Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

Oncogene (2002) 21, 3377 ± 3390

ã 2002 Nature Publishing Group All rights reserved 0950 ± 9232/02 $25.00
www.nature.com/onc

Transcriptional regulation of granulocyte and monocyte development

Alan D Friedman*,1
1
Division of Pediatric Oncology, Johns Hopkins University, Baltimore, Maryland, MD 21231, USA

Granulocytes and monocytes develop from a common granulocyte/macrophage progenitors (GMPs), charac-
myeloid progenitor. Early granulopoiesis requires the C/ terized by expression of CD34 and Fcg Receptor(II/III)
EBPa, PU.1, RAR, CBF, and c-Myb transcription (Akashi et al., 2000; Traver et al., 2001). These
factors, and terminal neutrophil di€erentiation is progenitors in turn give rise to granulocyte-, mono-
dependent upon C/EBPe, PU.1, Sp1, CDP, and cyte-, and granulocyte/monocyte-colony forming units
HoxA10. Monopoiesis can be induced by Maf-B, c- (CFU-G, CFU-M, and CFU-GM). Granulocyte-Col-
Jun, or Egr-1 and is dependent upon PU.1, Sp1, and ony Stimulating Factor (G-CSF) enables the formation
ICSBP. Signals eminating from cytokine receptors of CFU-G in methylcellulose cultures, Macrophage-
modulate factor activities but do not determine cell Colony Stimulating Factor (M-CSF, CSF-1) enables
fates. Orchestration of the myeloid developmental the growth of CFU-M, and Granulocyte/Macrophage-
program is achieved via cooperative gene regulation, Colony Stimulating Factor (GM-CSF) or Interleukin-3
via synergistic and inhibitory protein ± protein interac- (IL-3) enable the proliferation of CFU-G, CFU-M,
tions, via promoter auto-regulation and cross-regulation, and CFU-GM. In addition to these features of normal
via regulation of factor levels, and via induction of cell cells, the ®nding that a common subset of acute
cycle arrest: For example, c-Myb and C/EBPa myeloid leukemia has both a granulocytic and
cooperate to activate the mim-1 and NE promoters, monocytic cellular component further underscores the
PU.1, C/EBPa, and CBF, regulate the NE, MPO, and close developmental relationship of neutrophils and
M-CSF Receptor genes. PU.1:GATA-1 interaction and monocytes. Fetal liver and adult marrow also contain
C/EBP suppression of FOG transcription inhibits progenitors capable of producing B-lymphoid and
erythroid and megakaryocyte gene expression. c-Jun:- macrophage cells in single colonies, re¯ecting both
PU.1, ICSBP:PU.1, and perhaps Maf:Jun complexes the plasticity of pluripotent stem cell commitment and
induce monocytic genes. PU.1 and C/EBPa activate the relatedness of these lineages (Lacaud et al., 1998;
their own promoters, C/EBPa rapidly induces PU.1 and Montecino-Rodriguez et al., 2001; Traver et al., 2001).
C/EBPe RNA expression, and RARa activates the C/ Monocyte progenitors not only develop into macro-
EBPe promoter. Higher levels of PU.1 are required for phages but are also capable of osteoclast di€erentiation
monopoiesis than for B-lymphopoiesis, and higher C/ (Roodman, 1996), and GMPs share with common
EBP levels may favor granulopoiesis over monopoiesis. lymphoid progenitors (CLPs) the capacity to generate
CBF and c-Myb stimulate proliferation whereas C/EBPa myeloid dendritic cells (Manz et al., 2001).
induces a G1/S arrest; cell cycle arrest is required for Neutrophil maturation proceeds from the myeloblast
terminal myelopoiesis, perhaps due to expression of p53 to the promyelocyte, when primary granules become
or hypo-phosphorylated Rb. apparent, to the myelocyte, when cell division ceases
Oncogene (2002) 21, 3377 ± 3390. DOI: 10.1038/sj/ and secondary or speci®c granules begin to appear, to
onc/1205324 the metamyelocyte, band, and ®nally the neutrophil,
which has a three-lobed nucleus and tertiary granules.
Keywords: granulocyte; monocyte; di€erentiation; Early markers of granulopoiesis include the G-CSF
C/EBP; PU.1 Receptor, the CD33 and CD13 surface markers, and
primary granule components, such as myeloperoxidase
(MPO), neutrophil elastase (NE), myeloblastin (MBN),
Introduction lysozyme and avian mim-1. Oxidation system compo-
nents, such as the gp91-phox cytochrome heavy chain,
Polymorphonuclear and mononuclear phagocytes func- are expressed from the myelocyte stage onward and
tion in host defense against infections and are capable later markers of neutrophil development include
of recognizing, ingesting, and destroying foreign secondary granule components, such as lactoferrin
materials and organisms. Pluripotent hematopoietic (LF) and neutrophil gelatinase (NG), and the Gr-1
stem cells in fetal liver or in adult marrow give rise to surface marker.
Monopoiesis proceeds from the monoblast to the
circulating monocyte, which matures without prolifera-
*Correspondence: AD Friedman, Johns Hopkins University, Cancer
tion to the tissue macrophage. Early markers of this
Research Building, Room 253, 1650 Orleans Street, Baltimore, lineage include the M-CSF Receptor, lysozyme, and
Maryland, MD 21231; E-mail: adfrdman@jhmi.edu the Fcg Receptor(II/III), and latter markers include
Regulation of granulopoiesis and monopoiesis
AD Friedman
3378
gp91-phox, and several surface proteins: Macrosialin, Table 1 Transcription factors regulating granulocytic or monocytic
scavenger receptor (SR), F4/80, CD14, CD11b, and genes
CD18. Fcg Receptor(II/III) is also expressed on Gene Transcription factors
myeloid progenitors (Carlsson et al., 1995); the Immature granulocytes
CD11b:CD18 integrin is also expressed on neutrophils, mim-1 C/EBP's, c-Myb
B-lymphocytes, and some T-cell subsets (Christensen et Myeloperoxidase (MPO) C/EBPs, PU.1, CBF, c-Myb, CDP/HoxA10
al., 2001); and CD14 is also expressed in hepatocytes Neutrophil elastase (NE) C/EBPs, PU.1/GABP, CBF, c-Myb, Sp1
Myeloblastin (MBN) C/EBPs, PU.1, c-Myb
(Hetherington et al., 1999). Phagocytic capacity is an G-CSF receptor C/EBPs, PU.1
additional hallmark of maturing monocytes. GM-CSF receptor C/EBPs, PU.1
CD13 c-Myb, Ets-1 or Ets, c-Maf
Lysozyme C/EBPs, PU.1
Receptor signaling c-fes PU.1, Sp1
Mature granulocytes
gp91phox PU.1, ICSBP, CDP, HoxA10
We have recently reviewed how G-CSF Receptor Lactoferrin (LF) C/EBPs, Sp1, CDP
signals might modulate granulopoiesis (Ward et al., Immature monocytes
2000). Redundancy of receptor signaling is evident M-CSF receptor C/EBPs, CBF, PU.1, c-Jun
Lysozyme C/EBPs, PU.1
from the ®nding that G-CSF (7/7), G-CSF; GM- Mature monocytes
CSF (7/7; 7/7); G-CSFR (7/7), or G-CSFR; IL- Macrosialin PU.1, c-Jun
6 Receptor (7/7; 7/7) mice have only about a Scavenger receptor PU.1, c-Jun
twofold reduction in marrow neutrophils (Lieschke et CD14 C/EBPs, Sp1
al., 1994; Liu et al., 1996a, 1997; Seymour et al., 1997). gp91phox PU.1, ICSBP, CDP, HoxA10
CD11b PU.1, Sp1
Bcl-2 did not replace G-CSF receptor signals during CD18 PU.1, GABP, Sp1
32D cl3 maturation-MPO and Cathepsin G induction
did not occur, but nuclear morphologic changes were
observed (Rodel and Link, 1996). Thus, G-CSF
receptor signals provide more than a survival function insights gained from characterizing transcription factor
in this setting. Similarly, G-CSF receptor signals expression patterns, the phenotypes of knockout mice,
cooperated with exogenous C/EBPa to allow induction and the e€ects of factor over-expression.
of MPO and NE in Ba/F3 lymphoid cells and were The promoter of the avian mim-1 promoter is
required for induction of C/EBPe in 32D cl3 cells activated cooperatively by C/EBPs and c-Myb in
(Wang et al., 2001; Nakajima and Ihle, 2001). Also, immature granulocytic cells (Ness et al., 1993). This
introduction of exogenous GM-CSF or IL-2 receptors section will not refer to individual C/EBPs, as C/EBP
allows CLPs to be redirected to the granulocyte and family members bind a common DNA motif and often
monocyte lineages (Kondo et al., 2000). activate reporter constructs similarly in transient
M-CSFR signaling has also recently been reviewed assays. The MPO promoter region is activated
(Bourette and Rohrschneider, 2000). Transgenic ex- cooperatively by CBF and c-Myb (Suzow and Fried-
pression of bcl-2 in monocytes enabled M-CSF (7/7) man, 1993; Nuchprayoon et al., 1994; Britos-Bray and
mice to develop macrophages (Lagasse and Weissman, Friedman, 1997), and its distal enhancer is regulated by
1997). While this ®nding precludes an absolute C/EBPs and PU.1 (Ford et al., 1996). The NE
requirement for M-CSF Receptor signals in macro- promoter is regulated cooperatively by C/EBPs, PU.1,
phage development, redundant signals available from a and c-Myb; the murine but not the human NE
subset of other cytokine receptors may be necessary. promoter is activated weakly by CBF; GABP can
The ability of phorbol esters to induce monocytic substitute for PU.1 to cooperatively active the NE
di€erentiation of hematopoietic cell lines via activation promoter; and a 76 kb NE enhancer is activated by
of protein kinase Ca (PKCa) or PKCd suggests that Sp1 and an Ets family member other than PU.1
this signaling pathway is important in normal mono- (Nuchprayoon et al., 1994; 1997, 1999; Oelgeschlager et
poiesis (Mischak et al., 1993). Overall, with possible al., 1996). MBN and azurocidin are serine proteases
notable exceptions, signals eminating from cytokine highly related to NE. Their promoters contain
receptors appear to modulate but not determine sequences elements homologous to the NE elements
myeloid di€erentiation. which bind C/EBPs, PU.1, and c-Myb (Friedman,
1996a), and indeed these proteins regulate the MBN
promoter (Lutz et al., 2001). The promoters of both
Regulation of gene expression the G-CSF and GM ± CSF receptor genes are activated
by C/EBPs and PU.1 (Hohaus et al., 1995; Smith et al.,
Di€erentiation is de®ned by gene expression patterns, 1996). The CD13 gene is activated directly by c-Myb
and so regulatory factors must act either directly or and Ets-1 or Ets-2, and c-Maf indirectly inhibits the
indirectly to induce the transcription of lineage transcription of this gene by complexing with c-Myb in
markers. Therefore, studies investigating the regulation immature but not maturing granulocyte precursors
of granulocyte- and monocyte-speci®c genes, as (Shapiro, 1995; Hegde et al., 1998). The avian
summarized in Table 1, will be reviewed initially. The lysozyme gene 72.7 kb enhancer is regulated by C/
subsequent sections will then describe additional EBPs and PU.1, and its 76.1 kb enhancer is activated
Oncogene
Regulation of granulopoiesis and monopoiesis
AD Friedman
3379
by C/EBPs (Ahne and Stratling, 1994; Goethe and mature monocytes. The next section will describe
Loc, 1994; Faust et al., 1999). Consistent with these additional evidence which con®rms the importance of
results, we recently found that activation of exogenous these factors, as well as evidence implicating
C/EBPa-ER with estradiol in the murine 32D cl3 additional transcription factors, in the regulation of
myeloblast cell line rapidly induces lysozyme RNA granulocyte and monocyte development. The subse-
expression in the presence of cycloheximide but not quent section will then address the question of how
actinomycin D (Wang and Friedman, 2002). Finally, these factors orchestrate the myeloid developmental
the c-fes tyrosine kinase promoter region is regulated program.
by PU.1 and Sp1 in immature granulocytic and
monocytic cells (Heydemann et al., 1996).
In maturing neutrophils, the gp91-phox promoter
Transcription factors
region is activated by the PU.1:ICSBP dimer and is
repressed by both CDP and HoxA10 ± the levels of
C/EBPs
the latter factors decrease during terminal neutrophil
and monocyte di€erentiation (Skalnik et al., 1991; The C/EBPs homo- and hetero-dimerize via their C-
Eklund and Kakar, 1999; Eklund et al., 2000). The LF terminal leucine zipper domains and bind DNA as
promoter is activated by C/EBPs and Sp1 and is dimers via the adjacent basic regions (Landschulz et
repressed by CDP (Khanna-Gupta et al., 1997, 2000). al., 1989). The C/EBP consensus binding site is 5'-T(T/
CDP represses both the gp91-phox and LF promoters G)NNGNAA(T/G)-3'. C/EBPa, C/EBPb, and C/EBPd
via interaction with sites upstream of activator binding have N-terminal trans-activation domains, and transla-
sites (Luo and Skalnik, 1996; Khanna-Gupta et al., tion initiation from internal methionines produces
1997), and CDP also represses the gp91-phox promoter truncated, dominant-inhibitory polypeptides which
via a site 790 which overlaps an activator-binding site retain the bZIP domain (Friedman et al., 1989,
(Catt et al., 1999). HoxA10 interacts with an AT-rich Friedman and McKnight, 1990; Descombes and
element located just upstream of the 5'-CCAAT Schibler, 1991; Calkhoven et al., 2000). C/EBPe has
sequence at 790, the binding site for CDP, and so both a trans-activation and a trans-repression domain
like CDP may repress gp91-phox transcription, in part, (Williamson et al., 1998), and C/EBPg and CHOP are
via competition with activators (Eklund et al., 2000). dominant-inhibitory due to their ability to dimerize
The MPO promoter contains a potent repressor with other C/EBPs and their lack of intact basic
element active in 32D cl3 cells which has been mapped regions (Habener and Ron, 1992; Cooper et al., 1995).
to a 28 bp segment which includes a 5'-GAAATC Trans-activation by C/EBPa may be regulated via Ras-
sequence conserved in the homologous region of the induced phosphorylation of serine 248 (Behre et al.,
human MPO gene (Suzow and Friedman, 1993). 1999b).
Perhaps this element binds CDP or HoxA10, repressors Within hematopoiesis, full-length C/EBPa, C/EBPb,
thought to be more active in immature than mature and C/EBPd are predominantly expressed in the
granulocytes. granulocyte, monocyte, and eosinophil lineages (Scott
In immature monocytic cells, the M-CSF Receptor et al., 1992; Muller et al., 1995; Radomska et al., 1998).
promoter is activated synergistically by C/EBP, PU.1, C/EBPa is the isoform most prominently detected in
and CBF (Zhang et al., 1994a,b, 1996a; Petrovick et immature granulocytes (Scott et al., 1992; Hohaus et
al., 1998), and c-Jun activates the M-CSF receptor al., 1995), whereas C/EBPe is found in later-stage
gene indirectly, via its ability to interact with PU.1 granulocytes and in T-cells (Antonson et al., 1996).
(Bassuk and Leiden, 1995; Behre et al., 1999a). In cells CHOP is only detected in granulocytic cells subjected
di€erentiating into macrophages, the macrosialin and to stress, such as DNA-damage (Friedman, 1996b),
scavenger receptor promoters are activated coopera- and C/EBPg expression in this lineage is not well
tively by PU.1 and c-Jun, via binding sites separated by characterized.
33 or 135 bps, respectively (Moulton et al., 1994; Li et C/EBPa (7/7) mice lack neutrophils and eosino-
al., 1998). The CD14 promoter is activated by C/EBP phils, but retain monocytes, lymphocytes, erythroid
and Sp1 (Zhang et al., 1994c; Pan et al., 1999). And cells, and immature myeloblasts (Zhang et al., 1997).
®nally, PU.1 and Sp1 activate both the CD11b and Fetal liver cells from these mice lack Granulocyte-
CD18 promoters; optimal transcription of the latter Colony Stimulating Factor (G-CSF) Receptor RNA,
gene also requires GABP, which like PU.1 is a member consistent with the ability of C/EBPa to trans-activate
of the Ets family of transcription factors (Pahl et al., the G-CSF Receptor promoter (Smith et al., 1996).
1993; Chen et al., 1993; Rosmarin et al., 1995a,b, Induction of G-CSF or IL-6 Receptor cDNAs into C/
1998). EBPa (7/7) fetal liver cells restores their ability to
From these studies, C/EBPs and PU.1 emerge as generate neutrophils in vitro in response to G-CSF or
the most consistent regulators of genes expressed in IL-6 (Zhang et al., 1998). C/EBPb (7/7) mice retain
granulocytic and immature monocytic cells. PU.1 all of the hematopoietic lineages, and hematopoietic
also activates most genes expressed in mature defects in mice lacking C/EBPd or both C/EBPb and
monocytic cells, c-Myb and CBF contribute to the C/EBPd were not severe (Screpanti et al., 1995; Tanaka
activation of several genes in proliferating cells, and et al., 1995, 1997). C/EBPe (7/7) mice also retain
Sp1 activates several promoters in neutrophils and neutrophils, although they lack secondary granules
Oncogene
Regulation of granulopoiesis and monopoiesis
AD Friedman
3380
(Yamanaka et al., 1997; Chumakov et al., 1997; In addition to regulating di€erentiation in several
Lekstrom-Himes et al., 1999; Gombart et al., 2001). lineages, C/EBPa regulates proliferation directly.
Expression of C/EBPa in U937 cells led to the Decreased expression in proliferating versus quiescent
development of neutrophilic cells after 2 weeks, with hepatocytes and its ability to slow 3T3-L1 preadipocyte
expression of the mRNAs encoding G-CSF Receptor, proliferation provided the initial evidence that C/EBPs
lactoferrin, and neutrophil collagenase (Radomska et a€ect cell cycle progression (Friedman et al., 1989;
al., 1998). Introduction of C/EBPa into avian multi- Umek et al., 1991). Both C/EBPaWT-ER and KaER
potent progenitors also led to expression of myeloid inhibit progression from G1 to S phase in 32D cl3 cells
markers (Nerlov et al., 1998). Similarly, estradiol- (Wang et al., 1999; Friedman and Wang, 2000). These
mediated activation of C/EBPa-ER in 32D cl3 cells, a proteins may inhibit the G1/S transition via interaction
factor-dependent, diploid cell line, induced neutrophils with E2F (Timchenko et al., 1999; Slomiany et al.,
within 4 days, with induction of MPO, lactoferrin, and 2000; Johansen et al., 2001). A KaER variant with a
G-CSF Receptor RNAs (Wang et al., 1999). MPO defective leucine zipper domain does not slow 32D cl3
RNA was induced within 8 h of C/EBPa-ER activa- proliferation (Wang and Friedman, 2002). Perhaps C/
tion, but not in the presence of cycloheximide, EBPa dimers are required for interaction with E2F or
indicating the need for an additional C/EBP-dependent with another protein associated with the E2F com-
factor. A likely candidate is PU.1, as C/EBPa-ER plexes, or perhaps C/EBPa monomers interact with one
induced PU.1 RNA within 4 h, even in the presence of of these proteins via its leucine zipper. In fact, E2F
cycloheximide. C/EBPa-ER also rapidly induced the contains a leucine zipper, as noted (Slomiany et al.,
lysozyme and C/EBPe RNAs in the presence of 2000). C/EBPe also slows 32D cl3 proliferation
cycloheximide, but not actinomycin D, suggesting (Nakajima and Ihle, 2001). Inhibition of proliferation
direct regulation of the murine lysozyme and C/EBPe by C/EBPa or C/EBPe may contribute to granulopoi-
genes by C/EBPs (Wang and Friedman, 2002). The esis, as maneuvers which stimulate proliferation, such
signi®cance of C/EBPa regulation of PU.1 and C/EBPe as over-expression of c-Myb or cdk4, prevent induction
RNA expression will be discussed further below. As C/ of late markers (Bies et al., 1995; Lou et al., 2000).
EBPa is expressed in non-hematopoietic cells, it is Similarly, expression of p21WAF1/CIP1 or p27Kip1 in U937
interesting to speculate that early regulators of cells induces monocytic markers (Liu et al., 1996b). On
hematopoiesis, such as c-Myb, CBF, or GATA-2, the other hand, induction of p27Kip1 with mimosine
prime the PU.1 and C/EBPe genes for activation by C/ induced Lysozyme RNA but inhibited MPO RNA, an
EBPa. The ability of CEBPa to activate its own earlier di€erentiation marker, in 32D cl3 cells (Q Wang
promoter may also assist in committing progenitors to and AD Friedman, unpublished). The potential roles
the myeloid lineages (Christy et al., 1993). of p53 and hypo-phosphorylated Rb in terminal
The presence of several C/EBPs in myeloid cells myeloid di€erentiation will be discussed below.
suggests that family members might compensate in vivo
for the lack of a single isoform, just as GATA family
PU.1 and interacting proteins, ICSBP, c-Jun, and
members partially compensate for the lack of GATA-1
GATA-1
(Kulessa et al., 1995; Tsai et al., 1998). Indeed, as with
C/EBPa, C/EBPb, C/EBPd, or C/EBPe can induce PU.1 binds DNA as a monomer, via its C-terminal Ets
granulocytic di€erentiation in myeloblastic cell lines domain, to the consensus site 5'-AAAG(A/C/
(Park et al., 1999; Nakajima and Ihle, 2001; Wang and G)GGAAG-3' (Klemsz et al., 1990). The DNA-binding
Friedman, 2002), and C/EBPa, C/EBPb, or C/EBPd domain of PU.1 and its N-terminal, glutamine-rich
induced monocytic genes in P388 lymphoblasts (Hu et trans-activating domain were both required to rescue
al., 1998). Redundancy is also evident from the ability myelopoiesis in PU.1 (7/7) ES cells, whereas its
of C/EBPb to compensate for the loss of C/EBPa in acidic trans-activating domain was dispensable (Fisher
hepatocytes and granulocytes in vivo (Chen et al., 2000; et al., 1998).
Jones et al., 2002). In addition, compensatory e€ects Phosphorylation of serine 148 in PU.1 allows
may account for the ability of immortalized C/EBPa interaction with Pip, also known as Interferon
(7/7) progenitors to express high levels of G-CSF response factor-4 (IRF-4), an essential co-factor in B
Receptor in vitro, even though they could not respond cells (Eisenbeis et al., 1995). Pip or Interferon
to G-CSF in vivo (Collins et al., 2001). Global consensus sequence binding protein (ICSBP, IRF-8)
inhibition of C/EBP-regulated genes can be achieved can interact with PU.1 in monocytic cells, enabling
by expression of KRAB-C/EBPa-ER (KaER), in which trans-activation via a hybrid DNA-element binding
the C/EBPa DNA-binding domain is linked to both a element (Meraro et al., 1999; Marecki et al., 2001).
KRAB trans-repression domain and the Estrogen However, an essential role for Pip or ICSBP
Receptor ligand-binding domain. In 32D cl3 cells, interaction for PU.1 activity in myeloid cells has not
KaER inhibits granulocytic di€erentiation despite the been established. Notably, ICSBP (7/7) mice have
presence of exogenous G-CSF receptor (Wang and reduced macrophages and increased granulocytic cells,
Friedman, 2002). And in marrow cells, KaER inhibits and introduction of ICSBP into a cell line derived
the formation of CFU-G, CFU-M, and CFU-GM in from the knockout mice increased monocytic markers
multiple cytokines, without a€ecting BFU-E produc- at the expense of granulocytic markers (Holtschke et
tion (Wang and Friedman, 2002). al., 1996; Tamura et al., 2000).
Oncogene
Regulation of granulopoiesis and monopoiesis
AD Friedman
3381
As discussed, c-Jun also cooperates with PU.1 to apparent relaxed `stringency' re¯ects redundancy
regulate several monocytic genes, either via adjacent cis among Ets family members which is more e€ective in
DNA elements or via direct interaction. Notably, c- vitro. For example, GABP can substitute for PU.1 to
Jun, JunB, and JunD levels increase during monocytic activate the NE promoter in 32D cl3 cells (Nuch-
di€erentiation (Lord et al., 1993), and exogenous c-Fos prayoon et al., 1997).
or c-Jun induced partial monocytic di€erentiation in Activation of exogenous PU.1-ER in 32D cl3 cells is
M1, U937, or WEHI-B D+ cells and increased the not sucient to induce terminal granulopoiesis,
responsiveness of U937 cells to phorbol esters (Lord et although increased MPO RNA is detected (Wang et
al., 1993; Szabo et al., 1994; Li et al., 1994). Phorbol al., 1999). Similarly, stable expression of PU.1 in 32D
esters, an agent which stimulates the monocytic cl3 cells is of no consequence in IL-3, but accelerates
di€erentiation of several myeloid cell lines, directly their di€erentiation in G-CSF (Bellon et al., 1998). In
induces the c-Jun promoter via binding sites for c- contrast, over-expression of PU.1 in normal B-
Jun:ATF-2 dimers (van Damm et al., 1993). Phorbol lymphoid progenitors leads to the outgrowth of
esters or other stimuli active N-terminal Jun Kinases macrophages at high levels of PU.1 expression and to
(JNKs), which in turn phosphorylate c-Jun and ATF-2, the generation of B-cells at lower levels of PU.1
increasing their trans-activation potency (Arias et al., (DeKoter and Singh, 2000). The ability of PU.1 to
1994). Phorbol esters also induce the JunB promoter activate its own promoter may play a role in
(de Groot et al., 1991). JunB (7/7) mice in which maintaining high levels of PU.1 expression in myeloid
JunB expression has been rescued in tissues other than cells (Chen et al., 1995b).
marrow retain normal monocyte numbers but develop
granulocytic hyperplasia (Passegue et al., 2001). c-Jun
Core binding factors (CBFs)
(7/7) fetal liver cells reconstitute hematopoiesis in
syngeneic recipients, indicating that, as with JunB, c- The role of CBFs in normal hematopoiesis and in
Jun is not required for myeloid development (Eferl et leukemia was recently reviewed (Friedman, 1999). The
al., 1999). These ®ndings may re¯ect both compensa- CBFs are a family of heterodimeric proteins containing
tory e€ects among Jun family members during a common CBFb subunit and one of three CBFa
monocytic di€erentiation and a speci®c role for JunB subunits, CBFa1, CBFa2 (commonly referred to as
as an inhibitor of proliferation in the granulocyte AML1), and CBFa3 (Bae et al., 1993; Wang et al.,
lineage. c-Jun accelerates cell proliferation in ®bro- 1993; Ogawa et al., 1993a, Levanon et al., 1994). CBFs
blasts and hepatoblasts, suggesting that c-Jun and bind the consensus site, 5'-PuACCPuCA-3' via their N-
JunB are mutually antagonistic in this regard in terminal Runt homology domains (Ogawa et al.,
myeloid cells (Jochum et al., 2001). In fact, JunB 1993a; Meyers et al., 1993). CBFb does not contact
antagonizes the trans-activation potential of c-Jun DNA, but interacts with the CBFa subunits via their
(Chiu et al., 1989; Schutte et al., 1989). Runt domains and increases their anity for DNA
PU.1 also interacts with GATA-1 and inhibits its (Wang et al., 1993; Ogawa et al., 1993b). AML1 can
ability to activate erythroid genes, thereby likely also interact with DNA indirectly, via interaction with
contributing to lineage-speci®c expression in myeloid a subset of Ets family members, including MEF, Ets-1,
cells (Rekhtman et al., 1999; Zhang et al., 2000). and PU.1, and via interaction with C/EBPa (Westen-
Similarly, C/EBPb, and potentially other C/EBPs, dorf et al., 1998; Mao et al., 1999).
reduces FOG but not GATA-1 RNA expression, CBFa2 (AML1) expression is largely restricted to
potentially directing CMPs to the GMP or to an myeloid and lymphoid cells, including CD34+ pre-
eosinophil progenitor (Querfurth et al., 2000). cursors, in adult mice, whereas CBFa1 is most highly
PU.1 is expressed in B lymphoid, granulocytic, and expressed in osteoblasts, and CBFb is widely expressed
monocytic cells (Klemsz et al., 1990; Chen et al., (Satake et al., 1992; Wang et al., 1993; Erickson et al.,
1995a). PU.1 levels increase during granulocytic and 1996; Corsetti and Calabi, 1997; Ducy et al., 1997).
monocytic di€erentiation (Cheng et al., 1996). PU.1 AML1 (7/7) and CBFb (7/7) mice die as embryos
(7/7) mice lack B lymphoid cells and monocytes and without developing de®nitive hematopoiesis, whereas
have greatly reduced neutrophil development (Scott et CBFa1 (7/7) mice have normal blood formation but
al., 1994; McKercher et al., 1996). Introduction of the do not develop calci®ed bones (Okuda et al., 1996;
G-CSF Receptor or M-CSF Receptor into PU.1 (7/ Wang et al., 1996a,b; Sasaki et al., 1996; Niki et al.,
7) marrow cells did not enable generation of 1997; Komori et al., 1997). Adult mice chimeric for
monocytes or neutrophils (DeKoter et al., 1998; CBFb-SMMHC, a dominant-inhibitor of CBFs, devel-
Anderson et al., 1999). Just as C/EBPa (7/7) op erythroid but not myeloid or lymphoid cells
progenitors gain the ability to generate neutrophils expressing CBFb-SMMHC, consistent with a speci®c
when cultured in vitro, PU.1 (7/7) progenitors role for CBFs in the maturation of these lineages in
express myeloperoxidase in IL-3 and G-CSF and adult marrow (Castilla et al., 1999).
express markers of immature monocytes when cultured In addition to activating lineage-speci®c markers
in the combination of IL-3, SCF, GM-CSF, and M- such as MPO and the M-CSF receptor, CBFs stimulate
CSF (DeKoter et al., 1998; Henkel et al., 1999). the G1 to S transition in myeloid and lymphoid cell
Similarly, PU.1 (7/7) ES cells express several early lines (Cao et al., 1997, 1998; Lou et al., 2000; Strom et
myeloid RNAs (Olson et al., 1995). Perhaps this al., 2000). Exposure of 32D cl3 cells to mimosine,
Oncogene
Regulation of granulopoiesis and monopoiesis
AD Friedman
3382
which inhibits G1 progression via induction of p27Kip1, and expression of MafB in myeloblasts directed their
prevented G-CSF-mediated induction of MPO, an di€erentiation to macrophages (Kelly et al., 2000).
early granulocytic marker, but accelerated induction Similarly, expression of c-Maf in HL-60 or U937 cells
of two later markers, LF and Lysozyme (Q Wang and led to monocytic di€erentiation (Hegde et al., 1999). c-
AD Friedman, unpublished). This ®nding suggests that Maf can interact with c-Myb, interfering with its
proliferation is a prerequisite for the activation of activation of early myeloid genes; however, interference
proteins such as CBF and c-Myb expressed in with c-Myb activities was not sucient to induce
immature cells and meant to serve a dual role by monocytic di€erentiation in HL-60 cells (Hegde et al.,
simultaneously stimulating proliferation and di€eren- 1998, 1999).
tiation.
Egr-1 and WT1
c-Myb
Egr-1 is a member of a family of zinc-®nger
c-Myb binds the consensus DNA site 5'-(C/T)AAC(G/ transcription factors which binds the consensus 5'-
T)-3' (Biedenkapp et al., 1988). The 52 amino acid R2 GCGGGGCG-3' (Crosby et al., 1991). The DNA-
and R3 segments located near the N-terminus of c- binding domain of WT1 isoforms lacking a KTS insert
Myb contain tryptophans every 18 ± 19 residues which is strongly homologous, and these WT1 isoforms bind
are part of the hydrophobic core of a helix ± turn ± the same consensus site (Rauscher et al., 1990). Egr-1 is
helix structure (Ogata et al., 1992). c-Myb has a expressed in multiple tissue including the terminal
central trans-activating domain, and its C-terminus stages of macrophage and neutrophil di€erentiation
has an EVES motif which interferes with DNA- (Nguyen et al., 1993; Krishnaraju et al., 1995). Egr-1 is
binding by the N-terminus (Weston and Bishop, necessary for monocytic di€erentiation of U937 or M1
1989; Dash et al., 1999). The C-terminal region of c- cells, prevents the granulocytic di€erentiation of HL-60
Myb also contains a leucine zipper, which has been or 32D cl3 cells, induces the macrophage di€erentiation
shown to interact with a protein termed p160 (Tavner of M1 cells in the absence of IL-6 and endows 32D cl3
et al., 1998). c-Myb is expressed in immature myeloid, cells with the potential to di€erentiation to macro-
lymphoid, and erythroid cells, and c-Myb (7/7) mice phages in response to GM-CSF (Nguyen et al., 1993;
lack each of these lineages (Sheiness and Gardinier, Krishnaraju et al., 1995, 1998). In addition, ectopic
1984; Mucenski et al., 1991). Expression of exogenous expression of Egr-1 in myeloid marrow progenitors
c-Myb in 32D cl3 cells allowed G-CSF induction of increased the proportion of CFU-M at the expense of
MPO but prevented growth arrest and associated CFU-G and, to a lesser extent, BFU-E (Krishnaraju et
induction of late markers such as LF (Bies et al., al., 2001). Mice lacking Egr-1 develop normal numbers
1995). A-Myb and B-Myb are highly homologous to of macrophages, perhaps due to compensatory e€ects
c-Myb within its DNA-binding domain and bind the from other Egr family members (Lee et al., 1996). As
identical consensus sequence (Trauth et al., 1994). A- Egr-1 is induced rapidly by phorbol esters even in the
Myb is expressed in several tissues, including a subset absence of protein synthesis, Egr-1 may be an
of B cells, while B-Myb is expressed widely in important target of M-CSF receptor signaling (Nguyen
proliferating cells and may stimulate cell cycle et al., 1993).
progression (Golay et al., 1991; Reiss et al., 1991; Several WT1 isoforms are expressed as a result of
Trauth et al., 1994). alternative splicing. The transcriptional regulatory
domain of WT1 is a€ected by the presence or absence
of exon 5 and its DNA-binding domain is a€ected by
MafB and c-Maf
the presence or absence of a three amino acid sequence,
MafB and c-Maf are members of a family of basic KTS (Haber et al., 1991). The presence of the KTS
region-leucine zipper DNA-binding proteins. Mafs insert alters the DNA-binding speci®city of WT1
bind and activate transcription weakly as dimers via (Drummond et al., 1994). Lack of WT1 is lethal at
a 13 bp palindrome, 5'-TGCTGACTCAGCA-3', which or before birth, and results in urogenital defects and
contains a central AP-1 site, 5'-TGACTCA-3' (Katao- lack of spleen development (Kreidberg et al., 1993;
ka et al., 1994a,b; Kerppola and Curran, 1994). Mafs Herzer et al., 1999). Marrow development has not been
can also form heterodimers with Fos or Jun family characterized in these mice. Expression of WT1
members which bind modi®ed consensus sites (Katao- isoforms containing the KTS insert induces monocytic
ka et al., 1996). Mafs interact with another bZIP di€erentiation of M1 cells, whereas isoforms lacking
protein, NF-E2, in erythroid cells, enabling induction this insert, and so mimicking Egr-1 DNA-binding
of globin and other erythroid-speci®c genes (Andrews speci®city, prompt a G1 arrest (Smith et al., 1998). In
et al., 1993). MafB is expressed in myeloid cells, contrast, WT1(7KTS) accelerates whereas
including marrow macrophages, but not in erythroid WT1(+KTS) blocks 32D cl3 granulocytic di€erentia-
cells, and it inhibits erythroid gene expression via direct tion in response to G-CSF (Inoue et al., 1998; Loeb et
interaction with Ets-1 (Sieweke et al., 1996). When al., 2000). Inhibition of monocyte development by
avian progenitors were transformed by a Myb-Ets WT1(7KTS) may re¯ect dominant inhibition of Egr-1,
fusion protein in the presence of exogenous MafB, an via competition for DNA-binding. In addition, accel-
increased number of myeloid colonies were obtained, eration of granulocytic di€erentiation by WT1(7KTS)
Oncogene
Regulation of granulopoiesis and monopoiesis
AD Friedman
3383
may re¯ect limitation of G1 progression via direct regulating granulopoiesis in normal cells remains to be
repression of the cyclin E promoter (Loeb et al., 2000). con®rmed.
WT1 is normally expressed in immature, CD34+ HoxA10 is a homeobox protein which binds
marrow cells and is down-regulated in response to preferentially to 5'-TTAT-3' as a heterodimer with
stem cell factor and G-CSF (Maurer et al., 1997). The PBX (Chang et al., 1996). The ability of HoxA10 to
expression of speci®c WT1 isoforms in marrow repress the gp91-phox promoter led to the suggestion
progenitors remains to be determined. Perhaps that HoxA10 plays a role analogous to CDP during
WT1(7KTS) serves to interfere with monocyte granulopoiesis (Eklund et al., 2000). HoxA10 is
development and WT1(+KTS) serves to interfere with highly expressed in CD34+ and is only weakly
granulocytic development. detected in CD347 marrow cells and is not present
in mature neutrophils or monocytes (Sauvageau et
al., 1994; Lawrence et al., 1995). HoxA10 is
Retinoic acid receptors
preferentially expressed in myeloid cell lines, HoxA10
Retinoic acid receptors a, b, g(RARa, RARb, and (7/7) mice have increased numbers of granulocytes,
RARg) bind DNA via their zinc-®nger domains as and over-expression of HoxA10 inhibits monocytic
heterodimers with RXR proteins. RARs are broadly colony formation in vitro and produces AML in vivo
expressed, with RARa being preferentially expressed in (Lawrence et al., 1995; Zhang et al., 1996b;
myeloid cells (de The et al., 1989). The DNA consensus Thorsteinsdottir et al., 1997). Overall, as HoxA10
for RAR-RXR DNA-binding is a direct repeat of 5'- expression is apparently lost prior to CDP activity,
(A/G)G(G/T)TCA-3' separated by 2 ± 5 bps (Umesano it may play a role in maintaining an earlier stage in
et al., 1991; Naar et al., 1991). Dominant-inhibition of myelopoiesis. While HoxA10 can activate the
RARa arrests granulocytic di€erentiation at the p21WAF1/Cip1 promoter in U937 cells, it is tempting
promyelocyte stage (Tsai and Collins, 1993). Mice to speculate that HoxA10 in fact represses this
lacking RARa1 or RARg have normal myeloid promoter in immature myeloid cells, enabling cell
development, whereas while mice lacking both of these proliferation and thereby inhibiting terminal di€er-
RAR isoforms have normal myeloid progenitor entiation (Bromleigh and Freedman, 2000). As we
numbers, their neutrophilic di€erentiation arrests at have discussed, other Hox proteins may play a role
the myelocyte stage (Labrecque et al., 1998). This in regulating myelopoiesis as well (Ward et al.,
phenotype is similar to that observed in C/EBPe (7/ 2000).
7) mice, and RARa directly activates the C/EBPe Like CDP, Sp1 is found in multiple tissues, but Sp1
promoter (Yamanaka et al., 1997; Chih et al., 1997). is expressed at particularly high levels in maturing
While regulation of the C/EBPe promoter by RARs granulocytes (Sa€er et al., 1991). This expression
may account for the requirement of RARs for terminal pattern combined with the ®nding that Sp1 regulates
granulopoiesis, it remains to be established that several granulocytic and monocytic genes via its
regulation of RAR activities plays a role in normal consensus-binding site, 5'-GGGCGG-3', raises the
hematopoiesis. possibility that Sp1 plays a role in regulating terminal
myelopoiesis.
CCAAT displacement protein (CDP) and HoxA10
Myeloid zinc finger 1 (MZF-1)
CDP is a widely expressed protein that represses gene
expression, at least in part, via competition for MZF-1 was isolated as a zinc ®nger protein preferen-
transactivator binding to DNA elements which loosely tially expressed in myeloid cell lines (Hromas et al.,
®t the 5'-CCAAT-3' motif (Barberis et al., 1987; Luo 1991). MZF-1 is preferentially expressed in immature
and Skalnik, 1996). Decreased CDP DNA-binding myeloid cells, and reduction of its expression inhibits
during the terminal stages of granulopoiesis combined the formation of CFU-G (Bavisotto et al., 1991). The
with its ability to repress the gp91-phox promoter led two zinc ®nger clusters within MZF-1 bind the
to the proposal that inactivation and/or decreased consensus sites 5'-AGTGGGGA-3' and 5'-
expression of CDP is required for terminal neutrophil CGGGnGAGGGGGAA-3' (Morris et al., 1994). The
maturation (Skalnik et al., 1991). The mechanism MPO and LF promoters contain bindings sites capable
whereby CDP DNA-binding becomes inactivated of interacting with both sets of zinc ®ngers, and the
during granulopoiesis remains to be elucidated. 32D CD34 promoter contains two such sites located at
cl3 cells overexpressing CDP do not express C/EBPe in about 7500 bp. MZF-1 activates the CD34 promoter
response to G-CSF, and the human C/EBPe promoter via these sites in hematopoietic cell lines (Morris et al.,
contains a binding site for CDP at 71472 bp which 1995). MZF-1 (7/7) mice develop normal blood cells,
potentially accounts for the reduced activity of an but develop a progressive increase in monocytic cells
1800 bp compared to a 726 bp promoter fragment in followed by the late formation of an in®ltrating
32Dwt18 cells (Khanna-Gupta et al., 2001). Inhibition monocytic leukemia. In addition, a much greater
of C/EBPe expression may account for the combined proportion of MZF-1 (7/7) myeloid and erythroid
loss of LF, neutrophil collagenase, and neutrophil progenitors are in cell cycle without increased expres-
gelatinase in 32D cells expressing exogenous CDP sion of c-Myb (Gaboli et al., 2001). This phenotype
(Lawson et al., 1998). However, a role for CDP in may not have been detected during antisense inhibition
Oncogene
Regulation of granulopoiesis and monopoiesis
AD Friedman
3384
of MZF-1 in CFU-G due to residual expression Lineage commitment and progression
(Bavisotto et al., 1991).
The widespread role of C/EBPs and PU.1 in regulating
myeloid genes combined with the defects evident in C/
p53, retinoblastoma protein (Rb), STAT3, and BLIMP-1
EBPa (7/7) and PU.1 (7/7) mice make these
When induced by DNA strand breaks, p53 activates transcription factors leading candidates for roles in
pathways leading to apoptosis or cell cycle arrest determining the myeloid lineages. As PU.1 (7/7) mice
(Canman et al., 1995). p53 is detected at low levels in lack B cells and monocytes and have greatly reduced
mature myeloid and lymphoid cells and p53 levels numbers of neutrophils, whereas C/EBPa (7/7) mice
increase in ML-1 cells as they di€erentiate to only lack maturing granulocytes, it would seem
monocytes in response to phorbol esters (Kastan et reasonable to place PU.1 `upstream' of C/EBPs in this
al., 1991). As c-Jun represses the p53 gene, this e€ect of regard. Further support for this idea comes from the
phorbol esters is likely indirect, perhaps via PKC ®nding that PU.1 and GATA-1 inhibit each others
activation or cell cycle arrest (Schreiber et al., 1999). activities, suggesting their relative importance in the
Exogenous p53 prompts monocytic di€erentiation in myeloid versus the erythrod/megakaryocyte lineages.
U937 cells or in 32D cl3 cells also expressing v-src or However, several lines of evidence suggest that C/EBPs
an activated M-CSF receptor, without inducing G1 might play a role in specifying the bipotent granulo-
arrest or apoptosis (Soddu et al., 1996; Martinelli et cyte-monocyte progenitor (GMP): C/EBP-mediated
al., 1997; Ehinger et al., 1998). The induction of down-regulation of FOG RNA is essential for
monocytic di€erentiation in 32D cl3 cells is striking, eosinophil development, and perhaps is also required
given their propensity for granulocytic maturation. for GMP formation (Querfurth et al., 2000). Second,
Induction of U937 cell di€erentiation by p53 depended fractionation studies suggest that pluripotent stem cells
upon the integrity of its trans-activating domain and ®rst commit to a common lymphoid progenitor (CLP)
was not inhibited by bcl-2 (Chylicki et al., 2000). and a common myeloid progenitor (CMP) and that B-
Although mice lacking p53 develop normally, the p53- cells and macrophages develop from the CLP and the
related protein, p73, potentially compensates for p53 CMP respectively (Akashi et al., 2000); in this model, it
de®ciency as p73 is expressed in immature but not is dicult to draw a 1 : 1 correspondence between PU.1
terminally di€erentiated myeloid cells (Lowe et al., and a speci®c progenitor. Third, the CMP gives rise to
1993; Peters et al., 1999). the GMP, the megakaryocyte-erythroid progenitor
Rb cooperates with E2F family members to repress (MEP), and perhaps an eosinophilic progenitor
genes required for S phase entry (Weinberg, 1995). Rb (EoP), and evidence has been presented indicating that
is down-regulated during granulopoiesis but is up- high-level GATA-1 speci®es the MEP whereas the
regulated during monocyte development (Bergh et al., combination of a C/EBP and lower levels of GATA-1
1999). Consistent with this expression pattern, anti- speci®c to the eosinophil lineage (Kulessa et al., 1995;
sense inhibition of Rb expression in marrow progeni- McDevitt et al., 1997; Yamaguchi et al., 1999).
tors reduced CFU-M in favor of CFU-G (Bergh et al., Therefore, if the third branch eminating from the
1999). Perhaps complexes which form between hypo- CMP, the GMP, were determined by the expression of
phosphorylated Rb and either C/EBPb or PU.1 are C/EBP without GATA-1, then the tripartite decision of
essential for monocyte maturation (Hagemeier et al., the CMP would depend on only two factors ± such
1993; Chen et al., 1996). Rb (7/7) mice are simplicity is attractive. And fourth, higher levels of
embryonic lethal and have increased erythroblasts, PU.1 are present in and are required for the monocyte
whereas lack of the Rb-related protein p107 leads to lineage, compared to the B-lineage (DeKoter and
myeloid hyperplasia (Lee et al., 1992; Lecouter et al., Singh, 2000), and as C/EBPa rapidly induces PU.1
1998). As with p53, Rb family members may RNA expression in 32D cl3 and Ba/F3 cells, it might
compensate for each other in knockout mice to help do so in the GMP as well (Wang et al., 1999). One
mediate myelopoiesis. dicult with this alternative is that C/EBP would be
Dominant inhibition of STAT3 in 32D cl3 cells expected to induce PU.1 in eosinophils, which would
allows expression of early markers but prevents cell inhibit GATA-1 activity. PU.1 is present in mature
cycle arrest and induction of late markers, although C/ eosinophils, and PU.1 RNA is strongly upregulated
EBPe induction was not prevented (Shimozaki et al., when human CD34+ cells di€erentiate to eosinophils
1997; Nakajima and Ihle, 2001). These ®ndings suggest in response to IL-5 (J Du and SJ Ackerman, personal
that STAT3 is required for induction of G1 cell cycle communication). PU.1 7/7 mice have greatly reduced
arrest during granulopoiesis. Consistent with this numbers of mature eosinophils, and direct evidence for
model, dominant inhibition of STAT3 prevents the regulation of an eosinophil-speci®c gene by PU.1
p27Kip1 induction by G-CSF in 32D cl3 cells, and the has been presented (Du et al., 1998; van Dijk et al.,
p27Kip1 promoter is activated directly by STAT3 (De 1998). Perhaps, partial inhibition of GATA-1 by PU.1
Koning et al., 2000). in fact serves to direct CMPs to the EoP, as opposed to
BLIMP-1 is a zinc-®nger DNA-binding protein the MEP. Figure 1 illustrates two alternatives for the
which may contribute to terminal myeloid and B- speci®cation of the GMP, by high levels of PU.1 alone
lymphoid di€erentiation at least in part by repressing or by high levels of PU.1 achieved via expression of C/
transcription of the c-myc gene (Chang et al., 2000). EBPs, likely including C/EBPa as this isoform is
Oncogene
Regulation of granulopoiesis and monopoiesis
AD Friedman
3385

Figure 1 A model for transcriptional regulation of granulocyte and monocyte lineage commitment and progression. Detailed
discussions of lymphoid and erythroid lineage determination are presented elsewhere in this issue. Pluripotent hematopoietic stem
cells (HSC) are directed to the CLP in part by Ikaros, T cell development required GATA-3, and B-cell development requires Pax5
and low levels of PU.1. The speci®cation of the CMP from the HSC remains obscure ± perhaps this is a default pathway in the
absence of Ikaros. Development of the MEP requires high-level GATA-1, whereas the development of the eosinophil lineage
requires lower level (or lower activity) of GATA-1 along with a C/EBP. PU.1 induced by C/EBPs may in fact serve to reduce
GATA-1 activity in this lineage. High-level GATA-1 may further direct the MEP along the erythroid lineage (E), NF-E2, FOG, and
GATA-1 may specify megakaryocyte progenitors (Meg), and MITF in combination with GATA-1 or GATA-2 may allow
basophilic progenitors to form (Baso). Development of the GMP requires high level PU.1 expression, which may develop through
both C/EBP-dependent and C/EBP-independent mechanisms. Redundancy with respect to DNA-binding and trans-activation
potency might allow one or more C/EBP family members to play a role in this regard. Maf and/or Jun family members may then
direct the GMP towards a committed monocyte progenitor, and increased levels of C/EBPs may specify the neutrophil progenitor.
Monocyte lineage progression depends upon the continued presence of PU.1 and perhaps C/EBPs, and may require Egr-1, which
could also serve to repress the granulocytic program. ICSBP may be an important PU.1 co-factor during monocyte maturation.
Terminal maturation may in addition depend upon increased expression of hypo-phosphorylated Rb and/or p53 as a consequence
of cell cycle arrest. Granulocyte maturation requires PU.1, C/EBPs, RARs, C/EBPe, and Sp1 and may also require inactivation of
CDP DNA-binding. RARs, C/EBPa, and/or decreasing CDP activities may in fact serve to induce C/EBPe

expressed most prominently in immature hematopoietic speculation that Maf:Maf, Maf:Jun, Maf:Fos, Jun:Jun,
cells (Scott et al., 1992; Radomska et al., 1998). or Jun:Fos dimers initiate monopoiesis in conjunction
Detailed discussions of erythroid and lymphoid lineage with PU.1. The continued presence of C/EBPs, to
development are presented elsewhere in this issue ± of maintain elevated PU.1 levels, may be required as well.
note however is the ®nding that Pax5 suppresses GM- In the absence of Maf or Jun proteins, C/EBPs and
CSF Receptor RNA expression, again demonstrating PU.1 may specify the granulocyte lineage. The ®nding
the role of negative cross-talk between hematopoietic that increased levels of C/EBPa favor granulocytic over
lineages (Chiang and Monroe, 2001). Both C/EBP- monocytic di€erentiation in U937 cells suggests that, as
dependent and -independent commitment from the with PU.1 and GATA-1, C/EBP levels may play a role
CMP to the GMP might occur, with requisite elevation in lineage commitment decisions (Radomska et al.,
of PU.1 levels. Our ®nding that either C/EBPa-ER or 1998). A model for granulocyte versus monocyte
G-CSF receptor signals, acting in the presence of a lineage determination is illustrated in Figure 1.
dominant-inhibitory C/EBP, elevate PU.1 RNA in 32D Egr-1 may serve to simultaneously enable monocyte
cl3 cells is consistent with this idea (Wang et al., 1999; lineage progression and to prevent cross-over to
Wang and Friedman, 2002). neutrophil development. Granulocyte lineage progres-
The issue of granulocyte versus monocytic lineage sion requires C/EBPe, which may be induced by C/
choice also must be resolved. The ®ndings that Jun and EBPa and/or RARs. Loss of CDP DNA-binding is
Maf family members can each induce the monocyte also required for terminal granulopoiesis. In addition,
lineage in cell lines and that these proteins both progression to neutrophils and macrophages is asso-
interact with AP-1 consensus sites leads to the ciated with cell cycle arrest, elevating hypo-phosphory-
Oncogene
Regulation of granulopoiesis and monopoiesis
AD Friedman
3386
lated Rb and p53. These proteins may participate in co-repressors which participate in lineage speci®cation?
terminal di€erentiation, as best shown for monopoiesis. With respect to key transcription factors, further
And cell cycle arrest may inactivate factors such as clari®cation of the regulatory network illustrated in
CBF and c-Myb which participate in early stages of Figure 1 is needed: Which factor or factors specify
di€erentiation. A model of myeloid lineage progression each lineage and at what levels of expression; can
is shown in Figure 1. family members act redundantly in this regard; what
additional cooperative mechanisms operate among
transcriptional regulators; what roles do cytokine
Perspectives for the future receptor signaling and transcription factor modi®ca-
tions play in each commitment decision and in each
Many questions remain unanswered regarding the step of lineage progression? The answers to these
transcriptional regulation of granulocyte and monocyte questions will provide general lessons in developmental
development. With respect to the cellular basis for biology and insights into leukemogenesis and will
initiating these lineages: What are the relative con- enable applications in clinical hematology, oncology,
tributions of granulocyte/monocyte and B-cell/mono- and gene therapy.
cyte progenitors to mature blood elements; do some
granulocyte or monocyte progenitors develop directly
from pluripotent stem cells; how irreversible are
commitment decisions? With respect to gene regula-
tion: Are there additional important transcriptional Acknowledgments
regulators of myeloid genes remaining to be uncovered AD Friedman is a Scholar of the Leukemia and Lympho-
via detailed investigation of promoters and distal ma Society and his research is supported by the Children's
enhancers; are there lineage-restricted co-activators or Cancer Foundation.

References

Ahne B and Stratling WH. (1994). J. Biol. Chem., 269, Britos-Bray M and Friedman AD. (1997). Mol. Cell. Biol.,
17794 ± 17801. 17, 5127 ± 5135.
Akashi K, Traver D, Miyamoto T and Weissman IL. (2000). Bromleigh VC and Freedman LP. (2000). Genes Dev., 14,
Nature, 404, 193 ± 197. 2581 ± 2586.
Anderson KL, Smith KA, Perkin H, Hermanson G, Calkhoven CF, Muller C and Leutz A. (2000). Genes Dev.,
Anderson CG, Jolly DJ, Maki RA and Torbett BE. 14, 1920 ± 1932.
(1999). Blood, 94, 2310 ± 2318. Canman CE, Gilmer TM, Coutts SB and Kastan MB. (1995).
Andrews NC, Kotkow KJ, Ney PA, Eerdjument BH, Tempst Genes Dev., 9, 600 ± 611.
P and Orkin SH. (1993). Proc. Natl. Acad. Sci. USA, 90, Cao W, Britos-Bray M, Claxton DF, Kelley CA, Speck NA,
11488 ± 11492. Liu PP and Friedman AD. (1997). Oncogene, 15, 1315 ±
Antonson P, Stellan B, Yamanaka R and Xanthopoulos KG. 1327.
(1996). Genomics, 35, 30 ± 38. Cao W, Adya N, Britos-Bray M, Liu PP and Friedman AD.
Arias J, Alberts AS, Brindle P, Claret FX, Smeal T, Karin M, (1998). J. Biol. Chem., 273, 31534 ± 31540.
Feramisco J and Montminy M. (1994). Nature, 370, 226 ± Carlsson L, Candeias S, Staerz U and Keller G. (1995). Eur.
229. J. Immunol., 25, 2308 ± 2317.
Bae SC, Yamaguchi-Iwai Y, Ogawa E, Maruyama M, Castilla LH, Garrett L, Adya N, Orlic D, Dutra A, Anderson
Inuzuka M, Kagoshima H, Shigesada K, Satake M and S, Owens J, Eckhaus M, Bodine D and Liu PP. (1999).
Ito Y. (1993). Oncogene, 8, 809 ± 814. Nature Genet., 23, 144 ± 146.
Barberis A, Super-Furga G and Busslinger M. (1987). Cell, Catt D, Hawkins S, Roman A, Luo W and Skalnik DG.
50, 347 ± 359. (1999). Blood, 94, 3151 ± 3160.
Bassuk AG and Leiden JM. (1995). Immunity, 3, 223 ± 237. Chang CP, Brocchieri L, Shen WF, Largman C and Cleary
Bavisotto L, Kaushansky K, Lin N and Hromas R. (1991). J. ML. (1996). Mol. Cell. Biol., 16, 1734 ± 1745.
Exp. Med., 174, 1097 ± 1101. Chang DH, Angelin-Duclos C and Calame K. (2000). Nat.
Behre G, Whitmarsh AJ, Coghlan MP, Hoang T, Carpenter Immunol., 1, 169 ± 176.
CL, Zhang DE, Davis RJ and Tenen DG. (1999a). J. Biol. Chen HM, Pahl HL, Scheibe RJ, Zhang DE and Tenen DG.
Chem., 274, 4939 ± 4946. (1993). J. Biol. Chem., 268, 8230 ± 8239.
Behre G, Smith LT, Carpenter CL, Hiddemann W, Fried- Chen HM, Zhang P, Voso MT, Hohaus S, Gonzalez DA,
man AD and Tenen DG. (1999b). Blood, 94, 684a. Glass CK, Zhang DE and Tenen DG. (1995a). Blood, 85,
Bellon T, Perrotti D and Calabretta D. (1998). Blood, 90, 2918 ± 2928.
1828 ± 1839. Chen H, Ray-Gallet D, Zhang P, Hetherington CJ, Gonzalez
Bergh G, Ehinger M, Olsson I, Jacobsen SEW and Gullberg DA, Zheng DE, Moreau-Gachelin F and Tenen DG.
U. (1999). Blood, 94, 1971 ± 1978. (1995b). Oncogene, 11, 1549 ± 1560.
Biedenkapp H, Borgemeyer U, Sippel AE and Klempnauer Chen PL, Riley DJ, Chen-Kiang S and Lee WH. (1996).
KH. (1988). Nature, 335, 835 ± 837. Proc. Natl. Acad. Sci. USA, 93, 465 ± 469.
Bies J, Mukhopadhyaya R, Pierce J and Wol€ L. (1995). Cell Chen SS, Chen JF, Johnson PF, Muppala V and Lee YH.
Growth Di€er., 6, 59 ± 68. (2000). Mol. Cell. Biol., 20, 7292 ± 7299.
Bourette RP and Rohrschneider LR. (2000). Growth Factors, Cheng T, Shen H, Giokas D, Gere J, Tenen DG and Scadden
17, 155 ± 166. DT. (1996). Proc. Natl. Acad. Sci. USA, 93, 13158 ± 13163.

Oncogene
Regulation of granulopoiesis and monopoiesis
AD Friedman
3387
Chiang MY and Monroe JG. (2001). J. Immunol., 166, Friedman AD. (1996b). Cancer Res., 56, 3250 ± 3256.
6091 ± 6098. Friedman AD. (1999). Leukemia, 13, 1932 ± 1942.
Chih DY, Chumakov AM, Park DJ, Silla AG and Koe‚er Gaboli M, Kotsi PA, Gurrieri C, Cattoretti G, Ronchetti S,
HP. (1997). Blood, 90, 2987 ± 2994. Cordon-Cardo C, Broxmeyer HE, Hromas R and Pandol®
Chiu R, Angel P and Karin M. (1989). Cell, 59, 979 ± 986. PP. (2001). Genes Dev., 15, 1625 ± 1630.
Christensen JE, Andreasen SO and Christensen JP. (2001). Goethe R and Loc PV. (1994). J. Biol. Chem., 269, 31302 ±
Int. Immunol., 13, 593 ± 600. 31309.
Christy RJ, Kaestner KH, Geiman DE and Lane MD. Golay J, Capucci A, Arsura M, Castellano M, Rizzo V and
(1993). Proc. Natl. Acad. Sci. USA, 88, 2593 ± 2597. Introna M. (1991). Blood, 77, 149 ± 158.
Chumakov AM, Grillier I, Chumakova E, Chih D, Slater J Gombart AF, Shiohara M, Kwok SH, Agematsu K,
and Koe‚er HP. (1997). Mol. Cell. Biol., 17, 1375 ± 1386. Komiyama A and Koe‚er HP. (2001). Blood, 97, 2561 ±
Chylicki K, Ehinger M, Svedberg H and Gullberg U. (2000). 2567.
Cell Growth Di€er., 11, 561 ± 571. Habener JF and Ron D. (1992). Genes Dev., 6, 439 ± 453.
Collins SJ, Ulmer J, Purton LE and Darlington G. (2001). Haber DA, Sohn RL, Buckler AJ, Pelletier J, Call KM and
Blood, 98, 2382 ± 2388. Housman DE. (1991). Proc. Natl. Acad. Sci. USA, 88,
Cooper C, Henderson A, Artandi S, Avitahl N and Calame 9618 ± 9622.
K. (1995). Nucl. Acids Res., 23, 4371 ± 4377. Hagemeier C, Bannister AJ, Cook A and Kouzarides T.
Corsetti MT and Calabi F. (1997). Blood, 89, 2359 ± 2368. (1993). Proc. Natl. Acad. Sci. USA, 90, 1580 ± 1584.
Crosby SD, Puetz JJ, Simburger KS, Fahrner TJ and Hegde SP, Kumar A, Kurschner C and Shapiro LH. (1998).
Milbrandt J. (1991). Mol. Cell. Biol., 11, 3835 ± 3841. Mol. Cell. Biol., 18, 2729 ± 2737.
Dash AB, Orrico FC and Ness SA. (1999). Genes Dev., 10, Hegde SP, Zhao J, Ashmun RA and Shapiro LH. (1999).
1858 ± 1869. Blood, 94, 1578 ± 1589.
de Groot R, Auwerx J, Karperien M, Staels B and Kruijer W. Henkel GW, McKercher SR, Leenen PJ and Maki RA.
(1991). Nucl. Acids Res., 19, 775 ± 781. (1999). Blood, 93, 2849 ± 2858.
De Koning JP, Soede-Bobok AA, Ward AC, Schelen AM, Herzer U, Crocoll A, Barton D, Howells N and Englert C.
Antonissen C, van Leeuwen D, Lowenberg B and Touw (1999). Curr. Biol., 9, 837 ± 840.
IP. (2000). Oncogene, 19, 3290 ± 3298. Hetherington CJ, Kingsley PD, Crocicchio F, Zhang P,
DeKoter RP and Singh H. (2000). Science, 288, 1439 ± 1441. Rabin MS, Palis J and Zhang DE. (1999). J. Immunol.,
DeKoter RP, Walsh JC and Singh H. (1998). EMBO J., 17, 162, 503 ± 509.
4456 ± 4468. Heydemann A, Juan G, Hennessy K, Parmacek MS and
Descombes P and Schibler U. (1991). Cell, 67, 569 ± 580. Simon MC. (1996). Mol. Cell. Biol., 16, 1676 ± 1686.
de The H, Marchio A, Tiollais P and Dejean A. (1989). Hohaus S, Petrovick MS, Voso MT, Sun Z, Zhang DE and
EMBO J., 8, 429 ± 433. Tenen DG. (1995). Mol. Cell. Biol., 15, 5830 ± 5845.
Drummond IA, Rupprecht HD, Rohwer-Nutter P, Lopez- Holtschke T, Lohler J, Kanno Y, Fehr T, Giese N,
Guisa J, Maddel SL, Rauscher FJ and Sukhatme VP. Rosenbauer F, Lou J, Knobeloch KP, Gabriele L, Waring
(1994). Mol. Cell. Biol., 14, 3800 ± 3809. JF, Bachmann MF, Zinkernagel RM, Morse III HC,
Du J, Savage MP, DeKoter R, McKercher SR, Maki R, Ozato K and Horak I. (1996). Cell, 87, 307 ± 317.
Singh H and Ackerman SJ. (1998). Blood, 92, 191a. Hromas R, Collins SJ, Hickstein D, Raskind W, Deaven LL,
Ducy P, Zhang R, Geo€roy V, Ridall AL and Karsenty G. O'hara P, Hagen FS and Kaushanksky K. (1991). J. Biol.
(1997). Cell, 89, 747 ± 754. Chem., 266, 14183 ± 14187.
Ehinger M, Bergh G, Johnsson E, Baldetorp B, Olsson I and Hu HM, Baer M, Williams SC, Johnson PF and Schwartz
Gullberg U. (1998). Exp. Hematol., 26, 1043 ± 1052. RC. (1998). J. Immunol., 160, 2334 ± 2342.
Eisenbeis CF, Singh H and Storb U. (1995). Genes Dev., 9, Inoue K, Tamaki H, Ogawa H, Oka Y, Soma T, Tatekawa T,
1377 ± 1387. Oji Y, Tsuboi A, Kim EH, Kawakami M, Akiyama T,
Eklund EA and Kakar R. (1999). J. Immunol., 163, 6095 ± Kishimoto T and Sugiyama H. (1998). Blood, 91, 2969 ±
6105. 2976.
Eklund EA, Jalava A and Kakar R. (2000). J. Biol. Chem., Jochum W, Passegue E and Wagner F. (2001). Oncogene, 20,
275, 20117 ± 20126. 2401 ± 2412.
Eferl R, Sibilia M, Hilberg F, Fuchsbichler A, Ku€erath I, Johansen LM, Iwama A, Lodie TA, Sasaki K, Felsher DW,
Guertl B, Zenz R, Wagner EF and Zatloukal K. (1999). J. Golub TR and Tenen DG. (2001). Mol. Cell. Biol., 21,
Cell. Biol., 145, 1049 ± 1061. 3789 ± 3806.
Erickson P, Dessev G, Lasher RS, Philips G, Robinson M Jones LC, Lin ML, Chen SS, Krug U, Hofmann WK, Lee S,
and Drabkin H. (1996). Blood, 88, 1813 ± 1823. Lee YH and Koe‚er HP. (2002). Blood, 99, 2032 ± 2036.
Faust N, Bonifer C and Sippel AE. (1999). DNA Cell Biol., Kastan MB, Radin AI, Kuerbitz SJ, Onyekwere O, Wolkow
18, 631 ± 642. CA, Civin CI, Stone KD, Woo T, Ravindranath Y and
Fisher RC, Olson MC, Pongubala JM, Perkel JM, Atchinson Craig RW. (1991). Cancer Res., 51, 4279 ± 4286.
ML, Scott EW and Simon MC. (1998). Mol. Cell. Biol., 18, Kataoka K, Fujiwara KT, Noda M and Nishizawa M.
4347 ± 4357. (1994a). Mol. Cell. Biol., 14, 7581 ± 7591.
Ford AM, Bennett CA, Healy LE, Towatari M, Greaves MF Kataoka K, Noda M and Nishizawa M. (1994b). Mol. Cell.
and Enver T. (1996). Proc. Natl. Acad. Sci. USA, 93, Biol., 14, 700 ± 712.
10838 ± 10843. Kataoka K, Noda M and Nishizawa M. (1996). Oncogene,
Friedman AD, Landschulz WH and McKnight SL. (1989). 12, 53 ± 62.
Genes Dev., 3, 1314 ± 1322. Kelly LM, Englmeier U, Lafon I, Sieweke MH and Graf T.
Friedman AD and McKnight SL. (1990). Genes Dev., 4, (2000). EMBO J., 19, 1987 ± 1997.
1416 ± 1426. Kerppola TK and Curran TK. (1994). Oncogene, 9, 675 ±
Friedman AD. (1996a). Curr. Topics Microb. Immun., 211, 684.
149 ± 158.

Oncogene
Regulation of granulopoiesis and monopoiesis
AD Friedman
3388
Khanna-Gupta A, Zibello T, Kolla S, Neufeld EJ and Lou J, Cao W, Bernardin F, Ayyanathan K, Rauscher III FJ
Berliner N. (1997). Blood, 90, 2784 ± 2795. and Friedman AD. (2000). Oncogene, 19, 2695 ± 2703.
Khanna-Gupta A, Zibello T, Simkevich C, Rosmarin AG Lowe SW, Schmitt EM, Smith SW, Osborne BA and Jacks T.
and Berliner N. (2000). Blood, 95, 3734 ± 3741. (1993). Nature, 362, 847 ± 849.
Khanna-Gupta A, Zibello T, Sun H, Lekstrom-Himes J and Luo W and Skalnik DG. (1996). J. Biol. Chem., 271, 18203 ±
Berliner N. (2001). Proc. Natl. Acad. Sci., USA, 98, 8000 ± 18210.
8005. Lutz PG, Houzel-Charavel A, Moog-Lutz C and Cayre YE.
Klemsz MJ, McKercher SR, Celada A, Van Beveran C and (2001). Blood, 97, 2449 ± 2456.
Maki RA. (1990). Cell, 61, 113 ± 124. Manz MG, Traver D, Miyamoto T, Weissman IL and
Komori T, Yagi H, Nomura S, Yamaguchi A, Sasaki K, Akashi K. (2001). Blood, 97, 3333 ± 3341.
Deguchi K, Shimizu Y, Bronson RT, Gao YH, Inada M, Mao S, Frank RC, Zhang J, Miyazaki Y and Nimer SD.
Sato M, Okamoto R, Kitamura Y, Yoshiki S and (1999). Mol. Cell. Biol., 19, 3635 ± 3644.
Kishimoto T. (1997). Cell, 89, 755 ± 764. Marecki S, Riendeau CJ, Liang MD and Fenton MJ. (2001).
Kondo M, Scherer DC, Miyamoto T, King AG, Akashi K, J. Immunol., 166, 6829 ± 6838.
Sugamura K and Weissman IL. (2000). Nature, 407, 383 ± Martinelli R, Blandino G, Scardigli R, Crescenzi M,
386. Lombardi D, Sacchi A and Soddu S. (1997). Oncogene,
Kreidberg JA, Sariola H, Loring JM, Maeda M, Pelletier J, 15, 607 ± 611.
Housman D and Jaenisch R. (1993). Cell, 74, 679 ± 691. Maurer U, Brieger J, Weidmann E, Mitrou PS, Hoelzer D
Krishnaraju K, Nguyen HQ, Liebermann DA and Ho€man and Bergmann L. (1997). Exp. Hematol., 25, 945 ± 950.
B. (1995). Mol. Cell. Biol., 15, 5499 ± 5507. McDevitt MA, Shivdasani RA, Fujiwara Y, Yang H and
Krishnaraju K, Ho€man B and Liebermann DA. (1998). Orkin SH. (1997). Proc. Natl. Acad. Sci. USA, 94, 6781 ±
Blood, 92, 1957 ± 1966. 6785.
Krishnaraju K, Ho€man B and Liebermann DA. (2001). McKercher SR, Torbett BE, Anderson KL, Henkel GW,
Blood, 97, 1298 ± 1305. Vestal DJ, Baribault H, Klemsz M, Feeney AJ, Wu GE,
Kulessa H, Frampton J and Graf T. (1995). Genes Dev., 9, Paige CJ and Maki RA. (1996). EMBO J., 15, 5647 ± 5658.
1250 ± 1262. Meraro D, Hashmueli S, Koren B, Azriel A, Oumard A,
Labrecque J, Allan D, Chambon P, Iscove NN, Lohnes D Kircho€ S, Hauser N, Nagulapalli S, Atchinson ML and
and Hoang T. (1998). Blood, 92, 607 ± 615. Levi BZ. (1999). J. Immunol., 163, 6468 ± 6478.
Lacaud G, Carlsson L and Keller G. (1998). Immunity, 9, Meyers S, Downing JR and Hiebert SW. (1993). Mol. Cell.
827 ± 838. Biol., 13, 6336 ± 6345.
Lagasse E and Weissman IL. (1997). Cell, 89, 1021 ± 1031. Mischak H, Pierce JH, Goodnight J, Kazanietz MG,
Landschulz WH, Johnson PF and McKnight SL. (1989). Blumberg PM and Mushinski JF. (1993). J. Biol. Chem.,
Science, 246, 1681 ± 1688. 268, 20110 ± 20115.
Lawrence HJ, Sauvageau G, Ahmadi N, Lopez AR, LeBeau Montecino-Rodriguez E, Leathers H and Dorshkind K.
MM, Link M, Humphries K and Largman C. (1995). Exp. (2001). Nature Immunol., 2, 83 ± 88.
Hematol., 23, 1160 ± 1166. Morris JF, Hromas R and Rauscher FJ. (1994). Mol. Cell.
Lawson ND, Khanna-Gupta A and Berliner N. (1998). Biol., 14, 1786 ± 1795.
Blood, 91, 2517 ± 2524. Morris JF, Rauscher JF, Davis P, Klemsz M, Xu D, Tenen D
LeCouter JE, Kablar B, Hardy WR, Ying C, Megeney LA, and Hromas R. (1995). Blood, 86, 3640 ± 3647.
May LL and Rudnicki MA. (1998). Mol. Cell. Biol., 18, Moulton KS, Semple K, Wu H and Glass CK. (1994). Mol.
7455 ± 7465. Cell. Biol., 14, 4408 ± 4418.
Lee EY, Chang CY, Wang YC, Lai CC, Herrup K, Lee WH Mucenski ML, McClain K, Kier AB, Swerdlow SH,
and Bradley A. (1992). Nature, 359, 288 ± 294. Schreiner CM, Miller TA, Pietryga DW, Scott WJ and
Lee SL, Wang Y and Milbrandt J. (1996). Mol. Cell. Biol., Potter SS. (1991). Cell, 65, 677 ± 689.
16, 4566 ± 4572. Muller C, Kowenz-Leutz E, Grieser-Ada S, Graf T and
Lekstrom-Himes JA, Dorman SE, Dopar P, Holland SM and Leutz A. (1995). EMBO J., 14, 6127 ± 6135.
Gallin JI. (1999). J. Exp. Med., 189, 1847 ± 1852. Naar AM, Boutin JM, Lipkin SM, Yu VC, Holloway JM,
Levanon D, Negreanu V, Bernstein Y, Bar-Am I, Avivi L Glass CK and Rosenfeld MG. (1991). Cell, 65, 1267 ±
and Groner Y. (1994). Genomics, 23, 425 ± 432. 1279.
Li AC, Guidez FRB, Collier JG and Glass CK. (1998). J. Nakajima H and Ihle N. (2001). Blood, 98, 897 ± 905.
Biol. Chem., 273, 5389 ± 5399. Nerlov C, McNagny KM, Doderlein G, Kowentz-Leutz E
Li J, King I and Sartorelli AC. (1994). Cell Growth Di€er., 5, and Graf T. (1998). Genes Dev., 12, 2413 ± 2422.
743 ± 751. Ness SA, Kowentz-Leutz E, Casini T, Graf T and Leutz A.
Lieschke GJ, Grail D, Hodgson G, Metcalf D, Stanley E, (1993). Genes Dev., 7, 749 ± 759.
Cheers C, Fowler KJ, Basu S, Zhan YF and Dunn AR. Nguyen HQ, Ho€man-Liebermann B and Liebermann DA.
(1994). Blood, 84, 1737 ± 1746. (1993). Cell, 72, 197 ± 209.
Liu F, Wu HY, Wesselschmidt R, Kornaga T and Link DC. Niki M, Okada H, Takano H, Kujo J, Tani K, Hibino H,
(1996a). Cell, 5, 491 ± 501. Asano S, Ito Y, Satake M and Noda T. (1997). Proc. Natl.
Liu M, Lee MH, Cohen M, Bommakanti M and Freedman Acad. Sci. USA., 94, 5697 ± 5702.
LP. (1996b). Genes Dev., 10, 142 ± 153. Nuchprayoon I, Meyers S, Scott LM, Suzow J, Hiebert S and
Liu F, Poursine-Laurent J, Wu HY and Link DC. (1997). Friedman AD. (1994). Mol. Cell. Biol., 14, 5558 ± 5568.
Blood, 90, 2583 ± 2590. Nuchprayoon I, Simkevich CP, Menglin L, Friedman AD
Loeb DM, Friedman AD and Sukumar SV. (2000). Blood, and Rosmarin AD. (1997). Blood, 89, 4546 ± 4554.
96, 284a. Nuchprayoon I, Simkevich CP, Luo M, Rosmarin AG and
Lord KA, Abdollahi A, Ho€man-Liebermann B and Friedman AD. (1999). J. Biol. Chem., 274, 1085 ± 1091.
Liebermann DA. (1993). Mol. Cell. Biol., 13, 841 ± 851.

Oncogene
Regulation of granulopoiesis and monopoiesis
AD Friedman
3389
Oelgeschlager M, Nuchprayoon I, Luscher B and Friedman Scott LM, Civin CI, Rorth P and Friedman AD. (1992).
AD. (1996). Mol. Cell. Biol., 16, 4717 ± 4725. Blood, 80, 1725 ± 1735.
Ogata K, Hojo H, Aimoto S, Nakai T, Nakamura H, Sarai Scott EW, Simon MC, Anastasi J and Singh H. (1994).
A, Ishii S and Nishimura Y. (1992). Proc. Natl. Acad. Sci. Science, 265, 1573 ± 1577.
USA, 89, 6428 ± 6432. Screpanti I, Romani L, Musiani P, Modesti A, Fattori E,
Ogawa E, Maruyama M, Kagoshima H, Inuzuka M, Lu J, Lazzaro D, Sellito C, Scarpa S, Bellavia D, Lattanzio G,
Satake M, Shigesada K and Ito Y. (1993a). Proc. Natl. Bistoni F, Frati L, Cortese R, Gulino A, Ciliberto G,
Acad. Sci. USA, 90, 6859 ± 6863. Coslantini F and Poli V. (1995). EMBO J., 14, 1932 ± 1941.
Ogawa E, Inuzuka M, Maruyamna M, Satake M, Naito- Seymour JF, Lieschke GJ, Grail D, Quilici C, Hodgson G
Fujimoto M, Ito Y and Shigesada K. (1993b). Virol., 194, and Dunn AR. (1997). Blood, 90, 3037 ± 3049.
314 ± 331. Shapiro LH. (1995). J. Biol. Chem., 270, 8763 ± 8771.
Okuda T, van Deursen J, Hiebert SW, Grosveld G and Sheiness D and Gardinier M. (1984). Mol. Cell. Biol., 4,
Downing JR. (1996). Cell, 84, 321 ± 330. 1206 ± 1212.
Olson MC, Scott EW, Hack AA, Su GH, Tenen DG, Singh H Shimozaki K, Nakajima K, Hirano T and Nagata S. (1997).
and Simon MC. (1995). Immunity, 3, 703 ± 714. J. Biol. Chem., 272, 25184 ± 25189.
Pahl HL, Scheibe RJ, Zhang DE, Chen HM, Galson DL, Sieweke MH, Tekotte H, Frampton J and Graf T. (1996).
Maki RA and Tenen DG. (1993). J. Biol. Chem., 268, Cell, 85, 49 ± 60.
5014 ± 5020. Skalnik DG, Strauss EC and Orkin SH. (1991). J. Biol.
Pan Z, Hetherington CJ and Zhang DE. (1999). J. Biol. Chem., 266, 16376 ± 16744.
Chem., 274, 23242 ± 23248. Slomiany BA, D'Arigo KL, Kelly MM and Kurtz DT.
Park DJ, Chumakov A, Vuong P, Chih D, Gombart A, (2000). Mol. Cell. Biol., 20, 5986 ± 5997.
Miller Jr WH and Koe‚er HP. (1999). J. Clin. Invest., 103, Smith LT, Hohaus S, Gonzalez DA, Dziennis SE and Tenen
1399 ± 1408. DG. (1996). Blood, 88, 1234 ± 1247.
Peters UR, Tschan MP, Kreuzer KA, Baskaynak G, Lass U, Smith SI, Weil D, Johnson GR, Boyd AW and Li CL. (1998).
Tobler A, Fey MF and Schmidt CA. (1999). Cancer Res., Blood, 91, 764 ± 773.
59, 4233 ± 4236. Soddu S, Blandino G, Scardigli R, Martinelli R, Rizzo MG,
Petrovick MS, Hiebert SW, Friedman AD, Hetherington CJ, Crescenzi M and Sacchi A. (1996). Mol. Cell. Biol., 16,
Tenen DG and Zhang DE. (1998). Mol. Cell. Biol., 18, 487 ± 495.
3915 ± 3925. Strom DK, Nip J, Westendorf JJ, Linggi B, Lutterbach B,
Passegue E, Jochum W, Schorpp-Kistner M, Mohle- Downing JR, Lenny N and Hiebert SW. (2000). J. Biol.
Steinlein U and Wagner EF. (2001). Cell, 104, 21 ± 32. Chem., 275, 3438 ± 3445.
Querfurth E, Schuster M, Kulessa H, Crispino JD, Suzow J and Friedman AD. (1993). Mol. Cell. Biol., 13,
Doderlien G, Orkin SH, Graf T and Nerlov C. (2000). 2141 ± 2151.
Genes Dev., 14, 2515 ± 2525. Szabo E, Preis LH and Birrer MJ. (1994). Cell Growth
Radomska HS, Huettner CS, Zhang P and Tenen DG. Di€er., 5, 439 ± 446.
(1998). Mol. Cell. Biol., 18, 4301 ± 4314. Tamura T, Nagamura-Inoue T, Shmeltzer Z, Kuwata T and
Rauscher FJ, Morris JF, Tournary OE, Cook DM and Ozato K. (2000). Immunity, 13, 155 ± 165.
Curran T. (1990). Science, 250, 1259 ± 1262. Tanaka T, Akira S, Yoshida M, Umemoto Y, Yoneda N,
Reiss K, Travali S, Calabretta B and Baserga R. (1991). J. Shirafuji H, Suematsu S, Yoshida N and Kishimoto T.
Cell Physiol., 148, 338 ± 343. (1995). Cell, 80, 353 ± 361.
Rekhtman N, Radparvar F, Evans T and Skoultchi AI. Tanaka T, Yoshida N, Kishimoto T and Akira S. (1997).
(1999). Genes Dev., 13, 1398 ± 1411. EMBO J., 16, 7432 ± 7443.
Rodel JE and Link DC. (1996). Blood, 87, 1309 ± 1312. Tavner FJ, Simpson R, Tashiro S, Favier D, Jenkins NA,
Roodman GD. (1996). Endocr. Rev., 17, 308 ± 332. Gilbert DJ, Copeland NG, Macmillan EM, Lutwyche J,
Rosmarin AG, Caprio D, Levy R and Simkevich C. (1995a). Keough RA, Ishii S and Gonda TJ. (1998). Mol. Cell.
Proc. Natl. Acad. Sci. USA, 92, 801 ± 805. Biol., 18, 989 ± 1002.
Rosmarin AG, Caprio DG, Kirsch DG, Handa H and Thorsteinsdottir U, Sauvageau G, Hough MR, Dragowska
Simkevich CP. (1995b). J. Biol. Chem., 270, 23627 ± 23633. W, Lansdorp PM, Lawrence HJ, Largman C and
Rosmarin AG, Luo M, Caprio DG, Shang J and Simkevich Humphries RK. (1997). Mol. Cell. Biol., 17, 495 ± 505.
CP. (1998). J. Biol. Chem., 273, 13097 ± 13103. Timchenko NA, Wilde M and Darlington GJ. (1999). Mol.
Sa€er JD, Jackson SP and Annarella MB. (1991). Mol. Cell. Cell. Biol., 19, 2936 ± 2945.
Biol., 11, 2189 ± 2199. Trauth K, Mutschler B, Jenkins NA, Gilbert DJ, Copeland
Sasaki K, Yagi H, Bronson RT, Tominaga K, Matsunashi T, NG and Klempnauer KH. (1994). EMBO J., 13, 5994 ±
Deguchi K, Tani Y, Kishimoto T and Komori T. (1996). 6005.
Proc. Natl. Acad. Sci. USA, 93, 12359 ± 12363. Traver D, Miyamoto T, Christensen J, Iwasaki-Arai J,
Satake M, Inuzuka M, Shigesada K, Oikawa T and Ito Y. Askashi K and Weissman IL. (2001). Blood, 98, 627 ± 635.
(1992). Jpn. J. Can. Res., 83, 714 ± 722. Tsai S and Collins SJ. (1993). Proc. Natl. Acad. Sci. USA, 90,
Sauvageau G, Lansdorp PM, Eaves CJ, Hogge DE, 7153 ± 7157.
Dragowsak WH, Reid DS, Largman C, Lawrence JH Tsai FY, Browne CP and Orkin SH. (1998). Dev. Biol., 196,
and Humphries RK. (1994). Proc. Natl. Acad. Sci. USA, 218 ± 227.
91, 12223 ± 12227. Umek RH, Friedman AD and McKnight SL. (1991).
Schreiber M, Kolbus A, Piu F, Szabowski A, Mohle- Science, 25, 288 ± 292.
Steinlein U, Tian J, Karin M, Angel P and Wagner E. Umesano K, Murakami KK, Thompson CC and Evans RM.
(1999). Genes Dev., 13, 607 ± 619. (1991). Cell, 65, 1255 ± 1266.
Schutte J, Viallet J, Nau M, Segal S, Fedorko J and Minna J.
(1989). Cell, 59, 987 ± 997.

Oncogene
Regulation of granulopoiesis and monopoiesis
AD Friedman
3390
van Damm HH, Duyndam M, Rottier R, Bosch A, de Vries- Yamaguchi Y, Nishio H, Kishi K, Ackerman SJ and Suda T.
Smits L, Herrlich P, Zantema A, Angel P and van der Eb. (1999). Blood, 94, 1429 ± 1439.
(1993). EMBO J., 12, 479 ± 487. Yamanaka R, Barlow C, Lekstrom-Himes J, Castilla LH,
van Dijk TB, Caldenhoven E, Raaijmakers JA, Lammer JW, Liu PP, Eckhaus M, Decker T, Wynshaw-Boris A and
Koendeerman L and De Groot RP. (1998). Blood, 91, Xanthopolous KG. (1997). Proc. Natl. Acad. Sci. USA,
2126 ± 2132. 94, 13187 ± 13192.
Wang S, Wang Q, Crute BE, Melnikova IN, Keller SR and Zhang DE, Hetherington CJ, Chen HM and Tenen DG.
Speck NA. (1993). Mol. Cell. Biol., 13, 3324 ± 3339. (1994a). Mol. Cell. Biol., 14, 373 ± 381.
Wang Q, Stacy T, Binder M, Marin-Padilla M, Sharpe AH Zhang DE, Fujioka KI, Hetherington CJ, Shapiro LH, Chen
and Speck NA. (1996a). Proc. Natl. Acad. Sci. USA, 93, HM, Look AT and Tenen DG. (1994b). Mol. Cell. Biol.,
3444 ± 3449. 14, 8085 ± 8095.
Wang Q, Stacy T, Miller JD, Lewis AF, Gu T-L, Huang X, Zhang DE, Hetherington CJ, Tan S, Dziennis SE, Gonzalez
Bushweller JH, Bories J-C, Alt FW, Ryan G, Liu PP, DA, Chen HM and Tenen DG. (1994c). J. Biol. Chem.,
Wynshaw-Boris A, Binder M, Marin-Padilla M, Sharpe 269, 11425 ± 11434.
AH and Speck NA. (1996b). Cell, 87, 697 ± 708. Zhang DE, Hetherington CJ, Meyer S, Rhoades KL, Larson
Wang X, Scott E, Sawyers CL and Friedman AD. (1999). CJ, Chen HM, Hiebert SW and Tenen DG. (1996a). Mol.
Blood, 94, 560 ± 571. Cell. Biol., 16, 1231 ± 1240.
Wang W, Wang X, Ward AC, Touw IP and Friedman AD. Zhang P, Benson GV, Rhoades KL, Maas RL and Tenen
(2001). Leukemia, 15, 779 ± 786. DG. (1996b). Blood, 88, 631a.
Wang Q and Friedman AD. (2002). Blood, 99, (in press). Zhang DE, Zhang P, Wang ND, Hetherington CJ,
Ward AC, Loeb DM, Soede-Bobok AA, Touw I and Darlington GJ and Tenen DG. (1997). Proc. Natl. Acad.
Friedman AD. (2000). Leukemia, 14, 973 ± 990. Sci. USA, 94, 569 ± 574.
Weinberg RA. (1995). Cell, 81, 323 ± 330. Zhang P, Iwama A, Datta MW, Darlington GJ, Link DC
Westendorf JJ, Yamamoto CM, Lenny N, Downing JR, and Tenen DG. (1998). J. Exp. Med., 188, 1173 ± 1184.
Selsted ME and Hiebert SW. (1998). Mol. Cell. Biol., 18, Zhang P, Zhang X, Iwama A, Yu C, Smith KA, Mueller BU,
322 ± 333. Narravula S, Torbett BE, Orkin SH and Tenen DG.
Weston K and Bishop MJ. (1989). Cell, 58, 85 ± 93. (2000). Blood, 96, 2641 ± 1648.
Williamson E, Xu HN, Gombart AF, Verbeek W, Chuma-
kov AM, Friedman AD and Koe‚er HP. (1998). J. Biol.
Chem., 273, 14796 ± 14808.

Oncogene

You might also like