem
266
1991
8102
82
Pandit
J
Bohm
A
Jancarik
J
Halenbeck
R
Koths
K
Kim
S-H
Three-dimensional structure of dimeric human recombinant macrophage colony-stimulating factor.
Science
258
1992
1358
83
Nishikawa
M
Tojo
A
Ikebuchi
K
Katayama
K
Fujii
N
Ozawa
K
Asano
S
Deletion mutagenesis of stem cell factor defines the C-terminal sequences essential for its biological activity.
Biochem Biophys Res Commun
188
1992
292
84
Langley KE, Mendiaz EA, Liu N, Narhi LO, Zeni L, Parseghian CM, Clogston CL, Leslie I, Pope JA, Lu HS, Zsebo KM: Properties of variant forms of human stem cell factor recombinantly expressed inEscherichia coli. Arch Biochem Biophys 311:55, 1994
85
(abstr, suppl 1)
Escobar
S
Brasel
K
Anderberg
R
Lyman
SD
Structure function studies of human flt3 ligand.
Blood
86
1995
21a
86
Zsebo
KM
Wypych
J
McNiece
IK
Lu
HS
Smith
KA
Karkare
SB
Sachdev
RK
Yuschenkoff
VN
Birkett
NC
Williams
LR
Satyagal
VN
Tung
W
Bosselman
RA
Mendiaz
EA
Langley
KE
Identification, purification, and biological characterization of hemopoietic stem cell factor from buffalo rat liver-conditioned medium.
Cell
63
1990
195
87
Majumdar
MK
Feng
L
Medlock
E
Toksoz
D
Williams
DA
Identification and mutation of primary and secondary proteolytic cleavage sites in murine stem cell factor cDNA yields biologically active, cell-associated protein.
J Biol Chem
269
1994
1237
88
Flanagan
JG
Chan
DC
Leder
P
Transmembrane form of the kit ligand growth factor is determined by alternative splicing and is missing in the Sld mutant.
Cell
64
1991
1025
89
Toksoz
D
Zsebo
KM
Smith
KA
Hu
S
Brankow
D
Suggs
SV
Martin
FH
Williams
DA
Support of human hematopoiesis in long-term bone marrow cultures by murine stromal cells selectively expressing the membrane-bound and secreted forms of the human homolog of the steel gene product, stem cell factor.
Proc Natl Acad Sci USA
89
1992
7350
90
Huang
EJ
Nocka
KH
Buck
J
Besmer
P
Differential expression and processing of two cell associated forms of the kit-ligand: KL-1 and KL-2.
Mol Biol Cell
3
1992
349
91
Lyman SD, Williams DE: Biological control of mast cell growth factor c-kit interactions may be mediated through alternate splicing of mRNAs, in Murphy MJ Jr (eds): Blood Cell Growth Factors: Their Present and Future Use in Hematology and Oncology. Proceedings of the Beijing Symposium, August 21-24, 1991. Dayton, OH, AlphaMed, 1991, p 183
92
Brannan
CI
Lyman
SD
Williams
DE
Eisenman
J
Anderson
DM
Cosman
D
Bedell
MA
Jenkins
NA
Copeland
NG
Steel-Dickie mutation encodes a c-Kit ligand lacking transmembrane and cytoplasmic domains.
Proc Natl Acad Sci USA
88
1991
4671
93
Cerretti
DP
Wignall
J
Anderson
D
Tushinski
RJ
Gallis
BM
Stya
M
Gillis
S
Urdal
DL
Cosman
D
Human macrophage-colony stimulating factor: Alternative RNA and protein processing from a single gene.
Mol Immunol
25
1988
761
94
Lyman
SD
James
L
Escobar
S
Downey
H
de Vries
P
Brasel
K
Stocking
K
Beckmann
MP
Copeland
NG
Cleveland
LS
Jenkins
NA
Belmont
JW
Davison
BL
Identification of soluble and membrane-bound isoforms of the murine flt3 ligand generated by alternative splicing of mRNAs.
Oncogene
10
1995
149
95
Lyman
SD
Stocking
K
Davison
B
Fletcher
F
Johnson
L
Escobar
S
Structural analysis of human and murine flt3 ligand genomic loci.
Oncogene
11
1995
1165
96
Agnès
F
Shamoon
B
Dina
C
Rosnet
O
Birnbaum
D
Galibert
F
Genomic structure of the downstream part of the human FLT3 gene: Exon/intron structure conservation among genes encoding receptor tyrosine kinases (RTK) of subclass III.
Gene
145
1994
283
97
Gokkel
E
Grossman
Z
Ramot
B
Yarden
Y
Rechavi
G
Givol
D
Structural organization of the murine c-kit proto-oncogene.
Oncogene
7
1992
1423
98
André
C
Martin
E
Cornu
F
Hu
W-X
Wang
X-P
Galibert
F
Genomic organization of the human c-kit gene: Evolution of the receptor tyrosine kinase subclass III.
Oncogene
7
1992
685
99
Vandenbark
GR
deCastro
CM
Taylor
H
Dew-Knight
S
Kaufman
RE
Cloning and structural analysis of the human c-kit gene.
Oncogene
7
1992
1259
100
Giebel
LB
Strunk
KM
Holmes
SA
Spritz
RA
Organization and nucleotide sequence of the human KIT (mast/stem cell growth factor receptor) proto-oncogene.
Oncogene
7
1992
2207
101
Imbert
A
Rosnet
O
Marchetto
S
Ollendorff
V
Birnbaum
D
Pebusque
MJ
Characterization of a yeast artificial chromosome from human chromosome band 13q12 containing the FLT1 and FLT3 receptor-type tyrosine kinase genes.
Cytogenet Cell Genet
67
1994
175
102
(abstr, suppl 1)
Wang
Z
Kim
E
Chinault
AC
Civin
CI
Small
D
Genomic organization of the human Stk-1 (flt3/flk2) gene.
Blood
88
1996
111b
103
Brannan
CI
Bedell
MA
Resnick
JL
Eppig
JJ
Handel
MA
Williams
DE
Lyman
SD
Donovan
PJ
Jenkins
NA
Copeland
NG
Developmental abnormalities in Steel17H mice result from a splicing defect in the steel factor cytoplasmic tail.
Genes Dev
6
1992
1832
104
Ladner
MB
Martin
GA
Noble
JA
Nikoloff
DM
Tal
R
Kawasaki
ES
White
TJ
Human CSF-1: Gene structure and alternative splicing of mRNA precursors.
EMBO J
6
1987
2693
105
Rosnet
O
Stephenson
D
Mattei
M-G
Marchetto
S
Shibuya
M
Chapman
VM
Birnbaum
D
Close physical linkage of the FLT1 and FLT3 genes on chromosome 13 in man and chromosome 5 in mouse.
Oncogene
8
1993
173
106
Shibuya
M
Yamaguchi
S
Yamane
A
Ikeda
T
Tojo
A
Matsushime
H
Sato
M
Nucleotide sequence and expression of a novel human receptor-type tyrosine kinase gene (flt) closely related to the fms family.
Oncogene
5
1990
519
107
d'Auriol
L
Mattei
MG
Andre
C
Galibert
F
Localization of the human c-kit protooncogene on the q11-q12 region of chromosome 4.
Hum Genet
78
1988
374
108
Geissler
EN
Liao
M
Brook
JD
Martin
FH
Zsebo
KM
Housman
DE
Galli
SJ
Stem cell factor (SCF), a novel hematopoietic growth factor and ligand for c-kit tyrosine kinase receptor, maps on human chromosome 12 between 12q14.3 and 12qter.
Somat Cell Mol Genet
17
1991
207
109
McClanahan
T
Culpepper
J
Campbell
D
Wagner
J
Franz-Bacon
K
Mattson
J
Tsai
S
Luh
J
Guimaraes
MJ
Mattei
M-G
Rosnet
O
Birnbaum
D
Hannum
CH
Biochemical and genetic characterization of multiple splice variants of the Flt3 ligand.
Blood
88
1996
3371
110
Ezoe
K
Holmes
SA
Ho
L
Bennett
CP
Bolognia
JL
Brueton
L
Burn
J
Falabella
R
Gatto
EM
Ishii
N
Moss
C
Pittelkow
MR
Thompson
E
Ward
KA
Spritz
RA
Novel mutations and deletion of the KIT (steel factor receptor) gene in human piebaldism.
Am J Hum Genet
56
1995
58
111
Keller
SA
Liptay
S
Hajra
A
Meisler
MH
Transgene-induced mutation of the murine steel locus.
Proc Natl Acad Sci USA
87
1990
10019
112
Bedell
MA
Brannan
CI
Evans
EP
Copeland
NG
Jenkins
NA
Donovan
PJ
DNA rearrangements located over 100 kb 5′ of the Steel (Sl)-coding region in Steel-panda and Steel-contrasted mice deregulate Sl expression and cause female sterility by disrupting ovarian follicle development.
Genes Dev
9
1995
455
113
Johansson
B
Billstrom
R
Mauritzson
N
Mitelman
F
Trisomy 19 as the sole chromosomal anomaly in hematologic neoplasms.
Cancer Genet Cytogenet
74
1994
62
114
Keshet
E
Lyman
SD
Williams
DE
Anderson
DM
Jenkins
NA
Copeland
NG
Parada
LF
Embryonic RNA expression patterns of the c-kit receptor and its cognate ligand suggest multiple functional roles in mouse development.
EMBO J
10
1991
2425
115
Matsui
Y
Zsebo
KM
Hogan
BLM
Embryonic expression of a haematopoietic growth factor encoded by the Sl locus and the ligand for c-kit.
Nature
347
1990
667
116
Motro
B
van der Kooy
D
Rossant
J
Reith
A
Bernstein
A
Contiguous patterns of c-kit and steel expression: analysis of mutations at the W and Sl loci.
Development
113
1991
1207
117
Aye
MT
Hashemi
S
Leclair
B
Zeibdawi
A
Trudel
E
Halpenny
M
Fuller
V
Cheng
G
Expression of stem cell factor and c-kit mRNA in cultured endothelial cells, monocytes and cloned human bone marrow stromal cells (CFU-RF).
Exp Hematol
20
1992
523
118
McNiece
IK
Langley
KE
Zsebo
KM
The role of recombinant stem cell factor in early B cell development. Synergistic interaction with IL-7.
J Immunol
146
1991
3785
119
Flanagan
JG
Leder
P
The kit ligand: A cell surface molecule altered in Steel mutant fibroblasts.
Cell
63
1990
185
120
deLapeyriere
O
Naquet
P
Planche
J
Marchetto
S
Rottapel
R
Gambarelli
D
Rosnet
O
Birnbaum
D
Expression of Flt3 tyrosine kinase receptor gene in mouse hematopoietic and nervous tissues.
Differentiation
58
1995
351
121
Hu
ZB
Ma
W
Uphoff
CC
Quentmeier
H
Drexler
HG
c-kit expression in human megakaryoblastic leukemia cell lines.
Blood
83
1994
2133
122
André
C
d'Auriol
L
Lacombe
C
Gisselbrecht
S
Galibert
F
c-kit mRNA expression in human and murine hematopoietic cell lines.
Oncogene
4
1989
1047
123
Da Silva
N
Hu
ZB
Ma
W
Rosnet
O
Birnbaum
D
Drexler
HG
Expression of the FLT3 gene in human leukemia-lymphoma cell lines.
Leukemia
8
1994
885
124
Wang
C
Curtis
JE
Geissler
EN
McCulloch
EA
Minden
MD
The expression of the proto-oncogene C-kit in the blast cells of acute myeloblastic leukemia.
Leukemia
3
1989
699
125
Morita
S
Tsuchiya
S
Fujie
H
Itano
M
Ohashi
Y
Minegishi
M
Imaizumi
M
Endo
M
Takano
N
Konno
T
Cell surface c-kit receptors in human leukemia cell lines and pediatric leukemia: Selective preservation of c-kit expression on megakaryoblastic cell lines during adaptation to in vitro culture.
Leukemia
10
1996
102
126
de Castro
CM
Denning
SM
Langdon
S
Vandenbark
GR
Kurtzberg
J
Scearce
R
Haynes
BF
Kaufman
RE
The c-kit proto-oncogene receptor is expressed on a subset of human CD3−CD4−CD8− (triple-negative) thymocytes.
Exp Hematol
22
1994
1025
127
Moriyama Y, Tsujimura T, Hashimoto K, Morimoto M, Kitayama H, Matsuzawa, Kitamura Y, Kanakura Y: Role of aspartic acid 814 in the function and expression of c-kit receptor tyrosine kinase. J Biol Chem 271:3347, 1996
128
Hjertson
M
Sundström
C
Lyman
SD
Nilsson
K
Nilsson
G
Stem cell factor, but not flt3 ligand, induces differentiation and activation of human mast cells.
Exp Hematol
24
1996
748
129
Brasel
K
Escobar
S
Anderberg
R
de Vries
P
Gruss
H-J
Lyman
SD
Expression of the flt3 receptor and its ligand on hematopoietic cells.
Leukemia
9
1995
1212
130
Meierhoff
G
Dehmel
U
Gruss
H-J
Rosnet
O
Birnbaum
D
Quentmeier
H
Dirks
W
Drexler
HG
Expression of flt3 receptor and flt3-ligand in human leukemia-lymphoma cell lines.
Leukemia
9
1995
1368
131
Ikeda
H
Kanakura
Y
Tamaki
T
Kuriu
A
Kitayama
H
Ishikawa
J
Kanayama
Y
Yonezawa
T
Tarui
S
Griffin
JD
Expression and functional role of the proto-oncogene c-kit in acute myeloblastic leukemia cells.
Blood
78
1991
2962
132
Lerner
NB
Nocka
KH
Cole
SR
Qiu
FH
Strife
A
Ashman
LK
Besmer
P
Monoclonal antibody YB5.B8 identifies the human c-kit protein product.
Blood
77
1991
1876
133
Kubota
A
Okamura
S
Shimoda
K
Harada
M
Niho
Y
The c-kit molecule and the surface immunophenotype of human acute leukemia.
Leuk Lymphoma
14
1994
421
134
Reuss-Borst
MA
Buhring
HJ
Schmidt
H
Muller
CA
AML: Immunophenotypic heterogeneity and prognostic significance of c-kit expression.
Leukemia
8
1994
258
135
Kanakura
Y
Ikeda
H
Kitayama
H
Sugahara
H
Furitsu
T
Expression, function and activation of the proto-oncogene c-kit product in human leukemia cells.
Leuk Lymphoma
10
1993
35
136
Lauria
F
Bagnara
GP
Rondelli
D
Raspadori
D
Strippoli
P
Bonsi
L
Ventura
MA
Montanaro
LL
Bubola
G
Tura
S
Broudy
VC
Cytofluorimetric and functional analysis of c-kit receptor in acute leukemia.
Leuk Lymphoma
18
1995
451
137
Carlesso
N
Pregno
P
Bresso
P
Gallo
E
Pileri
A
Zsebo
KM
Ferrero
D
Human recombinant stem cell factor stimulates in vitro proliferation of acute myeloid leukemia cells and expands the clonogenic cell pool.
Leukemia
6
1992
642
138
Goselink
HM
Williams
DE
Fibbe
WE
Wessels
HW
Beverstock
GC
Willemze
R
Falkenburg
JH
Effect of mast cell growth factor (c-kit ligand) on clonogenic leukemic precursor cells.
Blood
80
1992
750
139
Valverde
LR
Matutes
E
Farahat
N
Heffernan
A
Owusu-Ankomah
K
Morilla
R
Catovsky
D
C-kit receptor (CD117) expression in acute leukemia.
Ann Hematol
72
1996
11
140
Cole
SR
Aylett
GW
Harvey
NL
Cambareri
AC
Ashman
LK
Increased expression of c-kit or its ligand Steel factor is not a common feature of adult acute myeloid leukaemia.
Leukemia
10
1996
288
141
Drexler
HG
Expression of FLT3 receptor and response to FLT3 ligand by leukemic cells.
Leukemia
10
1996
588
142
McKenna
HJ
Smith
FO
Brasel
K
Hirschstein
D
Bernstein
ID
Williams
DE
Lyman
SD
Effects of flt3 ligand on acute myeloid and lymphocytic leukemic blast cells from children.
Exp Hematol
24
1996
378
143
Carow
CE
Levenstein
M
Kaufmann
SH
Chen
J
Amin
S
Rockwell
P
Witte
L
Borowitz
MJ
Civin
CI
Small
D
Expression of the hematopoietic growth factor receptor FLT3 (STK-1/Flk2) in human leukemias.
Blood
87
1996
1089
144
Birg
F
Courcoul
M
Rosnet
O
Bardin
F
Pébusque
M-J
Marchetto
S
Tabilio
A
Mannoni
P
Birnbaum
D
Expression of the FMS/KIT-like gene FLT3 in human acute leukemias of the myeloid and lymphoid lineages.
Blood
80
1992
2584
145
Stacchini
A
Fubini
L
Severino
A
Sanavio
F
Aglietta
M
Piacibello
W
Expression of type III receptor tyrosine kinases FLT3 and KIT and responses to their ligands by acute myeloid leukemia blasts.
Leukemia
10
1996
1584
146
Piacibello
W
Fubini
L
Sanavio
F
Brizzi
MF
Severino
A
Garetto
L
Stacchini
A
Pegoraro
L
Aglietta
M
Effects of human FLT3 ligand on myeloid leukemia cell growth: Heterogeneity in response and synergy with other hematopoietic growth factors.
Blood
86
1995
4105
147
Pinto
A
Gloghini
A
Gattei
V
Aldinucci
D
Zagonel
V
Carbone
A
Expression of the c-kit receptor in human lymphomas is restricted to Hodgkin's disease and CD30+ anaplastic large cell lymphomas.
Blood
83
1994
785
148
Kiyoi
H
Naoe
T
Yokota
S
Nakao
M
Minami
S
Kuriyama
K
Takeshita
A
Saito
K
Hasegawa
S
Shimodaira
S
Tamura
J
Shimazaki
C
Matsue
K
Kobayashi
H
Arima
N
Suzuki
R
Morishita
H
Saito
H
Ueda
R
Ohno
R
Internal tandem duplication of FLT3 associated with leukocytosis in acute promyelocytic leukemia.
Leukemia
11
1997
1447
149
Horiike
S
Yokota
S
Nakao
M
Iwai
T
Sasai
Y
Kaneko
H
Taniwaki
M
Kashima
K
Fujii
H
Abe
T
Misawa
S
Tandem duplications of the FLT3 receptor gene are associated with leukemic transformation of myelodysplasia.
Leukemia
11
1997
1442
150
Buhring
HJ
Ullrich
A
Schaudt
K
Muller
CA
Busch
FW
The product of the proto-oncogene c-kit (P145c-kit) is a human bone marrow surface antigen of hemopoietic precursor cells which is expressed on a subset of acute non-lymphoblastic leukemic cells.
Leukemia
5
1991
854
151
Muroi
K
Nakamura
M
Amemiya
Y
Suda
T
Miura
Y
Expression of c-kit receptor (CD117) and CD34 in leukemic cells.
Leuk Lymphoma
16
1995
297
152
Carson
WE
Haldar
S
Baiocchi
RA
Croce
CM
Caligiuri
MA
The c-kit ligand suppresses apoptosis of human natural killer cells through the upregulation of bcl-2.
Proc Natl Acad Sci USA
91
1994
7553
153
Lisovsky
M
Estrov
Z
Zhang
X
Consoli
U
Sanchez-Williams
G
Snell
V
Munker
R
Goodacre
A
Savchenko
V
Andreeff
M
Flt3 ligand stimulates proliferation and inhibits apoptosis of acute myeloid leukemia cells: Regulation of Bcl-2 and Bax.
Blood
88
1996
3987
154
Wang
C
Koistinen
P
Yang
GS
Williams
DE
Lyman
SD
Minden
MD
McCulloch
EA
Mast cell growth factor, a ligand for the receptor encoded by c-kit, affects the growth in culture of the blast cells of acute myeloblastic leukemia.
Leukemia
5
1991
493
155
Hassan
HT
Zander
A
Stem cell factor as a survival and growth factor in human normal and malignant hematopoiesis.
Acta Haematol
95
1996
257
156
Pietsch
T
Kyas
U
Steffens
U
Yakisan
E
Hadam
MR
Ludwig
WD
Zsebo
K
Welte
K
Effects of human stem cell factor (c-kit ligand) on proliferation of myeloid leukemia cells: Heterogeneity in response and synergy with other hematopoietic growth factors.
Blood
80
1992
1199
157
Agarwal
R
Doren
S
Hicks
B
Dunbar
CE
Long-term culture of chronic myelogenous leukemia marrow cells on stem cell factor-deficient stroma favors benign progenitors.
Blood
85
1995
1306
158
(abstr, suppl 1)
Mahon
FX
Pigeonnier
V
Chahine
H
Barbot
C
Jazwiec
B
Ripoche
J
Reiffers
J
Flt3 ligand preferentially stimulates normal immature progenitor (Philadelphia negative) in chronic myeloid leukemia (CML).
Blood
88
1996
234a
159
Eder
M
Hemmati
P
Kalina
U
Ottmann
OG
Hoelzer
D
Lyman
SD
Ganser
A
Effects of Flt3 ligand and interleukin-7 on in vitro growth of acute lymphoblastic leukemia cells.
Exp Hematol
24
1996
371
160
Fukuda
T
Kamishima
T
Tsuura
Y
Suzuki
T
Kakihara
T
Naito
M
Kishi
K
Matsumoto
K
Shibata
A
Seito
T
Expression of the c-kit gene product in normal and neoplastic mast cells but not in neoplastic basophil/mast cell precursors from chronic myelogenous leukemia.
J Pathol
177
1995
139
161
Tsujimura
T
Furitsu
T
Morimoto
M
Kanayama
Y
Nomura
S
Matsuzawa
Y
Kitamura
Y
Kanakura
Y
Substitution of an aspartic acid results in constitutive activation of c-kit receptor tyrosine kinase in a rat tumor mast cell line RBL-2H3.
Int Arch Allergy Immunol
106
1995
377
162
Metcalf
D
Nicola
NA
Direct proliferative actions of stem cell factor on murine bone marrow cells in vitro: Effects of combination with colony-stimulating factors.
Proc Natl Acad Sci USA
88
1991
6239
163
Rolink
A
Ghia
P
Grawunder
U
Haasner
D
Karasuyama
H
Kalberer
C
Winkler
T
Melchers
F
In-vitro analyses of mechanisms of B-cell development.
Semin Immunol
7
1995
155
164
Rico-Vargas
SA
Weiskopf
B
Nishikawa
S
Osmond
DG
c-kit expression by B cell precursors in mouse bone marrow. Stimulation of B cell genesis by in vivo treatment with anti-c-kit antibody.
J Immunol
152
1994
2845
165
Moore
TA
Zlotnik
A
T-cell lineage commitment and cytokine responses of thymic progenitors.
Blood
86
1995
1850
166
Rasko
JEJ
Metcalf
D
Rossner
MT
Begley
CG
Nicola
NA
The flt3/flk-2 ligand: receptor distribution and action on murine haemopoietic cell survival and proliferation.
Leukemia
9
1995
2058
167
Hunt
P
Zsebo
KM
Hokom
MM
Hornkohl
A
Birkett
NC
del Castillo
JC
Martin
F
Evidence that stem cell factor is involved in the rebound thrombocytosis that follows 5-fluorouracil treatment.
Blood
80
1992
904
168
Avraham
H
Vannier
E
Cowley
S
Jiang
SX
Chi
S
Dinarello
CA
Zsebo
KM
Groopman
JE
Effects of the stem cell factor, c-kit ligand, on human megakaryocytic cells.
Blood
79
1992
365
169
Ratajczak
MZ
Ratajczak
J
Ford
J
Kregenow
R
Marlicz
W
Gewirtz
AM
FLT3/FLK-2 (STK-1) ligand does not stimulate human megakaryopoiesis in vitro.
Stem Cells
14
1996
146
170
Grabarek
J
Groopman
JE
Lyles
YR
Jiang
S
Bennett
L
Zsebo
K
Avraham
H
Human kit ligand (stem cell factor) modulates platelet activation in vitro.
J Biol Chem
269
1994
21718
171
Wasserman
R
Li
YS
Hardy
RR
Differential expression of the blk and ret tyrosine kinases during B lineage development is dependent on Ig rearrangement.
J Immunol
155
1995
644
172
Papayannopoulou
T
Brice
M
Broudy
VC
Zsebo
KM
Isolation of c-kit receptor-expressing cells from bone marrow, peripheral blood, and fetal liver: Functional properties and composite antigenic profile.
Blood
78
1991
1403
173
Ashman
LK
Cambareri
AC
To
LB
Levinsky
RJ
Juttner
CA
Expression of the YB5.B8 antigen (c-kit proto-oncogene product) in normal human bone marrow.
Blood
78
1991
30
174
Broudy
VC
Lin
N
Zsebo
KM
Birkett
NC
Smith
KA
Bernstein
ID
Papayannopoulou
T
Isolation and characterization of a monoclonal antibody that recognizes the human c-kit receptor.
Blood
79
1992
338
175
Matos
ME
Schnier
GS
Beecher
MS
Ashman
LK
William
DE
Caligiuri
MA
Expression of a functional c-kit receptor on a subset of natural killer cells.
J Exp Med
178
1993
1079
176
Rappold
I
Ziegler
BL
Köhler
I
Marchetto
S
Rosnet
O
Birnbaum
D
Simmons
PJ
Zannettino
ACW
Hill
B
Neu
S
Knapp
W
Alitalo
R
Alitalo
K
Ullrich
A
Kanz
L
Büring
HJ
Functional and phenotypic characterization of cord blood and bone marrow subsets expressing FLT3 (CD135) receptor tyrosine kinase.
Blood
90
1997
111
177
Tsai
M
Takeishi
T
Thompson
H
Langley
KE
Zsebo
KM
Metcalfe
DD
Geissler
EN
Galli
SJ
Induction of mast cell proliferation, maturation and heparin synthesis by rat c-kit ligand, stem cell factor.
Proc Natl Acad Sci USA
88
1991
6382
178
Galli
SJ
Iemura
A
Garlick
DS
Gamba-Vitalo
C
Zsebo
KM
Andrews
RG
Reversible expansion of primate mast cell populations in vivo by stem cell factor.
J Clin Invest
91
1993
148
179
Costa
JJ
Demetri
GD
Hayes
DF
Merica
EA
Menchaca
DM
Galli
SJ
Increased skin mast cells and urine methyl histamine in patients receiving recombinant methionyl human stem cell factor.
Proc Am Assoc Cancer Res
34
1993
211
180
Lynch
DH
Jacobs
C
DuPont
D
Eisenman
J
Foxworthe
D
Martin
U
Miller
RE
Roux
E
Liggitt
D
Williams
DE
Pharmacokinetic parameters of recombinant mast cell growth factor (rMGF).
Lymphokine Cytokine Res
11
1992
233
181
Moskowitz
CH
Stiff
P
Gordon
MS
McNiece
I
Ho
AD
Costa
JJ
Broun
ER
Bayer
RA
Wyres
M
Hill
J
Jelaca-Maxwell
K
Nichols
CR
Brown
SL
Nimer
SD
Gabrilove
J
Recombinant methionyl human stem cell factor and filgrastim for peripheral blood progenitor cell mobilization and transplantation in non-Hodgkins lymphoma patients—Results of a phase I/II trial.
Blood
89
1997
3136
182
Demetri
G
Costa
J
Hayes
D
Sledge
G
Galli
S
Hoffman
R
Merica
E
Rich
W
Harkins
B
McGuire
B
Gordon
M
A phase I trial of recombinant methionyl human stem cell factor (SCF) in patients with advanced breast carcinoma pre- and post-chemotherapy with cyclophosphamide and doxorubicin.
Proc Am Assoc Clin Oncol
12
1993
A367
183
Ogawa
M
Matsuzaki
Y
Nishikawa
S
Hayashi
S-I
Kunisada
T
Sudo
T
Kina
T
Nakauchi
H
Nishikawa
S-I
Expression and function of c-kit in hemopoietic progenitor cells.
J Exp Med
174
1991
63
184
Okada
S
Nakauchi
H
Nagayoshi
K
Nishikawa
S-I
Miura
Y
Suda
T
In vivo and in vitro stem cell function of c-kit- and Sca-1-positive murine hematopoietic cells.
Blood
80
1992
3044
185
Okada
S
Nakauchi
H
Nagayoshi
K
Nishikawa
S
Nishikawa
S
Miura
Y
Suda
T
Enrichment and characterization of murine hematopoietic stem cells that express c-kit molecule.
Blood
78
1991
1706
186
Ikuta
K
Weissman
IL
Evidence that hematopoietic stem cells express mouse c-kit but do not depend on steel factor for their generation.
Proc Natl Acad Sci USA
89
1992
1502
187
Orlic
D
Fischer
R
Nishikawa
S
Nienhuis
AW
Bodine
DM
Purification and characterization of heterogeneous pluripotent hematopoietic stem cell populations expressing high levels of c-kit receptor.
Blood
82
1993
762
188
Li
CL
Johnson
GR
Murine hematopoietic stem and progenitor cells: I. Enrichment and biologic characterization.
Blood
85
1995
1472
189
Simmons
PJ
Aylett
GW
Niutta
S
To
LB
Juttner
CA
Ashman
LK
c-kit is expressed by primitive human hematopoietic cells that give rise to colony-forming cells in stroma-dependent or cytokine-supplemented culture.
Exp Hematol
22
1994
157
190
Olweus
J
Terstappen
LW
Thompson
PA
Lund-Johansen
F
Expression and function of receptors for stem cell factor and erythropoietin during lineage commitment of human hematopoietic progenitor cells.
Blood
88
1996
1594
191
Briddell
RA
Broudy
VC
Bruno
E
Brandt
JE
Srour
EF
Hoffman
R
Further phenotypic characterization and isolation of human hematopoietic progenitor cells using a monoclonal antibody to the c-kit receptor.
Blood
79
1992
3159
192
Gabbianelli
M
Pelosi
E
Montesoro
E
Valtieri
M
Luchetti
L
Samoggia
P
Vitelli
L
Barberi
T
Testa
U
Lyman
S
Peschle
C
Multi-level effects of flt3 ligand on human hematopoiesis: Expansion of putative stem cells and proliferation of granulomonocytic progenitors/monocytic precursors.
Blood
86
1995
1661
193
Saraya
K
Reid
CD
Stem cell factor and the regulation of dendritic cell production from CD34+ progenitors in bone marrow and cord blood.
Br J Haematol
93
1996
258
194
Szabolcs
P
Moore
MAS
Young
JW
Expansion of immunostimulatory dendritic cells among the myeloid progeny of human CD34+ bone marrow precursors cultured with c-kit ligand, granulocyte-macrophage colony-stimulating factor, and TNF-α.
J Immunol
154
1995
5851
195
Rosenzwajg
M
Canque
B
Gluckman
JC
Human dendritic cell differentiation pathway from CD34+ hematopoietic precursor cells.
Blood
87
1996
535
196
(abstr, suppl 1)
Maraskovsky
E
Roux
E
Tepee
M
McKenna
HJ
Brasel
K
Lyman
SD
Williams
DE
The effect of Flt3 ligand and/or c-kit ligand on the generation of dendritic cells from human CD34+ bone marrow.
Blood
86
1995
420a
197
Broxmeyer
HE
Hangoc
G
Cooper
S
Anderson
D
Cosman
D
Lyman
SD
Williams
DE
Influence of murine mast cell growth factor (c-kit ligand) on colony formation by mouse marrow hematopoietic progenitor cells.
Exp Hematol
19
1991
143
198
Xiao
M
Leemhuis
T
Broxmeyer
HE
Lu
L
Influence of combinations of cytokines on proliferation of isolated single cell-sorted human bone marrow hematopoietic progenitor cells in the absence and presence of serum.
Exp Hematol
20
1992
276
199
Broxmeyer
HE
Cooper
S
Lu
L
Hangoc
G
Anderson
D
Cosman
D
Lyman
SD
Williams
DE
Effect of murine mast cell growth factor (c-kit proto-oncogene ligand) on colony formation by human marrow hematopoietic progenitor cells.
Blood
77
1991
2142
200
McNiece
IK
Langley
KE
Zsebo
KM
Recombinant human stem cell factor synergizes with GM-CSF, G-CSF, IL-3 and Epo to stimulate human progenitor cells of the myeloid and the erythroid lineages.
Blood
19
1991
226
201
Uoshima
N
Ozawa
M
Kimura
S
Tanaka
K
Wada
K
Kobayashi
Y
Kondo
M
Changes in c-Kit expression and effects of SCF during differentiation of human erythroid progenitor cells.
Br J Haematol
91
1995
30
202
Sui
X
Tsuji
K
Tajima
S
Tanaka
R
Muraoka
K
Ebihara
Y
Ikebuchi
K
Yasukawa
K
Taga
T
Kishimoto
T
Nakahata
T
Erythropoietin-independent erythrocyte production: Signals through gp130 and c-kit dramatically promote erythropoiesis from human CD34+ cells.
J Exp Med
183
1996
837
203
Wu
H
Klingmuller
U
Besmer
P
Lodish
HF
Interaction of the erythropoietin and stem cell factor receptors.
Nature
377
1995
242
204
Levesque
JP
Haylock
DN
Simmons
PJ
Cytokine regulation of proliferation and cell adhesion are correlated events in human CD34+ hematopoietic progenitors.
Blood
88
1996
1168
205
Jacobsen
SEW
Okkenhaug
C
Myklebust
J
Veiby
OP
Lyman
SD
The FLT3 ligand potently and directly stimulates the growth and expansion of primitive murine bone marrow progenitor cells in vitro: synergistic interactions with interleukin (IL) 11, IL-12, and other hematopoietic growth factors.
J Exp Med
181
1995
1357
206
Hudak
S
Hunte
B
Culpepper
J
Menon
S
Hannum
C
Thompson-Snipes
L
Rennick
D
FLT3/FLK2 ligand promotes the growth of murine stem cells and the expansion of colony-forming cells and spleen colony-forming units.
Blood
85
1995
2747
207
Rusten
LS
Lyman
SD
Veiby
OP
Jacobsen
SEW
The FLT3 ligand is a direct and potent stimulator of the growth of primitive and committed human CD34+ bone marrow progenitor cells in vitro.
Blood
87
1996
1317
208
McKenna
HJ
de Vries
P
Brasel
K
Lyman
SD
Williams
DE
Effect of flt3 ligand on the ex vivo expansion of human CD34+ hematopoietic progenitor cells.
Blood
86
1995
3413
209
Ebbe
S
Phalen
E
Stohlman
FJ
Abnormal megakaryocytopoiesis in Sl/Sld mice.
Blood
42
1973
865
210
Adrados
C
Ebbe
S
Phalen
E
Garbutt
P
Allan
C
Macrocytic megakaryocytes in cultures of Sl/Sld bone marrow.
Exp Hematol
12
1984
237
211
Ebbe
S
Bentfeld-Barker
M
Adrados
C
Carpenter
D
Mortensen
C
Yee
T
Phalen
E
Functionally abnormal stromal cells and megakaryocyte size, ploidy, and ultrastructure in Sl/Sld mice.
Blood Cells
12
1986
217
212
Briddell
RA
Bruno
E
Cooper
RJ
Brandt
JE
Hoffman
R
Effect of c-kit ligand on in vitro human megakaryocytopoiesis.
Blood
78
1991
2854
213
Tanaka
R
Koike
K
Imai
T
Shiohara
M
Kubo
T
Amano
Y
Komiyama
A
Nakahata
T
Stem cell factor enhances proliferation, but not maturation, of murine megakaryocytic progenitors in serum-free culture.
Blood
80
1992
1743
214
Debili
N
Masse
JM
Katz
A
Guichard
J
Breton-Gorius
J
Vainchenker
W
Effects of the recombinant hematopoietic growth factors interleukin-3, interleukin-6, stem cell factor, and leukemia inhibitory factor on the megakaryocytic differentiation of CD34+ cells.
Blood
82
1993
84
215
Imai
T
Nakahata
T
Stem cell factor promotes proliferation of human primitive megakaryocytic progenitors, but not megakaryocytic maturation.
Int J Hematol
59
1994
91
216
(abstr, suppl 1)
Burstein
SA
Henthorn
J
Mei
R
Williams
DE
Mast cell growth factor (MGF) promotes human and murine megakaryocytic (MK) differentiation in vitro.
Blood
78
1991
160a
217
Kaushansky
K
Thrombopoietin: The primary regulator of platelet production.
Blood
86
1995
419
218
Nichol
JL
Hokom
MM
Hornkohl
A
Sheridan
WP
Ohashi
H
Kato
T
Li
YS
Bartley
TD
Choi
E
Bogenberger
J
Skrine
JD
Knudten
A
Chen
J
Trail
G
Sleeman
L
Cole
S
Grampp
G
Hunt
P
Megakaryocyte growth and development factor. Analyses of in vitro effects on human megakaryocytopoiesis and endogenous serum levels during chemotherapy-induced thrombocytopenia.
J Clin Invest
95
1995
2973
219
Broudy
VC
Lin
NL
Kaushansky
K
Thrombopoietin (c-mpl ligand) acts synergistically with erythropoietin, stem cell factor, and interleukin-11 to enhance murine megakaryocyte colony growth and increases megakaryocyte ploidy in vitro.
Blood
85
1995
1719
220
Hunt
P
Li
YS
Nichol
JL
Hokom
MM
Bogenberger
JM
Swift
SE
Skrine
JD
Hornkohl
AC
Lu
H
Clogston
C
Merewether
LA
Johnson
MJ
Parker
V
Knudten
A
Farese
A
Hsu
RY
Garcia
A
Stead
R
Bosselman
RA
Bartley
TD
Purification and biologic characterization of plasma-derived megakaryocyte growth and development factor.
Blood
86
1995
540
221
Banu
N
Wang
JF
Deng
B
Groopman
JE
Avraham
H
Modulation of megakaryocytopoiesis by thrombopoietin: The c-Mpl ligand.
Blood
86
1995
1331
222
Ku
H
Yonemura
Y
Kaushansky
K
Ogawa
M
Thrombopoietin, the ligand for the Mpl receptor, synergizes with steel factor and other early acting cytokines in supporting proliferation of primitive hematopoietic progenitors of mice.
Blood
87
1996
4544
223
Ramsfjell
V
Borge
OJ
Veiby
OP
Cardier
J
Murphy
MJ
Jr
Lyman
SD
Lok
S
Jacobsen
SEW
Thrombopoietin, but not erythropoietin, directly stimulates multilineage growth of primitive murine bone marrow progenitor cells in synergy with early acting cytokines: distinct interactions with the ligands for c-kit and FLT3.
Blood
88
1996
4481
224
Ramsfjell
V
Borge
OJ
Cui
L
Jacobsen
SEW
Thrombopoietin directly and potently stimulates multilineage growth and progenitor cell expansion from primitive (CD34+CD38−) human bone marrow progenitor cells: Distinct and key interactions with the ligands for c-kit and flt3, and inhibitory effects of TGF-β and TNF-α.
J Immunol
158
1997
5169
225
Sitnicka
E
Lin
N
Priestley
GV
Fox
N
Broudy
VC
Wolf
NS
Kaushansky
K
The effect of thrombopoietin on the proliferation and differentiation of murine hematopoietic stem cells.
Blood
87
1996
4998
226
Kobayashi
M
Laver
JH
Kato
T
Miyazaki
H
Ogawa
M
Thrombopoietin supports proliferation of human primitive hematopoietic cells in synergy with steel factor and/or interleukin-3.
Blood
88
1996
429
227
Mackarehtschian
K
Hardin
JD
Moore
KA
Boast
S
Goff
SP
Lemischka
IR
Targeted disruption of the flk2/flt3 gene leads to deficiencies in primitive hematopoietic progenitors.
Immunity
3
1995
147
228
Piacibello
W
Garetto
L
Sanavio
F
Severino
A
Fubini
L
Stacchini
A
Dragonetti
G
Aglietta
M
The effects of human FLT3 ligand on in vitro human megakaryocytopoiesis.
Exp Hematol
24
1996
340
229
Piacibello
W
Sanavio
F
Garetto
L
Severino
A
Bergandi
D
Ferrario
J
Fagioli
F
Berger
M
Aglietta
M
Extensive amplification and self-renewal of human primitive hematopoietic stem cells from cord blood.
Blood
89
1997
2644
230
Henderson
AJ
Narayanan
R
Collins
L
Dorshkind
K
Status of κL chain gene rearrangements and c-kit and IL-7 receptor expression in stromal cell-dependent pre-B cells.
J Immunol
149
1992
1973
231
Faust
EA
Saffran
DC
Toksoz
D
Williams
DA
Witte
ON
Distinctive growth requirements and gene expression patterns distinguish progenitor B cells from pre-B cells.
J Exp Med
177
1993
915
232
Hozumi
K
Kobori
A
Sato
T
Nozaki
H
Nishikawa
S
Nishimura
T
Habu
S
Pro-T cells in fetal thymus express c-kit and RAG-2 but do not rearrange the gene encoding the T cell receptor beta chain.
Eur J Immunol
24
1994
1339
233
Godfrey
DI
Zlotnik
A
Suda
T
Phenotypic and functional characterization of c-kit expression during intrathymic T cell development.
J Immunol
149
1992
2281
234
Godfrey
DI
Kennedy
J
Mombaerts
P
Tonegawa
S
Zlotnik
A
Onset of TCR-β gene rearrangement and role of TCR-beta expression during CD3−CD4−CD8− thymocyte differentiation.
J Immunol
152
1994
4783
235
Godfrey
DI
Kennedy
J
Gately
MK
Hakimi
J
Hubbard
BR
Zlotnik
A
IL-12 influences intrathymic T cell development.
J Immunol
152
1994
2729
236
Dehmel
U
Quentmeier
H
Drexler
HG
Effects of FLT3 ligand on human leukemia cells. II. Agonistic and antagonistic effects of other cytokines.
Leukemia
10
1996
271
237
Wu
L
Vremec
D
Ardavin
C
Winkel
K
Süss
G
Georgiou
H
Maraskovsky
E
Cook
W
Shortman
K
Mouse thymus dendritic cells: Kinetics of development and changes in surface markers during maturation.
Eur J Immunol
25
1995
418
238
Landreth
KS
Kincade
PW
Lee
G
Harrison
DE
B lymphocyte precursors in embryonic and adult W anemic mice.
J Immunol
132
1984
2724
239
Billips
LG
Petitte
D
Dorshkind
K
Narayanan
R
Chiu
C-P
Landreth
KS
Differential roles of stromal cells, interleukin-7, and kit-ligand in the regulation of B lymphopoiesis.
Blood
79
1992
1185
240
Funk
PE
Varas
A
Witte
PL
Activity of stem cell factor and IL-7 in combination on normal bone marrow B lineage cells.
J Immunol
150
1993
748
241
Rolink
A
Streb
M
Nishikawa
S-I
Melchers
F
The c-kit-encoded tyrosine kinase regulates the proliferation of early pre-B cells.
Eur J Immunol
21
1991
2609
242
Palacios
R
Samaridis
J
Fetal liver pro-B and pre-B lymphocyte clones: Expression of lymphoid-specific genes, surface markers, growth requirements, colonization of the bone marrow, and generation of B lymphocytes in vivo and in vitro.
Mol Cell Biol
12
1992
518
243
Hirayama
F
Shih
JP
Awgulewitsch
A
Warr
GW
Clark
SC
Ogawa
M
Clonal proliferation of murine lymphohemopoietic progenitors in culture.
Proc Natl Acad Sci USA
89
1992
5907
244
Ball
TC
Hirayama
F
Ogawa
M
Lymphohematopoietic progenitors of normal mice.
Blood
85
1995
3086
245
Hirayama
F
Lyman
SD
Clark
SC
Ogawa
M
The flt3 ligand supports proliferation of lymphohematopoietic progenitors and early B-lymphoid progenitors.
Blood
85
1995
1762
246
Kee
BL
Cumano
A
Iscove
NN
Paige
CJ
Stromal cell independent growth of bipotent B cell—macrophage precursors from murine fetal liver.
Int Immunol
6
1994
401
247
Takeda
S
Shimizu
T
Rodewald
HR
Interactions between c-kit and stem cell factor are not required for B-cell development in vivo.
Blood
89
1997
518
248
(abstr, suppl 1)
McKenna
HJ
Miller
RE
Brasel
KE
Maraskovsky
E
Maliszewski
C
Pulendran
B
Lynch
D
Teepe
M
Roux
ER
Smith
J
Williams
DE
Lyman
SD
Peschon
JJ
Stocking
K
Targeted disruption of the flt3 ligand gene in mice affects multiple hematopoietic lineages, including natural killer cells, B lymphocytes, and dendritic cells.
Blood
88
1996
474a
249
Hunte
BE
Hudak
S
Campbell
D
Xu
Y
Rennick
D
flk2/flt3 ligand is a potent cofactor for the growth of primitive B cell progenitors.
J Immunol
156
1996
489
250
Peschon
JJ
Morrissey
PJ
Grabstein
KH
Ramsdell
FJ
Maraskovsky
E
Gliniak
BC
Park
LS
Ziegler
SF
Williams
DE
Ware
CB
Meyer
JD
Davison
BL
Early lymphocyte expansion is severely impaired in interleukin 7 receptor-deficient mice.
J Exp Med
180
1994
1955
251
Ray
RJ
Paige
CJ
Furlonger
C
Lyman
SD
Rottapel
R
Flt3 ligand supports the differentiation of early B cell progenitors in the presence of interleukin-11 and interleukin-7.
Eur J Immunol
26
1996
1504
252
Veiby
OP
Lyman
SD
Jacobsen
SEW
Combined signaling through interleukin-7 receptors and flt3 but not c-kit potently and selectively promotes B-cell commitment and differentiation from uncommitted murine bone marrow progenitor cells.
Blood
88
1996
1256
253
Saeland
S
Moreau
I
Duvert
V
Pandrau
D
Bancherau
J
In vitro growth and maturation of human B-cell precursors.
Curr Top Microbiol Immunol
182
1992
85
254
Abboud
MR
Xu
F
Payne
A
Laver
J
Effects of recombinant human Steel factor (c-kit ligand) on early cord blood hematopoietic precursors.
Exp Hematol
22
1994
388
255
Rawlings
DJ
Quan
SG
Kato
RM
Witte
ON
Long-term culture system for selective growth of human B-cell progenitors.
Proc Natl Acad Sci USA
92
1995
1570
256
Namikawa
R
Muench
MO
de Vries
JE
Roncarolo
MG
The FLK2/FLT3 ligand synergizes with interleukin-7 in promoting stromal-cell-independent expansion and differentiation of human fetal pro-B cells in vitro.
Blood
87
1996
1881
257
Mekori
T
Phillips
RA
The immune response in mice of genotypes W-Wv and Sl-Sld1.
Proc Soc Exp Biol Med
132
1969
115
258
Asamoto
H
Mandel
TE
Thymus mice bearing the steel mutation. Morphologic studies on fetal, neonatal, organ-cultured, and grafted fetal thymus.
Lab Invest
45
1981
418
259
de Vries
P
Brasel
KA
McKenna
HJ
Williams
DE
Watson
JD
Thymus reconstitution by c-kit-expressing hematopoietic stem cells purified from adult mouse bone marrow.
J Exp Med
176
1992
1503
260
Matsuzaki
Y
Gyotoku
J
Ogawa
M
Nishikawa
S
Katsura
Y
Gachelin
G
Nakauchi
H
Characterization of c-kit positive intrathymic stem cells that are restricted to lymphoid differentiation.
J Exp Med
178
1993
1283
261
Morrissey
PJ
McKenna
H
Widmer
MB
Braddy
S
Voice
R
Charrier
K
Williams
DE
Watson
JD
Steel factor (c-kit ligand) stimulates the in vitro growth of immature CD3−/CD4−/CD8− thymocytes: Synergy with IL-7.
Cell Immunol
157
1994
118
262
Moore
TA
Zlotnik
A
Differential effects of Flk-2/Flt-3 ligand and stem cell factor on murine thymic progenitor cells.
J Immunol
158
1997
4187
263
Tjonnfjord
GE
Veiby
OP
Steen
R
Egeland
T
T lymphocyte differentiation in vitro from adult human prethymic CD34+ bone marrow cells.
J Exp Med
177
1993
1531
264
Freedman
AR
Zhu
H
Levine
JD
Kalams
S
Scadden
DT
Generation of human T lymphocytes from bone marrow CD34+ cells in vitro.
Nat Med
2
1996
46
265
Silva
MR
Hoffman
R
Srour
EF
Ascensao
JL
Generation of human natural killer cells from immature progenitors does not require marrow stromal cells.
Blood
84
1994
841
266
Shibuya
A
Nagayoshi
K
Nakamura
K
Nakauchi
H
Lymphokine requirement for the generation of natural killer cells from CD34+ hematopoietic progenitor cells.
Blood
85
1995
3538
267
Mrozek
E
Anderson
P
Caligiuri
MA
Role of interleukin-15 in the development of human CD56+ natural killer cells from CD34+ hematopoietic progenitor cells.
Blood
87
1996
2632
268
(abstr, suppl 1)
Yu
H
Carson
W
Caligiuri
M
The Flt3 ligand enhances expansion but not differentiation of human natural killer (NK) cells from CD34+ hematopoetic progenitor cells (HPCs) when combined with interleukin 15 (IL-15).
Blood
88
1996
105b
269
Peters
JH
Gieseler
R
Thiele
B
Steinbach
F
Dendritic cells: From ontogenetic orphans to myelomonocytic descendants.
Immunol Today
17
1996
273
270
Caux
C
Liu
YJ
Banchereau
J
Recent advances in the study of dendritic cells and follicular dendritic cells.
Immunol Today
16
1995
2
271
Siena
S
Di Nicola
M
Bregni
M
Mortarini
R
Anichini
A
Lombardi
L
Ravagnani
F
Parmiani
G
Gianni
AM
Massive ex vivo generation of functional dendritic cells from mobilized CD34+ blood progenitors for anticancer therapy.
Exp Hematol
23
1995
1463
272
Saunders
D
Lucas
K
Ismaili
J
Wu
J
Maraskovsky
E
Dunn
A
Shortman
K
Dendritic cell development in culture from thymic precursor cells in the absence of granulocyte/macrophage colony-stimulating factor.
J Exp Med
184
1996
2185
273
Maraskovsky
E
Brasel
K
Teepe
M
Roux
ER
Lyman
SD
Shortman
K
McKenna
HJ
Dramatic increase in the numbers of functionally mature dendritic cells in Flt3 ligand-treated mice: Multiple dendritic cell subpopulations identified.
J Exp Med
184
1996
1953
274
Sanchez
MJ
Holmes
A
Miles
C
Dzierzak
E
Characterization of the first definitive hematopoietic stem cells in the AGM and liver of the mouse embryo.
Immunity
5
1996
513
275
Wineman
JP
Nishikawa
S
Muller-Sieburg
CE
Maintenance of high levels of pluripotent hematopoietic stem cells in vitro: Effect of stromal cells and c-kit.
Blood
81
1993
365
276
de Jong
MO
Rozemuller
H
Kieboom
D
Visser
JW
Wognum
AW
Wagemaker
G
Purification of repopulating hemopoietic cells based on binding of biotinylated Kit ligand.
Leukemia
10
1996
1813
277
Osawa
M
Hamada
K-I
Hamada
H
Nakauchi
H
Long-term lympho-hematopoietic reconstitution by a single CD34− low/negative hematopoietic stem cell.
Science
273
1996
242
278
Keller
JR
Ortiz
M
Spence
SE
Lohrey
N
Ruscetti
FW
Characterization of a c-kit negative primitive murine hematopoietic stem cell.
Exp Hematol
23
1995
815a
279
Jones
RJ
Collector
MI
Barber
JP
Vala
MS
Fackler
MJ
May
WS
Griffin
CA
Hawkins
AL
Zehnbauer
BA
Hilton
J
Colvin
OM
Sharkis
SJ
Characterization of mouse lymphohematopoietic stem cells lacking spleen colony-forming activity.
Blood
88
1996
487
280
Doi
H
Inaba
M
Yamamoto
Y
Taketani
S
Mori
SI
Sugihara
A
Ogata
H
Toki
J
Hisha
H
Inaba
K
Sogo
S
Adachi
M
Matsuda
T
Good
RA
Ikehara
RA
Pluripotent hemopoietic stem cells are c-kit<low.
Proc Natl Acad Sci USA
94
1997
2513
281
Katayama
N
Shih
JP
Nishikawa
S
Kina
T
Clark
SC
Ogawa
M
Stage-specific expression of c-kit protein by murine hematopoietic progenitors.
Blood
82
1993
2353
282
Visser
JW
Rozemuller
H
de Jong
MO
Belyavsky
A
The expression of cytokine receptors by purified hemopoietic stem cells.
Stem Cells
11
1993
49
283
de Vries
P
Brasel
KA
Eisenman
JR
Alpert
AR
Williams
DE
The effect of recombinant mast cell growth factor on purified murine hematopoietic stem cells.
J Exp Med
173
1991
1205
284
Tsuji
K
Zsebo
KM
Ogawa
M
Enhancement of murine blast cell colony formation in culture by recombinant rat stem cell factor, ligand for c-kit.
Blood
78
1991
1223
285
Migliaccio
G
Migliaccio
AR
Valinsky
J
Langley
KE
Zsebo
KM
Visser
JMW
Adamson
JW
Stem cell factor induces proliferation and differentiation of highly enriched murine hemopoietic cells.
Proc Natl Acad Sci USA
88
1991
7420
286
Lowry
PA
Zsebo
KM
Deacon
DH
Eichman
CE
Quesenberry
PJ
Effects of rrSCF on multiple cytokine responsive HPP-CFC generated from SCA+Lin− murine hematopoietic progenitors.
Exp Hematol
19
1991
994
287
Williams
N
Bertoncello
I
Kavnoudias
H
Zsebo
K
McNiece
I
Recombinant rat stem cell factor stimulates the amplification and differentiation of fractionated mouse stem cell populations.
Blood
79
1992
58
288
Tsuji
K
Lyman
SD
Sudo
T
Clark
SC
Ogawa
M
Enhancement of murine hematopoiesis by synergistic interactions between Steel factor (ligand for c-kit), interleukin-11, and other early acting factors in culture.
Blood
79
1992
2855
289
Lowry PA, Deacon D, Whitefield P, McGrath HE, Quesenberry PJ: Stem cell factor induction of in vitro murine hematopoietic colony formation by “subliminal” cytokine combinations: the role of “anchor factors.” Blood 80:663, 1992
290
Muench
MO
Schneider
JG
Moore
MA
Interactions among colony-stimulating factors, IL-1 beta, IL-6, and kit-ligand in the regulation of primitive murine hematopoietic cells.
Exp Hematol
20
1992
339
291
Jacobsen
SEW
Veiby
OP
Smeland
EB
Cytotoxic lymphocyte maturation factor (interleukin 12) is a synergistic growth factor for hematopoietic stem cells.
J Exp Med
178
1993
413
292
Keller
JR
Gooya
JM
Ruscetti
FW
Direct synergistic effects of leukemia inhibitory factor on hematopoietic progenitor cell growth: Comparison with other hematopoietins that use the gp130 receptor subunit.
Blood
88
1996
863
293
Broxmeyer
HE
Lu
L
Cooper
S
Ruggieri
L
Li
ZH
Lyman
SD
Flt3 ligand stimulates/costimulates the growth of myeloid stem/progenitor cells.
Exp Hematol
23
1995
1121
294
Fujimoto
K
Lyman
SD
Hirayama
F
Ogawa
M
Isolation and characterization of primitive hematpoietic progenitors of murine fetal liver.
Exp Hematol
24
1996
285
295
Jacobsen
FW
Stokke
T
Jacobsen
SEW
Transforming growth factor-beta potently inhibits the viability-promoting activity of stem cell factor and other cytokines and induces apoptosis of primitive murine hematopoietic progenitor cells.
Blood
86
1995
2957
296
Jacobsen
FW
Dubois
CM
Rusten
LS
Veiby
OP
Jacobsen
SEW
Inhibition of stem cell factor-induced proliferation of primitive murine hematopoietic progenitor cells signaled through the 75-kilodalton tumor necrosis factor receptor. Regulation of c-kit and p53 expression.
J Immunol
154
1995
3732
297
Yonemura
Y
Ku
H
Lyman
SD
Ogawa
M
In vitro expansion of hematopoietic progenitors and maintenance of stem cells: Comparison between FLT3/FLK-2 ligand and KIT ligand.
Blood
89
1997
1915
298
(abstr, suppl 1)
Weiss
M
Yetz-Aldape
J
Crosier
PS
Nathan
DG
Sieff
CA
Committed hematopoietic progenitors of human bone marrow are restricted to the CD38+34+ fraction whereas c-kit expression is greatest in CD38−34+ cells.
Blood
78
1991
161a
299
Civin
C
Almaida-Porada
G
Lee
M
Olweus
J
Terstappen
L
Zanjani
E
Sustained, retransplantable, multilineage engraftment of highly purified adult human bone marrow stem cells in vivo.
Blood
88
1996
4102
300
Larochelle
A
Vormoor
J
Hanenberg
H
Wang
J
Bhatia
M
Lapidot
T
Moritz
T
Murdoch
B
Xiao
X
Kato
I
Williams
D
Dick
J
Identification of primitive human hematopoietic cells capable of repopulating NOD/SCID mouse bone marrow: Implications for gene therapy.
Nat Med
2
1996
1329
301
Berardi
AC
Wang
A
Levine
JD
Lopez
P
Scadden
DT
Functional isolation and characterization of human hematopoietic stem cells.
Science
267
1995
104
302
Gunji
Y
Nakamura
M
Osawa
H
Nagayoshi
K
Nakauchi
H
Miura
Y
Yanagisawa
M
Suda
T
Human primitive hematopoietic progenitor cells are more enriched in KITlow cells than in KIThigh cells.
Blood
82
1993
3283
303
Kawashima
I
Zanjani
ED
Almaida-Porada
G
Flake
AW
Zeng
H
Ogawa
M
CD34+ human marrow cells that express low levels of Kit protein are enriched for long-term marrow-engrafting cells.
Blood
87
1996
4136
304
Laver
JH
Abboud
MR
Kawashima
I
Leary
AG
Ashman
LK
Ogawa
M
Characterization of c-kit expression by primitive hematopoietic progenitors in umbilical cord blood.
Exp Hematol
23
1995
1515
305
Bernstein
ID
Andrews
RG
Zsebo
KM
Recombinant human stem cell factor enhances the formation of colonies by CD34+ and CD34+lin− cells, and the generation of colony-forming cell progeny from CD34+lin− cells cultured with interleukin-3, granulocyte colony-stimulating factor, or granulocyte-macrophage colony-stimulating factor.
Exp Hematol
77
1991
2316
306
Carow
CE
Hangoc
G
Cooper
SH
Williams
DE
Broxmeyer
HE
Mast cell growth factor (c-kit ligand) supports the growth of human multipotential progenitor cells with a high replating potential.
Blood
78
1991
2216
307
Brandt
J
Briddell
RA
Srour
EF
Leemhuis
TB
Hoffman
R
Role of c-kit ligand in the expansion of human hematopoietic progenitor cells.
Blood
79
1992
634
308
Migliaccio
G
Migliaccio
AR
Druzin
ML
Giardina
PJ
Zsebo
KM
Adamson
JW
Long-term generation of colony-forming cells in liquid culture of CD34+ cord blood cells in the presence of recombinant human stem cell factor.
Blood
79
1992
2620
309
Lemoli
RM
Fogli
M
Fortuna
A
Motta
MR
Rizzi
S
Benini
C
Tura
S
Interleukin-11 stimulates the proliferation of human hematopoietic CD34+ and CD34+CD33− DR− cells and synergizes with stem cell factor, interleukin-3, and granulocyte-macrophage colony-stimulating factor.
Exp Hematol
21
1993
1668
310
Sonoda
Y
Sakabe
H
Ohmisono
Y
Tanimukai
S
Yokota
S
Nakagawa
S
Clark
SC
Abe
T
Synergistic actions of stem cell factor and other burst-promoting activities on proliferation of CD34+ highly purified blood progenitors expressing HLA-DR or different levels of c-kit protein.
Blood
84
1994
4099
311
Muench
MO
Roncarolo
MG
Menon
S
Xu
Y
Kastelein
R
Zurawski
S
Hannum
CH
Culpepper
J
Lee
F
Namikawa
R
FLK-2/FLT-3 ligand regulates the growth of early myeloid progenitors isolated from human fetal liver.
Blood
85
1995
963
312
Brashem-Stein
C
Flowers
DA
Bernstein
ID
Regulation of colony forming cell generation by flt-3 ligand.
Br J Haematol
94
1996
17
313
Shah
AJ
Smogorzewska
EM
Hannum
C
Crooks
GM
Flt3 ligand induces proliferation of quiescent human bone marrow CD34+CD38− cells and maintains progenitor cells in vitro.
Blood
87
1996
3563
314
Petzer
AL
Zandstra
PW
Piret
JM
Eaves
CJ
Differential cytokine effects on primitive (CD34+CD38−) human hematopoietic cells: Novel responses to Flt3-ligand and thrombopoietin.
J Exp Med
183
1996
2551
315
Elwood
NJ
Zogos
H
Willson
T
Begley
CG
Retroviral transduction of human progenitor cells: Use of granulocyte colony-stimulating factor plus stem cell factor to mobilize progenitor cells in vivo and stimulation by Flt3/Flk-2 ligand in vitro.
Blood
88
1996
4452
316
Dao
MA
Hannum
CH
Kohn
DB
Nolta
JA
FLT3 ligand preserves the ability of human CD34+ progenitors to sustain long-term hematopoiesis in immune-deficient mice after ex vivo retroviral-mediated transduction.
Blood
89
1997
446
317
Eaves
CJ
Cashman
JD
Eaves
AC
Methodology of long-term culture of human hemopoietic cells.
J Tiss Cult Methods
13
1991
55
318
Gartner
S
Kaplan
HS
Long-term culture of human bone marrow cells.
Proc Natl Acad Sci USA
77
1980
4756
319
Miller
CL
Rebel
VI
Lemieux
ME
Helgason
CD
Lansdorp
PM
Eaves
CJ
Studies of W mutant mice provide evidence for alternate mechanisms capable of activating hematopoietic stem cells.
Exp Hematol
24
1996
185
320
Kodama
H
Nose
M
Yamaguchi
Y
Tsunoda
J
Suda
T
Nishikawa
S
Nishikawa
S
In vitro proliferation of primitive hemopoietic stem cells supported by stromal cells: Evidence for the presence of a mechanism(s) other than that involving c-kit receptor and its ligand.
J Exp Med
176
1992
351
321
Liesveld
JL
Broudy
VC
Harbol
AW
Abboud
CN
Effect of stem cell factor on myelopoiesis potential in human Dexter-type culture systems.
Exp Hematol
23
1995
202
322
Sutherland
HJ
Hogge
DE
Cook
D
Eaves
CJ
Alternative mechanisms with and without steel factor support primitive human hematopoiesis.
Blood
81
1993
1465
323
Heinrich
MC
Dooley
DC
Freed
AC
Band
L
Hoatlin
ME
Keeble
WW
Peters
ST
Silvey
KV
Ey
FS
Kabat
D
Maziarz
RT
Bagby
GC
Jr
Constitutive expression of steel factor gene by human stromal cells.
Blood
82
1993
771
324
Papayannopoulou
T
Craddock
C
Nakamoto
B
Priestley
GV
Wolf
NS
The VLA4/VCAM-1 adhesion pathway defines contrasting mechanisms of lodgement of transplanted murine hemopoietic progenitors between bone marrow and spleen.
Proc Natl Acad Sci USA
92
1995
9647
325
Miyake
K
Weissman
IL
Greenberger
JS
Kincade
PW
Evidence for a role of the integrin VLA-4 in lympho-hemopoiesis.
J Exp Med
173
1991
599
326
Williams
DA
Rios
M
Stephens
C
Patel
VP
Fibronectin and VLA-4 in haematopoietic stem cell-microenvironment interactions.
Nature
352
1991
438
327
Hirsch
E
Iglesias
A
Potocnik
AJ
Hartmann
U
Fassler
R
Impaired migration but not differentiation of haematopoietic stem cells in the absence of beta 1 integrins.
Nature
380
1996
171
328
Arroyo
AG
Yang
JT
Rayburn
H
Hynes
RO
Differential requirements for alpha4 integrins during fetal and adult hematopoiesis.
Cell
85
1996
997
329
Levesque
JP
Leavesley
DI
Niutta
S
Vadas
M
Simmons
PJ
Cytokines increase human hemopoietic cell adhesiveness by activation of very late antigen (VLA)-4 and VLA-5 integrins.
J Exp Med
181
1995
1805
330
Kovach
NL
Lin
N
Yednock
T
Harlan
JM
Broudy
VC
Stem cell factor modulates avidity of α4β1 and α5β1 integrins expressed on hematopoietic cell lines.
Blood
85
1995
159
331
Kinashi
T
Springer
TA
Steel factor and c-kit regulate cell-matrix adhesion.
Blood
83
1994
1033
332
Dastych
J
Metcalfe
DD
Stem cell factor induces mast cell adhesion to fibronectin.
J Immunol
152
1994
213
333
Hanenberg
H
Xiao
XL
Dilloo
D
Hashino
K
Kato
I
Williams
DA
Colocalization of retrovirus and target cells on specific fibronectin fragments increases genetic transduction of mammalian cells.
Nat Med
2
1996
876
334
Hurley
RW
McCarthy
JB
Verfaillie
CM
Direct adhesion to bone marrow stroma via fibronectin receptors inhibits hematopoietic progenitor cell proliferation.
J Clin Invest
96
1995
511
335
Moritz
T
Patel
VP
Williams
DA
Bone marrow extracellular matrix molecules improve gene transfer into human hematopoietic cells via retroviral vectors.
J Clin Invest
93
1994
1451
336
Kodama
H
Nose
M
Niida
S
Nishikawa
S
Nishikawa
S
Involvement of the c-kit receptor in the adhesion of hematopoietic stem cells to stromal cells.
Exp Hematol
22
1994
979
337
Long
MW
Briddell
R
Walter
AW
Bruno
E
Hoffman
R
Human hematopoietic stem cell adherence to cytokines and matrix molecules.
J Clin Invest
90
1992
251
338
Kaneko
Y
Takenawa
J
Yoshida
O
Fujita
K
Sugimoto
K
Nakayama
H
Fujita
J
Adhesion of mouse mast cells to fibroblasts: adverse effects of Steel (SI) mutation.
J Cell Physiol
147
1991
224
339
Avraham
H
Scadden
DT
Chi
S
Broudy
VC
Zsebo
KM
Groopman
JE
Interaction of human bone marrow fibroblasts with megakaryocytes: Role of the c-kit ligand.
Blood
80
1992
1679
340
Adachi
S
Ebi
Y
Nishikawa
S
Hayashi
S
Yamazaki
M
Kasugai
T
Yamamura
T
Nomura
S
Kitamura
Y
Necessity of extracellular domain of W (c-kit) receptors for attachment of murine cultured mast cells to fibroblasts.
Blood
79
1992
650
341
Broudy
VC
Lin
NL
Priestley
GV
Nocka
K
Wolf
NS
Interaction of stem cell factor and its receptor c-kit mediates lodgment and acute expansion of hematopoietic cells in the murine spleen.
Blood
88
1996
75
342
Okumura
N
Tsuji
K
Ebihara
Y
Tanaka
I
Sawai
N
Koike
K
Komiyama
A
Nakahata
T
Chemotactic and chemokinetic activities of stem cell factor on murine hematopoietic progenitor cells.
Blood
87
1996
4100
343
Nilsson
G
Butterfield
JH
Nilsson
K
Siegbahn
A
Stem cell factor is a chemotactic for human mast cells.
J Immunol
153
1994
3717
344
Meininger
CJ
Yano
H
Rottapel
R
Bernstein
A
Zsebo
KM
Zetter
BR
The c-kit receptor ligand functions as a mast cell chemoattractant.
Blood
79
1992
958
345
Bodine
DM
Orlic
D
Birkett
NC
Seidel
NE
Zsebo
KM
Stem cell factor increases colony-forming unit-spleen number in vitro in synergy with interleukin-6, and in vivo in Sl/Sld mice as a single factor.
Blood
79
1992
913
346
Katayama
N
Clark
SC
Ogawa
M
Growth factor requirement for survival in cell-cycle dormancy of primitive murine lymphohematopoietic progenitors.
Blood
81
1993
610
347
Li
CL
Johnson
GR
Stem cell factor enhances the survival but not the self-renewal of murine hematopoietic long-term repopulating cells.
Blood
84
1994
408
348
Keller
JR
Ortiz
M
Ruscetti
FW
Steel factor (c-kit ligand) promotes the survival of hematopoietic stem/progenitor cells in the absence of cell division.
Blood
86
1995
1757
349
Brandt
JE
Bhalla
K
Hoffman
R
Effects of interleukin-3 and c-kit ligand on the survival of various classes of human hematopoietic progenitor cells.
Blood
83
1994
1507
350
Hong
DS
Huss
R
Beckham
C
Hoy
CA
Storb
R
Deeg
HJ
Major histocompatibility complex class II-mediated inhibition of hemopoiesis in vitro and in vivo is abrogated by c-kit ligand.
Transplant Proc
27
1995
642
351
Veiby
OP
Jacobsen
FW
Cui
L
Lyman
SD
Jacobsen
SEW
The flt3 ligand promotes the survival of primitive hemopoietic progenitor cells with myeloid as well as B lymphoid potential. Suppression of apoptosis and counteraction by TNF-alpha and TGF-beta.
J Immunol
157
1996
2953
352
Takahira H, Lyman SD, Broxmeyer HE : Flt3 ligand prolongs survival of CD34+++ human umbilical cord blood myeloid progenitors in serum-depleted culture medium. Ann Hematol 72:131, 1996
353
Keller
JR
Jacobsen
SEW
Dubois
CM
Hestdal
K
Ruscetti
FW
Transforming growth factor-beta: A bidirectional regulator of hematopoietic cell growth.
Int J Cell Cloning
10
1992
2
354
McNiece
IK
Bertoncello
I
Keller
JR
Ruscetti
FW
Hartley
CA
Zsebo
KM
Transforming growth factor beta inhibits the action of stem cell factor on mouse and human hematopoietic progenitors.
Int J Cell Cloning
10
1992
80
355
Ohishi
K
Katayama
N
Itoh
R
Mahmud
N
Miwa
H
Kita
K
Minami
N
Shirakawa
S
Lyman
SD
Shiku
H
Accelerated cell-cycling of hematopoietic progenitors by the flt3 ligand that is modulated by transforming growth factor-β.
Blood
87
1996
1718
356
Jacobsen
SEW
Veiby
OP
Myklebust
J
Okkenhaug
C
Lyman
SD
Ability of flt3 ligand to stimulate the in vitro growth of primitive murine hematopoietic progenitors is potently and directly inhibited by transforming growth factor-β and tumor necrosis factor-α.
Blood
87
1996
5016
357
Jacobsen
SEW
Jacobsen
FW
Fahlman
C
Rusten
LS
TNF-alpha, the great imitator: Role of p55 and p75 TNF receptors in hematopoiesis.
Stem Cells
12
1994
111
358
Jacobsen
FW
Veiby
OP
Stokke
T
Jacobsen
SEW
TNF-alpha bidirectionally modulates the viability of primitive murine hematopoietic progenitor cells in vitro.
J Immunol
157
1996
1193
359
Jacobsen
SEW
Veiby
OP
Myklebust
J
Okkenhaug
C
Lyman
SD
Ability of flt3 ligand to stimulate the in vitro growth of primitive murine hematopoietic progenitors is potently and directly inhibited by transforming growth factor-β and tumor necrosis factor-α.
Blood
87
1996
5016
360
Rusten
LS
Smeland
EB
Jacobsen
FW
Lien
E
Lesslauer
W
Loetscher
H
Dubois
CM
Jacobsen
SEW
Tumor necrosis factor-alpha inhibits stem cell factor-induced proliferation of human bone marrow progenitor cells in vitro. Role of p55 and p75 tumor necrosis factor receptors.
J Clin Invest
94
1994
165
361
Kurosawa
K
Miyazawa
K
Gotoh
A
Katagiri
T
Nishimaki
J
Ashman
LK
Toyama
K
Immobilized anti-KIT monoclonal antibody induces ligand-independent dimerization and activation of Steel factor receptor: Biologic similarity with membrane-bound form of Steel factor rather than its soluble form.
Blood
87
1996
2235
362
Miyazawa
K
Williams
DA
Gotoh
A
Nishimaki
J
Broxmeyer
HE
Toyama
K
Membrane-bound Steel factor induces more persistent tyrosine kinase activation and longer life span of c-kit gene-encoded protein than its soluble form.
Blood
85
1995
641
363
Gurney
AL
Carver-Moore
K
de Sauvage
FJ
Moore
MW
Thrombocytopenia in c-mpl-deficient mice.
Science
265
1994
1445
364
Carver-Moore
K
Broxmeyer
HE
Luoh
S-M
Cooper
S
Peng
J
Burstein
SA
Moore
MW
de Sauvage
FJ
Low levels of erythroid and myeloid progenitors in thrombopoietin- and c-mpl-deficient mice.
Blood
88
1996
803
365
Miller
CL
Rebel
VI
Helgason
CD
Lansdorp
PM
Eaves
CJ
Impaired Steel factor responsiveness differentially affects the detection and long-term maintenance of fetal liver hematopoietic stem cells in vivo.
Blood
89
1997
1214
366
Yan
XQ
Briddell
R
Hartley
C
Stoney
G
Samal
B
McNiece
I
Mobilization of long-term hematopoietic reconstituting cells in mice by the combination of stem cell factor plus granulocyte colony-stimulating factor.
Blood
84
1994
795
367
Neta
R
Williams
D
Selzer
F
Abrams
J
Inhibition of c-kit ligand/Steel factor by antibodies reduces survival of lethally irradiated mice.
Blood
81
1993
324
368
Zsebo
KM
Smith
KA
Hartley
CA
Greenblatt
M
Cooke
K
Rich
W
McNiece
IK
Radioprotection of mice by recombinant rat stem cell factor.
Proc Natl Acad Sci USA
89
1992
9464
369
Patchen
ML
Fischer
R
Schmauder-Chock
EA
Williams
DE
Mast cell growth factor enhances multilineage hematopoietic recovery in vivo following radiation-induced aplasia.
Exp Hematol
22
1994
31
370
Schuening
FG
Appelbaum
FR
Deeg
HJ
Sullivan-Pepe
M
Graham
TC
Hackman
R
Zsebo
KM
Storb
R
Effects of recombinant canine stem cell factor, a c-kit ligand, and recombinant granulocyte colony-stimulating factor on hematopoietic recovery after otherwise lethal total body irradiation.
Blood
81
1993
20
371
Molineux
G
Migdalska
A
Szmitkowski
M
Zsebo
K
Dexter
TM
The effects on hematopoiesis of recombinant stem cell factor (ligand for c-kit) administered in vivo to mice either alone or in combination with granulocyte colony-stimulating factor.
Blood
78
1991
961
372
Fleming
WH
Alpern
EJ
Uchida
N
Ikuta
K
Weissman
IL
Steel factor influences the distribution and activity of murine hematopoietic stem cells in vivo.
Proc Natl Acad Sci USA
90
1993
3760
373
Briddell
RA
Hartley
CA
Smith
KA
McNiece
IK
Recombinant rat stem cell factor synergizes with recombinant human granulocyte colony-stimulating factor in vivo in mice to mobilize peripheral blood progenitor cells that have enhanced repopulating potential.
Blood
82
1993
1720
374
Yan
XQ
Hartley
C
McElroy
P
Chang
A
McCrea
C
McNiece
I
Peripheral blood progenitor cells mobilized by recombinant human granulocyte colony-stimulating factor plus recombinant rat stem cell factor contain long-term engrafting cells capable of cellular proliferation for more than two years as shown by serial transplantation in mice.
Blood
85
1995
2303
375
Bodine
DM
Seidel
NE
Zsebo
KM
Orlic
D
In vivo administration of stem cell factor to mice increases the absolute number of pluripotent hematopoietic stem cells.
Blood
82
1993
445
376
de Revel
T
Appelbaum
FR
Storb
R
Schuening
F
Nash
R
Deeg
J
McNiece
I
Andrews
R
Graham
T
Effects of granulocyte colony-stimulating factor and stem cell factor, alone and in combination, on the mobilization of peripheral blood cells that engraft lethally irradiated dogs.
Blood
83
1994
3795
377
Andrews
RG
Briddell
RA
Knitter
GH
Rowley
SD
Appelbaum
FR
McNiece
IK
Rapid engraftment by peripheral blood progenitor cells mobilized by recombinant human stem cell factor and recombinant human granulocyte colony-stimulating factor in nonhuman primates.
Blood
85
1995
15
378
Andrews
RG
Briddell
RA
Knitter
GH
Opie
T
Bronsden
M
Myerson
D
Appelbaum
FR
McNiece
IK
In vivo synergy between recombinant human stem cell factor and recombinant human granulocyte colony-stimulating factor in baboons: Enhanced circulation of progenitor cells.
Blood
84
1994
800
379
Andrews
RG
Bensinger
WI
Knitter
GH
Bartelmez
SH
Longin
K
Bernstein
ID
Appelbaum
FR
Zsebo
KM
The ligand for c-kit, stem cell factor, stimulates the circulation of cells that engraft lethally irradiated baboons.
Blood
80
1992
2715
380
Tong
J
Gordon
MS
Srour
EF
Cooper
RJ
Orazi
A
McNiece
I
Hoffman
R
In vivo administration of recombinant methionyl human stem cell factor expands the number of human marrow hematopoietic stem cells.
Blood
82
1993
784
381
McNiece
IK
Briddell
RA
Yan
XQ
Hartley
CA
Gringeri
A
Foote
MA
Andrews
RG
The role of stem cell factor in mobilization of peripheral blood progenitor cells.
Leuk Lymphoma
15
1994
405
382
Bodine
DM
Seidel
NE
Orlic
D
Bone marrow collected 14 days after in vivo administration of granulocyte colony-stimulating factor and stem cell factor to mice has 10-fold more repopulating ability than untreated bone marrow.
Blood
88
1996
89
383
Dunbar
CE
Seidel
NE
Doren
S
Sellers
S
Cline
AP
Metzger
ME
Agricola
BA
Donahue
RE
Bodine
DM
Improved retroviral gene transfer into murine and Rhesus peripheral blood or bone marrow repopulating cells primed in vivo with stem cell factor and granulocyte colony-stimulating factor.
Proc Natl Acad Sci USA
93
1996
11871
384
Brasel
K
McKenna
HJ
Morrissey
PJ
Charrier
K
Morris
AE
Lee
CC
Williams
DE
Lyman
SD
Hematologic effects of flt3 ligand in vivo in mice.
Blood
88
1996
2004
385
Brasel
K
McKenna
HJ
Charrier
K
Morrissey
P
Williams
DE
Lyman
SD
Flt3 ligand synergizes with granulocyte-macrophage colony-stimulating factor or granulocyte colony-stimulating factor to mobilize hematopoietic progenitor cells into the peripheral blood of mice.
Blood
90
1997
3781
386
(abstr, suppl 1)
Winton
EF
Bucur
SZ
Bond
LD
Hegwood
AJ
Hillyer
CD
Holland
HK
Williams
DE
McClure
HM
Troutt
AB
Lyman
SD
Recombinant human (rh) Flt3 ligand plus rhGM-CSF or rhG-CSF causes a marked CD34+ cell mobilization to blood in rhesus monkeys.
Blood
88
1996
642a
387
Langley
KE
Bennett
LG
Wypych
J
Yancik
SA
Liu
X-D
Westcott
KR
Chang
DG
Smith
KA
Zsebo
KM
Soluble stem cell factor in human serum.
Blood
81
1993
656
388
Abkowitz
JL
Hume
H
Yancik
SA
Bennett
LG
Matsumoto
AM
Stem cell factor serum levels may not be clinically relevant.
Blood
87
1996
4017
389
Lyman
SD
Seaberg
M
Hanna
R
Zappone
J
Brasel
K
Abkowitz
JL
Prchal
JT
Schultz
JC
Shahidi
NT
Plasma/serum levels of flt3 ligand are low in normal individuals and highly elevated in patients with Fanconi anemia and acquired aplastic anemia.
Blood
86
1995
4091
390
Wodnar-Filipowicz
A
Lyman
SD
Gratwohl
A
Tichelli
A
Speck
B
Nissen
C
Flt3 ligand level reflects hematopoietic progenitor cell function in multilineage bone marrow failure.
Blood
88
1996
4493
391
(abstr, suppl 1)
Zwierzina
H
Torok-Storb
B
Rollinger-Holzinger
I
Anderson
JE
Nuessler
V
Lyman
SD
Serum levels of flt3 ligand are associated with disease stage in patients with myelodysplastic syndrome.
Blood
88
1996
99a
392
Lebsack ME, Hoek JA, Maraskovsky E, McKenna HJ: FLT3 ligand induces stem and dendritic cell mobilization in healthy volunteers. International Society for Hematotherapy and Graft Engineering Meeting. Bordeaux, France, May 31-June 3, 1997
393
(abstr, suppl 1)
Winton
EF
Bucur
SZ
Bray
RA
Toba
K
Williams
DE
McClure
HM
Lyman
SD
The hematopoietic effects of recombinant human (rh) Flt3 ligand administered to non-human primates.
Blood
86
1995
424a
394
Bodine
DM
Seidel
NE
Gale
MS
Nienhuis
AW
Orlic
D
Efficient retrovirus transduction of mouse pluripotent hematopoietic stem cells mobilized into the peripheral blood by treatment with granulocyte colony-stimulating factor and stem cell factor.
Blood
84
1994
1482
395
Kohn
DB
Weinberg
KI
Nolta
JA
Heiss
LN
Lenarsky
C
Crooks
GM
Hanley
ME
Annett
G
Brooks
JS
el-Khoureiy
A
Lawrence
K
Wells
S
Moen
RC
Bastian
J
Williams-Herman
DE
Elder
M
Wara
D
Bowen
T
Hershfield
MS
Mullen
CA
Blaese
RM
Parkman
R
Engraftment of gene-modified umbilical cord blood cells in neonates with adenosine deaminase deficiency.
Nat Med
1
1995
1017
396
Dick
JE
Kamel-Reid
S
Murdoch
B
Doedens
M
Gene transfer into normal human hematopoietic cells using in vitro and in vivo assays.
Blood
78
1991
624
397
Nolta
JA
Dao
MA
Wells
S
Smogorzewska
EM
Kohn
DB
Transduction of pluripotent human hematopoietic stem cells demonstrated by clonal analysis after engraftment in immune-deficient mice.
Proc Natl Acad Sci USA
93
1996
2414
398
Williams
DA
Ex vivo expansion of hematopoietic stem and progenitor cells—Robbing Peter to pay Paul?
Blood
81
1993
3169
399
Lange
W
Henschler
R
Mertelsmann
R
Biological and clinical advances in stem cell expansion.
Leukemia
10
1996
943
400
Emerson
SG
Ex vivo expansion of hematopoietic precursors, progenitors, and stem cells: The next generation of cellular therapeutics.
Blood
87
1996
3082
401
Rill
DR
Santana
VM
Roberts
WM
Nilson
T
Bowman
LC
Krance
RA
Heslop
HE
Moen
RC
Ihle
JN
Brenner
MK
Direct demonstration that autologous bone marrow transplantation for solid tumors can return a multiplicity of tumorigenic cells.
Blood
84
1994
380
402
Deisseroth
AB
Zu
Z
Claxton
D
Hanania
EG
Fu
S
Ellerson
D
Goldberg
L
Thomas
M
Janicek
K
Anderson
WF
Hester
J
Korbling
M
Durrett
A
Moen
R
Berenson
R
Heimfeld
S
Hamer
J
Calvert
L
Tibbits
P
Talpaz
M
Kantarjian
H
Champlin
R
Reading
C
Genetic marking shows that Ph+ cells present in autologous transplants of chronic myelogenous leukemia (CML) contribute to relapse after autologous bone marrow in CML.
Blood
83
1994
3068
403
Muench
MO
Firpo
MT
Moore
MA
Bone marrow transplantation with interleukin-1 plus kit-ligand ex vivo expanded bone marrow accelerates hematopoietic reconstitution in mice without the loss of stem cell lineage and proliferative potential.
Blood
81
1993
3463
404
Yonemura
Y
Ku
H
Lyman
SD
Ogawa
M
In vitro expansion of hematopoietic progenitors and maintenance of stem cells: Comparison between flt3/flk-2 ligand and kit ligand.
Blood
89
1997
1915
405
Rebel
VI
Dragowska
W
Eaves
CJ
Humphries
RK
Lansdorp
PM
Amplification of Sca-1+ Lin− WGA+ cells in serum-free cultures containing steel factor, interleukin-6, and erythropoietin with maintenance of cells with long-term in vivo reconstituting potential.
Blood
83
1994
128
406
Holyoake
TL
Freshney
MG
McNair
L
Parker
AN
McKay
PJ
Steward
WP
Fitzsimons
E
Graham
GJ
Pragnell
IB
Ex vivo expansion with stem cell factor and interleukin-11 augments both short-term recovery posttransplant and the ability to serially transplant marrow.
Blood
87
1996
4589
407
Yonemura
Y
Ku
H
Hirayama
F
Souza
LM
Ogawa
M
Interleukin 3 or interleukin 1 abrogates the reconstituting ability of hematopoietic stem cells.
Proc Natl Acad Sci USA
93
1996
4040
408
Haylock
DN
To
LB
Dowse
TL
Juttner
CA
Simmons
PJ
Ex vivo expansion and maturation of peripheral blood CD34+ cells into the myeloid lineage.
Blood
80
1992
1405
409
Henschler
R
Brugger
W
Luft
T
Frey
T
Mertelsmann
R
Kanz
L
Maintenance of transplantation potential in ex vivo expanded CD34(+)-selected human peripheral blood progenitor cells.
Blood
84
1994
2898
410
Srour
EF
Brandt
JE
Briddell
RA
Grigsby
S
Leemhuis
T
Hoffman
R
Long-term generation and expansion of human primitive hematopoietic progenitor cells in vitro.
Blood
81
1993
661
411
Petzer
AL
Hogge
DE
Landsdorp
PM
Reid
DS
Eaves
CJ
Self-renewal of primitive human hematopoietic cells (long-term-culture-initiating cells) in vitro and their expansion in defined medium.
Proc Natl Acad Sci USA
93
1996
1470
412
Karlsson
S
Treatment of genetic defects in hematopoietic cell function by gene transfer.
Blood
78
1991
2481
413
Brenner
MK
Rill
DR
Holladay
MS
Heslop
HE
Moen
RC
Buschle
M
Krance
RA
Santana
VM
Anderson
WF
Ihle
JN
Gene marking to determine whether autologous marrow infusion restores long-term haemopoiesis in cancer patients.
Lancet
342
1993
1134
414
Koller
MR
Oxender
M
Brott
DA
Palsson
flt-3 ligand is more potent than c-kit ligand for the synergistic stimulation of ex vivo hematopoietic cell expansion.
J Hematother
5
1996
449
415
Miller
AD
Human gene therapy comes of age.
Nature
357
1992
455
416
Kohn
DB
The current status of gene therapy using hematopoietic stem cells.
Curr Opin Pediatr
7
1995
56
417
Williams
DA
Lemischka
IR
Nathan
DG
Mulligan
RC
Introduction of new genetic material into pluripotent haematopoietic stem cells of the mouse.
Nature
310
1984
476
418
Bodine
DM
Karlsson
S
Nienhuis
AW
Combination of interleukins 3 and 6 preserves stem cell function in culture and enhances retrovirus-mediated gene transfer into hematopoietic stem cells.
Proc Natl Acad Sci USA
86
1989
8897
419
Luskey
BD
Rosenblatt
M
Zsebo
K
Williams
DA
Stem cell factor, interleukin-3, and interleukin-6 promote retroviral-mediated gene transfer into murine hematopoietic stem cells.
Blood
80
1992
396
420
Fraser
CC
Eaves
CJ
Szilvassy
SJ
Humphries
RK
Expansion in vitro of retrovirally marked totipotent hematopoietic stem cells.
Blood
76
1990
1071
421
Correll
PH
Colilla
S
Dave
HP
Karlsson
S
High levels of human glucocerebrosidase activity in macrophages of long-term reconstituted mice after retroviral infection of hematopoietic stem cells.
Blood
80
1992
331
422
Cairo
MS
Law
P
van de Ven
C
Plunkett
JM
Williams
D
Ishizawa
L
Gee
A
The in vitro effects of stem cell factor and PIXY321 on myeloid progenitor formation (CFU-GM) from immunomagnetic separated CD34+ cord blood.
Pediatr Res
32
1992
277
423
Cassel
A
Cottler-Fox
M
Doren
S
Dunbar
CE
Retroviral-mediated gene transfer into CD34-enriched human peripheral blood stem cells.
Exp Hematol
21
1993
585
424
Dunbar
CE
Cottler-Fox
M
O'Shaughnessy
JA
Doren
S
Carter
C
Berenson
R
Brown
S
Moen
RC
Greenblatt
J
Stewart
FM
Leitman
SF
Wilson
WH
Cowan
K
Young
NS
Nienhuis
AW
Retrovirally marked CD34-enriched peripheral blood and bone marrow cells contribute to long-term engraftment after autologous transplantation.
Blood
85
1995
3048
425
Nolta
JA
Crooks
GM
Overell
RW
Williams
DE
Kohn
DB
Retroviral vector-mediated gene transfer into primitive human hematopoietic progenitor cells: Effects of mast cell growth factor (MGF) combined with other cytokines.
Exp Hematol
20
1992
1065
426
Nolta
JA
Smogorzewska
EM
Kohn
DB
Analysis of optimal conditions for retroviral-mediated transduction of primitive human hematopoietic cells.
Blood
86
1995
101
427
Schwarzenberger
P
Spence
SE
Gooya
JM
Michiel
D
Curiel
DT
Ruscetti
FW
Keller
JR
Targeted gene transfer to human hematopoietic progenitor cell lines through the c-kit receptor.
Blood
87
1996
472
428
Mayordomo
JI
Zorina
T
Storkus
WJ
Zitvogel
L
Celluzzi
C
Falo
LD
Melief
CJ
Ildstad
ST
Kast
WM
Deleo
AB
Lotze
MT
Bone marrow-derived dendritic cells pulsed with synthetic tumour peptides elicit protective and therapeutic antitumour immunity.
Nat Med
1
1995
1297
429
Thomson
AW
Lu
L
Murase
N
Demetris
AJ
Rao
AS
Starzl
TE
Microchimerism, dendritic cell progenitors and transplantation tolerance.
Stem Cells
13
1995
622
430
Young
JW
Inaba
K
Dendritic cells as adjuvants for class I major histocompatibility complex-restricted antitumor immunity.
J Exp Med
183
1996
7
431
Santiago-Schwarz
F
Rappa
DA
Laky
K
Carsons
SE
Stem cell factor augments tumor necrosis factor-granulocyte-macrophage colony-stimulating factor-mediated dendritic cell hematopoiesis.
Stem Cells
13
1995
186
432
Lynch
DH
Andreasen
A
Maraskovsky
E
Whitmore
J
Miller
RE
Schuh
JCL
Flt3 ligand induces tumor regression and anti-tumor immune responses in vivo.
Nat Med
3
1997
625
433
(abstr, suppl 1)
Chen
K
Braun
SE
Lyman
SD
Broxmeyer
HE
Cornetta
K
Soluble and membrane bound isoforms of FLT3-ligand induce antitumor immunity in vivo.
Blood
88
1996
274a
434
Chen
K
Braun
S
Lyman
S
Fan
Y
Traycoff
CM
Wiebke
EA
Gaddy
J
Sledge
G
Broxmeyer
HE
Cornetta
K
Antitumor activity and immunotherapeutic properties of Flt3-ligand in a murine breast cancer model.
Cancer Res
57
1997
3511
Sign in via your Institution