Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

Review article

Signaling pathways governing stem-cell fate


Ulrika Blank,1 Göran Karlsson,1 and Stefan Karlsson1
1Molecular Medicine and Gene Therapy, Institute of Laboratory Medicine and Lund Strategic Research Center for Stem Cell Biology and Cell Therapy, Lund

University Hospital, Lund, Sweden

Hematopoietic stem cells (HSCs) are his- mained elusive. Recently, several develop- tion occurs, concomitantly with provid-
torically the most thoroughly character- mentally conserved signaling pathways ing information of how the bone marrow
ized type of adult stem cell, and the have emerged as important control de- microenvironment couples and integrates
hematopoietic system has served as a vices of HSC fate, including Notch, extrinsic with intrinsic HSC fate determi-
principal model structure of stem-cell bi- Wingless-type (Wnt), Sonic hedgehog nants. It is the focus of this review to

Downloaded from http://ashpublications.org/blood/article-pdf/111/2/492/1220086/zh800208000492.pdf by guest on 29 January 2024


ology for several decades. However, para- (Shh), and Smad pathways. HSCs reside highlight some of the most recent devel-
doxically, although HSCs can be defined in a complex environment in the bone opments concerning signaling pathways
by function and even purified to near- marrow, providing a niche that optimally governing HSC fate. (Blood. 2008;111:
homogeneity, the intricate molecular ma- balances signals that control self- 492-503)
chinery and the signaling mechanisms renewal and differentiation. These signal-
regulating fate events, such as self- ing circuits provide a valuable structure
renewal and differentiation, have re- for our understanding of how HSC regula- © 2008 by The American Society of Hematology

Introduction
In tissues with a high cellular turnover, stem-cell populations and more restricted in their differentiation capacity, finally
are essential for lifelong maintenance of organ function. At generating functionally mature cells (Figure 2). Phenotypic
present, somatic stem cells have been identified in several characterization of HSCs has allowed for their prospective
self-renewing organs, including intestinal and epidermal epithe- isolation, and several groups have reported the successful
lia as well as the blood cell system. Residing in the bone marrow regeneration of all blood cell lineages after transplantation of a
(BM) of adults, hematopoietic stem cells (HSCs) are ultimately single HSC into lethally irradiated mice.2,3 All functional HSCs
responsible for the continuous daily production of all mature are associated with lack of expression of a range of cell surface
blood cell lineages.1 HSCs are functionally defined at the markers normally found on differentiating or mature blood cells
single-cell level by their dual capacity for self-renewal and while displaying high levels of Sca1 and c-kit.4,5 This population
multipotential differentiation. Self-renewal pertains to the pro- of cells is commonly referred to as the LSK compartment
cess whereby at least one daughter cell of a dividing HSC retains (lineage⫺/Sca1⫹/c-kit⫹) and 100 LSK cells have been shown to
stem-cell fate and may be either symmetric or asymmetric in its be sufficient for long-term multilineage repopulation of lethally
nature (Figure 1). A symmetric self-renewing division refers to irradiated hosts.5 Based on their ability to self-renew, HSCs can
the process whereby both daughter cells retain stem cell be divided into long-term and short-term reconstituting HSCs
properties. Theoretically, this type of cell division expands the (LT-HSCs and ST-HSCs, respectively). LT-HSCs have extensive
stem-cell pool and is therefore thought to be important after self-renewal capacity and can sustain lifelong hematopoiesis,
transplantation or after hematopoietic injury. In an asymmetric whereas ST-HSCs are restricted in self-renewal potential,
self-renewing division, the 2 daughter cells adopt different sustaining hematopoiesis only for a limited time in vivo.6-8 With
fates, resulting in one cell maintaining stem-cell properties. respect to self-renewal, the LSK compartment is heterogeneous,
On a population basis, this allows for steady-state main- containing a mixture of LT-HSCs, ST-HSCs, and multipotent
tenance of the HSC compartment. Self-renewal is critical progenitor cells (MPPs). Further fractionation based on differen-
for sustaining the HSC compartment and thus is a prerequisite for tial expression of CD34 and Flt3 has been shown to correlate
lifelong hematopoiesis. with distinct functional properties, with LT-HSCs being nega-
tive for both CD34 and Flt3.3,6,7 In addition to cell-surface
staining, exclusion of fluorescent dyes, such as Hoechst and
rhodamine, has been shown to correlate with HSC activity.9 The
Hematopoietic hierarchy side population defines a small and distinct subset of cells
with LT-HSC characteristics.10 More recently, Kiel et al identi-
The hematopoietic system is hierarchically organized, with a fied the SLAM family of cell surface receptors that were
rare population of HSCs giving rise to progeny that progres- shown to be differentially expressed among stem and
sively lose self-renewal potential and successively become more progenitor populations.11

Submitted July 20, 2007; accepted September 30, 2007. Prepublished online as © 2008 by The American Society of Hematology
Blood First Edition paper, October 3, 2007; 10.1182/blood-2007-07-075168.

492 BLOOD, 15 JANUARY 2008 䡠 VOLUME 111, NUMBER 2


BLOOD, 15 JANUARY 2008 䡠 VOLUME 111, NUMBER 2 SIGNALING PATHWAYS AND STEM CELL FATE 493

Blood vessel

Symmetric
HSC
HSC HSC
HSC

Migration

Downloaded from http://ashpublications.org/blood/article-pdf/111/2/492/1220086/zh800208000492.pdf by guest on 29 January 2024


Self-renewal

Asymmetric
Apoptosis HSC

Differentiation

Figure 1. HSC fate decisions. During or after cell division, the 2 daughter cells of an HSC are faced with several options, or fate decisions. These include the possibility to
remain a HSC (the process of self-renewal), which can be either symmetric or asymmetric in its nature, to commit along the path of differentiation or to go through apoptosis. In
addition, the option to move to other anatomic sites in or outside the BM (migration) can be regarded as a fate decision, which is particularly important during specific stages of
ontogeny when HSCs seed the fetal liver or later the BM.

Efficient isolation of human HSCs has proven more difficult, stress, which is why caution should be taken when interpreting
primarily as a consequence of lack of appropriate in vivo assays. obtained data.
As of today, the rhodaminelow fraction of the Lin⫺CD34⫹CD38⫺
population of human cord blood (CB), containing putative HSCs
at a frequency of 1 in 30 cells, provides the most stringent
isolation of human HSCs.12 Populations containing putative Regulation of hematopoietic stem cells, a
human HSCs can be functionally analyzed in nonobese diabetic/ dynamic state of balance
severe combined immune deficient (NOD/SCID) mouse strains.13
SCID repopulating cells (SRCs) represent the most primitive To meet the need of a variety of situations, ranging from normal
hematopoietic population assessable, but NOD/SCID assays are homeostasis to acute blood loss or infection, hematopoiesis must be
hampered by the generally low engraftment of human cells and rapidly yet meticulously controlled. Whether regarded as stochastic
the difficulty in keeping NOD/SCID mice for long-term analy- mainly regulating probabilities or deterministic providing more
ses. This predicament can be overcome by serial NOD/SCID precise instructions, both intrinsic and extrinsic signals undoubt-
transplantations, allowing for analysis of even more primitive edly participate in the regulation of HSCs.1,14-16 Through reduction-
populations. However, such assays keep HSCs under constant istic strategies, a wide variety of factors critical for HSC regulation
494 BLANK et al BLOOD, 15 JANUARY 2008 䡠 VOLUME 111, NUMBER 2

B cell

T cell

CLP

LSK compartment NK cell

Downloaded from http://ashpublications.org/blood/article-pdf/111/2/492/1220086/zh800208000492.pdf by guest on 29 January 2024


Granulocyte
LT-HSC ST-HSC MPP

GMP
Thy1.1 low Thy1.1 low Thy1.1- Macrophage
FLT3- FLT3- FLT3+
CD34- CD34+ CD34+
Endoglin+ CD11b low CD11b low
SP+ CD4 low
Rho low CMP
CD150+
Platelet

MEP
Erythrocyte

Figure 2. The hematopoietic hierarchy and phenotypic markers associated with HSCs. The LSK compartment contains LT-HSCs, ST-HSCs, and MPPs, each
subpopulation of which is associated with distinct phenotypic features as indicated. CLP indicates common lymphoid progenitor; CMP, common myeloid progenitor; GMP,
granulocyte/macrophage progenitor; and MEP, megakaryocyte/erythrocyte progenitor.

have been identified, including cytokines, growth factors, transcrip- HSCs have failed because of differentiation of HSCs and subse-
tion factors, chromatin modifiers, and cell-cycle regulators. Delin- quent loss of reconstitution capacity. In one study, a modest
eating the signaling pathways that integrate these diverse compo- expansion of repopulating murine HSCs was observed using a
nents remains an enduring challenge of foremost importance, 10-day culture protocol in the presence of IL-11, Flt-3 ligand, and
which would have tremendous impact on regenerative medicine. SCF.17 However, the same conditions failed to expand HSCs
During the course of development, a handful of signaling pathways derived from the fetal liver and later studies have found that the
shape the structures of the embryo and determine cell fate and Flt-3 receptor is not expressed on LT-HSCs.7,18 Furthermore, the
identity. In the adult organism, these signaling pathways are gp130 protein, which is a part of the receptor for IL-6 and IL-11
reiterated, and it is becoming increasingly clear that most somatic was suggested to play a role in the self-renewal of HSCs.19
stem-cell compartments are under the influence of developmentally However, elimination of the IL-11 receptor does not affect
conserved signals. hematopoiesis, making conclusions of the in vivo relevance
uncertain.20 Detailed studies of purified murine HSCs in recent
years have shown that the receptors for TPO and SCF, c-mpl and
Regulation of HSCs by classic hematopoietic c-kit, respectively, are both expressed on repopulating HSCs,6,7,21-24
cytokines and it has been shown that mice with genetic mutations in TPO or
c-mpl have a reduction in HSCs.21,22 Moreover, TPO has been
Numerous attempts have been made to use classic hematopoietic reported to support viability25 and suppressed apoptosis of HSCs.26
cytokines for the purpose of expanding HSCs in vitro. Many of the Thus, the main function of TPO may be to counteract apoptosis of
interleukins, particularly, interleukin (IL)-3, IL-6, and IL-11, as HSCs rather than promote their expansion.24
well as Flt-3 ligand, thrombopoietin (TPO) and stem-cell factor A further role for TPO in regulating HSC self-renewal was recently
(SCF) have been investigated. In most cases, efforts to expand suggested through the association with Lnk, an intracellular adaptor
BLOOD, 15 JANUARY 2008 䡠 VOLUME 111, NUMBER 2 SIGNALING PATHWAYS AND STEM CELL FATE 495

Downloaded from http://ashpublications.org/blood/article-pdf/111/2/492/1220086/zh800208000492.pdf by guest on 29 January 2024


Figure 3. Notch and Wnt signaling pathways. (A) Notch pathway: When ligands of the Delta (Delta1-3) or Jagged (Jagged 1-2) families bind to the Notch receptor,
proteolytic events involving ␥-secretase lead to release and translocation of the intracellular domain of the receptor (NICD) to the nucleus. Subsequently, NICD will form
a complex with the transcription factor CSL and cofactors of the Mastermind-like (MAML) family to activate transcription of target genes.138 (B) Wnt pathway: Wnt
signaling is initiated when a ligand binds to the Frizzled and lipoprotein receptor-related protein (LRP) receptors at the cell surface. In the absence of Wnt ligands, the
downstream signal transducer ␤-catenin is trapped by adenomatous polyposis coli (APC) and Axin in a destruction complex, where it is phosphorylated by
casein-kinase 1␣ (CK1␣) and glycogen synthase kinase (GSK3␤). Phosphorylation ultimately leads to ubiquitination and degradation of ␤-catenin. On ligand binding,
Frizzled forms a complex with disheveled (Dsh), whereas LRP is phosphorylated, resulting in Axin relocation to the cell membrane. Subsequently, the destruction
complex is dispersed and ␤-catenin accumulates and translocates to the nucleus where it interacts with the T-cell factor/lymphoid enhancer factor (TCF) transcription
factors to regulate gene expression.139,140

molecule functioning as a broad inhibitor of several cytokine signaling progenitors in vitro as measured by reconstitution assays.35-41
pathways, including TPO, SCF, erythropoietin, IL-3, and IL-7.24,27-30 Importantly, enforced activation of the Notch signaling pathway
Mice deficient in Lnk have been reported to exhibit increased HSC or the downstream target Hes-1, have been demonstrated to
numbers and their repopulation function was elevated, a phenotype increase the self-renewal capacity of long-term in vivo repopu-
attributable to augmented proliferation and self-renewal.24,27,31 Interest- lating HSCs42,43 or even to immortalize primitive hematopoietic
ingly, the increased self-renewal capacity of Lnk⫺/⫺ HSCs was depen- progenitor cells.44 When Calvi et al established osteoblasts as
dent on TPO, and Lnk-negative HSCs were shown to be hypersensitive putative members of the HSC niche, they demonstrated how
to stimulation by TPO.24,27 Thus, TPO and Lnk serve opposing roles in constitutively active parathyroid hormone receptor expanded
the regulation of HSC self-renewal and Lnk acts as a powerful inhibitor these cells and stimulated their expression of Jagged-1.45 The
of TPO signaling in HSCs. To conclude, of the classic hematopoietic activated osteoblasts were able to increase self-renewal of
cytokines, the most important positive regulators of HSCs are
primitive hematopoietic cells as measured by LTC-ICs and
TPO and SCF.
competitive repopulation assays.45 Moreover, LSK cells from
these transgenic mice had increased levels of Notch intracellular
domain (NICD), and the elevated LTC-IC numbers could be
Notch pathway: at the interface of osteoblasts normalized with a ␥-secretase inhibitor.45 Supporting these data
and HSCs were the observation by Duncan et al that a Notch reporter was
active in c-kit⫹ hematopoietic cells in the trabecular bone and
In humans, Notch was discovered 15 years ago as an oncogene
isolated LSK and side population cells.46
that, after chromosomal translocation, gave rise to T-cell
leukemia.32 Since then, several loss- and gain-of-function Together these findings indicate not only that osteoblasts are
studies have demonstrated a critical role for Notch in lineage part of the so-called HSC niche but also that Jagged/Notch
specification of lymphopoiesis, where active Notch signaling signaling activated by these cells might be important in the
(Figure 3A) drives differentiation toward T cells.33 It was soon extracellular regulation of HSC self-renewal.45 However, HSCs
suggested that Notch could have a role in HSC biology as deficient in Notch-1 could reconstitute Jagged-1 deficient hosts in a
primitive hematopoietic cells as well as cells in the putative normal fashion, suggesting that Notch signaling through mecha-
HSC microenvironment expressed both Notch and Notch-ligand nisms including these molecules is dispensable for in vivo HSC
mRNA.34,35 Indeed, through various approaches, several groups function.47 It is probable that the lack of phenotype observed in this
have shown that members of the Notch ligand families, Delta setting is the result of compensatory mechanisms mediated by
and Jagged, can expand both murine and human hematopoietic other Notch receptors and ligands, which is why approaches to
496 BLANK et al BLOOD, 15 JANUARY 2008 䡠 VOLUME 111, NUMBER 2

broadly eliminate Notch signaling would be informative. Regard-


less of what the case may be, manipulation of the Notch signaling Integration of Wnt and Notch signals in
pathway holds great promise for ex vivo expansion of HSCs in HSC regulation
future clinical protocols.
Recently, an intriguing report presented evidence that Notch and
Wnt pathways act in synergy to maintain the HSC pool as both
signaling circuits were shown to be active simultaneously in a large
HSC self-renewal under control by the Wnt fraction of cells in the trabecular bone.46 Even though Notch- and
signaling pathway Wnt signaling both are implied to stimulate self-renewal of HSCs,
Duncan et al propose that they do so through different mecha-
A role for Wnt signaling in the regulation of murine HSCs was nisms.46 As Wnt signaling seems to induce proliferation and
suggested a decade ago when expression of different Wnts was support viability of LSK cells, inhibition of the Notch pathway had
discovered at sites of fetal hematopoiesis and Wnt5a was shown to no effect on these functions.46 Instead, Notch signaling was
stimulate proliferation and self-renewal of fetal HSCs/progenitors demonstrated to be imperative in maintaining HSCs in an undiffer-

Downloaded from http://ashpublications.org/blood/article-pdf/111/2/492/1220086/zh800208000492.pdf by guest on 29 January 2024


in vitro.48 Adult HSCs also appear to respond to Wnt treatment entiated state, even if Wnt3a was present in the culture.46 These
because exposure to purified Wnt3a was recently shown to increase findings suggest that Wnt and Notch signaling together could play a
self-renewal of murine HSCs in vitro, as determined by in vivo role in self-renewal of HSCs. Moreover, observations that Wnt3a
reconstituting assays.49 Moreover, Van den Berg et al demonstrated regulate expression of established Notch target genes46 and that
that the positive effects on HSCs were not exclusive to the murine inhibition of the Wnt signaling component GSK-3 affect HSC fate
options through mechanisms involving regulation of both Wnt and
setting because human Lin⫺CD34⫹ cells exposed to WNTs
Notch target genes54 suggest that the 2 pathways belong to a
also expanded in vitro as measured by immunophenotype and
network of regulatory circuits controlling the HSC pool.
colony assays.50
Not only is there evidence supporting the hypothesis that Wnts
could be used as tools for expanding HSCs in culture, but WNT Smad signaling pathway
proteins have also been reported to be expressed in both human
fetal BM stroma and adult BM, where WNT5A has been shown to The Smad signaling pathway embodies an evolutionary ancient
be expressed in the primitive Lin⫺CD34⫹ population.50 This would signaling circuitry, which functions to transduce signals down-
suggest that Wnts are physiologically important and act on HSCs stream of the transforming growth factor-␤ (TGF-␤) family of
through both paracrine and autocrine mechanisms. In support of the ligands (Figure 4). Apart from TGF-␤, activins and bone morphoge-
latter statement was the finding that exposure of HSCs in vitro to a netic proteins (BMPs) also belong to this superfamily, which
soluble Wnt binding antagonist reduced their proliferation capac- regulate a bewildering array of fundamental biologic processes
ity.51 However, the intracellular mechanisms through which Wnts during development and postnatally. Because of the highly redun-
exercise their effect on HSCs are somewhat inconclusive. Canoni- dant nature of the Smad pathway and early embryonic lethality of
cal Wnt signaling (Figure 3B) occurs through mechanisms involv- most knockout mouse models, the precise role of the Smad
ing the stabilization and nuclear translocation of ␤-catenin, fol- circuitry in hematopoiesis has remained nebulous. However, re-
lowed by transcriptional regulation of target genes by a ␤-catenin/ cently several reports have shed new light suggesting that this
T-cell factor complex. Reya et al detected active canonical Wnt pathway plays a pivotal role in the regulation of HSC fate
signaling, as measured by a T-cell factor reporter construct, in a decisions.
large proportion of the HSC enriched LSK compartment in adult TGF-␤, key negative modulator of HSCs in vitro
BM.51 Intriguingly, retroviral–mediated enforced expression of
␤-catenin resulted in a 102 to 103 fold increase in HSCs during TGF-␤ is generally catalogued as one of the most potent inhibitors
long-term cultures of KTLS cells as defined by both phenotype and of HSC growth in vitro, and ample evidence from a variety of
function.51 In accordance with the observed phenotype resulting culture systems exist to support this fact.55-57 A hallmark feature of
from forced expression of ␤-catenin was an up-regulation in HSCs is their relative quiescence, and given the strong growth
expression of the HSC self-renewal stimulating genes Notch1 and inhibitory properties of TGF-␤, it has naturally been hypothesized
HoxB4.51 However, when ␤-catenin was conditionally expressed in to be a cardinal regulator of HSC quiescence in vivo. In keeping
with this, neutralization of TGF-␤ in vitro was shown to release
vivo under the control of the ROSA26 locus, no increase in HSC
early hematopoietic progenitor cells from quiescence.58-60 Several
self-renewal was observed.52 Adding to the contradiction, deletion
molecular mechanisms have been proposed to account for TGF-␤–
of the ␤-catenin gene in HSCs failed to result in an in vivo
mediated growth inhibition, including alterations in cytokine
phenotype.53
receptor expression and up-regulation of cyclin-dependent kinase
The reasons for these discrepancies are unclear but may reflect a inhibitors, such as p21 and p57.59,61-69
stronger role for Wnt signaling in in vitro expansion of HSCs as TGF-␤ can affect most cell types throughout the hematopoietic
opposed to its function in vivo. The more complex and enduring in hierarchy and depending on the context and differentiation stage of
vivo setting might allow for redundant mechanisms to develop. the target cell different biologic responses are ultimately elic-
Alternatively, the HSC expansion observed when ␤-catenin was ited.70-72 In vivo, TGF-␤ plays a principal role as regulator of
retrovirally overexpressed might be affected by the use of Bcl-2 immune cell homeostasis and function, as unambiguously evi-
transgenic HSCs in that particular study.51 Thus, even though Wnts denced by the development of a lethal inflammatory disorder of
may possibly be useful for expanding HSCs in vitro, the unresolved both TGF-␤1- ligand and receptor knockout mice.73-75 Further-
mechanisms behind the effect and the question pertaining to the more, Tgf-␤1 null mice exhibited enhanced myelopoiesis, suggest-
role of canonical Wnt signaling remains to be determined. ing that TGF-␤ acts as a negative regulator of myelopoiesis in
BLOOD, 15 JANUARY 2008 䡠 VOLUME 111, NUMBER 2 SIGNALING PATHWAYS AND STEM CELL FATE 497

Figure 4. Smad signaling pathway. Smad pathway:


TGF-␤ family members bind and signal through 2 types
of serine/threonine kinase receptors, type I and type II,
both of which are necessary for signal transduction. On
ligand binding and receptor activation, the Smad pro-
teins are activated through phosphorylation by type I
receptors.141,142 Three groups of Smads have been Ligand Activin/Nodal TGF-beta BMP
identified: receptor-activated Smads (R-Smads), common-
partner Smads (Co-Smads), and inhibitory Smads
(I-Smads). In general, TGF-␤ and activin signal via
R-Smad2 and 3, whereas BMP signals are transduced
Receptor
through R-Smad1, 5, and 8. Phosphorylated R-Smads
subsequently associate with the Co-Smad4, creating a
complex that translocates to the nucleus where target
gene transcription is modified. In contrast to R- and ActR-IIA BMPR-II
ActR-IIB ActR-IIA
Co-Smads, the I-Smads, Smad6 and Smad7, function TGF-b RII
ActR-IIB
in a negative feedback loop to prevent activation of
ALK4
R-Smads.143-145 Divergence and convergence of the ALK1 ALK2
ALK7 ALK5
Smad signaling circuitry are depicted. Commonly used ALK3

Downloaded from http://ashpublications.org/blood/article-pdf/111/2/492/1220086/zh800208000492.pdf by guest on 29 January 2024


alternative names include: ALK2/Activin type I receptor, ALK6
ALK3/BMP type IA receptor, ALK4/Activin type IB recep- I-Smads Smad1
tor, ALK5/TGF-␤ type I receptor, and ALK6/BMP type IB
Smad6
receptor. The shaded portion indicates the nucleus. Smad2 Smad5 R-Smads
P indicates phosphorylation. R-Smads Smad7
Smad3 Smad8

Smad4 Co-Smad

Nucleus Cytoplasm

DNA-binding partner
P
R-Smad
Smad4
P
R-Smad
Smad4
Target gene

vivo.76 However, despite the pronounced role of TGF-␤ in vitro, formation was assessed, indicative of increased self-renewal.
mice deficient of the TGF-␤ type I receptor displayed normal HSC However, conditional deletion of Smad5 in adult mice does not
self-renewal and regenerative capacity in vivo, even under extreme affect hematopoiesis, and HSCs lacking Smad5 have a normal
hematopoietic stress.77,78 The apparent discrepancy between in differentiation and regenerative potential in vivo.88 It is possible
vitro and in vivo findings may reflect mechanisms of redundancy that BMP–activated Smad signaling plays a more pronounced role
between other type I receptors or alternatively other ligands such as early in hematopoietic development, as opposed to adult hematopoi-
activin, which signals through the same R-Smad pathway. esis. Alternatively, redundant mechanisms within the Smad path-
way may explain the lack of phenotype in the adult setting.
BMP promotes maintenance of HSCs in culture
Smad signaling: toward a deeper understanding
BMPs have been implicated as key regulators of hematopoietic develop-
ment in a variety of species, from zebrafish to mouse.79-83 In the context To block the entire Smad signaling pathway, thus sidestepping
of adult hematopoiesis, BMP-4 was shown to promote maintenance of redundant mechanisms, the inhibitory Smad7 was overexpressed in
HSCs in culture, whereas lower concentrations of BMP-4 induced murine HSCs using a retroviral gene transfer approach.89 Forced
proliferation and differentiation of human hematopoietic progenitors.84 expression of Smad7 significantly increased the self-renewal
Furthermore, Shh induced proliferation of primitive human hematopoi- capacity of HSCs in vivo, indicating that the Smad pathway
etic progenitors in vitro, apparently through a BMP-4–dependent negatively regulates self-renewal in vivo. In a similar approach
mechanism.85 However, although BMP-4 has been shown to maintain using human SCID repopulating cells (SRCs), overexpression of
human NOD/SCID repopulating cells in culture, it does not seem to Smad7 resulted in a shift from lymphoid dominant engraftment
cause an expansion, suggesting that Shh may act through additional toward increased myeloid contribution.90 Thus, in the xenograft
mechanisms. In the murine system, BMP-4 does not appear to affect model system, forced expression of Smad7 modulates cell fate
proliferation of purified HSCs in vitro, although it is currently unclear decisions of primitive multipotent human SRCs.
whether BMP-4 can extend the maintenance of murine HSCs in It is beyond doubt that the Smad signaling pathway lies at the
culture.86 Furthermore, BMPs function to regulate the HSC niche, thus very core of mediating signals from the TGF-␤ family of ligands.
indirectly controlling HSC numbers as discussed in “HSC niche.” However, a considerably more diversified picture is now emerging,
A role for Smad5, traditionally characterized as a mediator of which indicates that the Smad circuitry is more varied than
BMP signals, in self-renewal was implicated through a series of in previously thought.91 Using a conditional knockout mouse model,
vitro studies. Interestingly, the number of colonies, derived from disruption of the entire Smad pathway at the level of Smad4 was
yolk sacs or ES cell differentiated embryoid bodies deficient of recently investigated. Intriguingly, Smad4 deficient HSCs dis-
Smad5, were found to be increased.87 Smad5 deficient colonies also played a significantly reduced repopulative capacity of primary and
had an enhanced regenerative potential when secondary colony secondary recipients, indicating that Smad4 is critical for HSC
498 BLANK et al BLOOD, 15 JANUARY 2008 䡠 VOLUME 111, NUMBER 2

self-renewal in vivo.92 Because overexpression of Smad7 versus specifically, a 24- to 30-fold increase in HSC number was
deletion of Smad4 would be anticipated to yield similar hematopoi- observed. Angptl proteins 5 and 7 were also reported to expand
etic phenotypes, it is conceivable that Smad4 functions as a murine HSCs, whereas Angptl protein 4 failed to show a similar
positive regulator of self-renewal independently of its role as a effect. The molecular mechanisms and the signaling events down-
central mediator of the canonical Smad pathway. The precise stream of Angptl proteins remain unidentified and their receptors
molecular mechanism for this is currently unknown, but it is are currently unknown, which is why it is not clear how the Angptl
possible that Smad4 participates in other signaling cascades such as proteins expand HSCs. However, Angptl protein 2 has been shown
Wnt or Notch.93-95 Moreover, He et al proposed an elegant model to bind to a highly enriched population of HSCs. Therefore, the
for how TGF-␤ mediates erythroid differentiation concomitantly expansion effect attributable to these proteins is likely to be a result
with balancing growth inhibition in human hematopoietic stem/ of a direct action on HSCs. It will be exciting to see whether the
progenitor cells.96 According to this model, transcriptional interme- Angptl proteins can also be used for robust expansion of human
diary factor1␥ (TIF1␥) selectively binds Smad2 and Smad3 in HSCs, particularly HSCs derived from cord blood.
competition with Smad4. In response to TGF-␤, the TIF1␥/ These findings are important especially in light of the convenience of
Smad2/3 complex was shown to stimulate erythroid differentiation adding these factors to cultures of HSCs in vitro, and they can therefore
of human hematopoietic progenitors, whereas Smad2/3 in associa- be used in future attempts to expand HSCs for the clinic and possibly

Downloaded from http://ashpublications.org/blood/article-pdf/111/2/492/1220086/zh800208000492.pdf by guest on 29 January 2024


tion with Smad4 inhibited proliferation of the same cells. Thus, the also to increase the efficiency of gene delivery to HSCs.
relative abundance of Smad4 and TIF1␥ is likely to be important
for determining the effect of TGF-␤ stimulation. Interestingly, the
zebrafish homolog of TIF1␥, encoded by moonshine, has been HSC niche
shown to be essential for blood formation with mutants displaying
severe red cell aplasia, indicating that the function of TIF1␥ may be On the role of osteoblasts
preserved across species.97 The spatial organization of hematopoietic cells within the BM,
However, the mechanism through which TIF1␥ cooperates with primitive cells distributed closer to the bone surface and
with Smads is not entirely clear. Evidence from Xenopus has more differentiated cells positioned closer to the BM longitudi-
suggested that TIF1␥, known as Ectodermin, acts as a ubiquitin nal axis of the femur, has long been recognized.102,103 As early as
ligase for Smad4, thus functioning as a direct inhibitor of Smad4 1978, Schofield proposed the niche hypothesis of a physiologi-
downstream of TGF-␤ and BMP signaling.98 This dichotomy cally limited microenvironment supporting stem cells in vivo.104
may reflect differences in cellular context or alternatively Because of the anatomic complexity of mammalian stem cell
species or temporal differences. Another interesting observation niches, the precise location as well as the cellular and molecular
is the association between Smad4 and Hox proteins. Homeobox basis for these structures has only recently begun to be
(hox) genes encode transcription factors that function as para- uncovered. Several lines of evidence now converge to suggest
mount regulators of hematopoiesis and are frequently dysregu- that the osteoblasts, cells of mesenchymal origin positioned at
lated in human leukemia, particularly acute myeloid leukemia. the endosteal surface of bone, are essential components of the
Enforced expression of HoxA9 immortalizes and blocks my- HSC niche.105 Using a transgenic mouse model expressing a
eloid differentiation, eventually causing acute myeloid leuke- constitutively active Parathyroid hormone receptor under an
mia.99 Recently, Wang et al described a mechanism whereby osteoblast specific promoter (␣1(I) collagen), Calvi et al demon-
TGF-␤/BMP inhibited the bone marrow transformation capacity strated that an increase in trabecular osteoblasts was accompa-
of HoxA9 and HoxA9-Nup98 fusion protein through a Smad4- nied by an increase in HSC numbers.45 Importantly, Calvi et al45
dependent mechanism. Accordingly, Smad4 was shown to propose a model in which increased parathyroid hormone
interact directly with HoxA9 and Nup98-HoxA9 fusion protein, signaling in osteoblasts results in elevated levels of the Notch1
thus precluding their DNA binding capacity and subsequent ligand, Jagged1, ultimately generating increased activation of
transcriptional activity.100 Whether Smad4 mutations facilitate Notch1 signaling in HSCs. An independent study investigated
leukemogenesis through increased Hox activity remains an the role of the BMP signaling pathway to regulate HSC
unanswered question. numbers, by conditional inactivation of the BMP type IA
receptor Alk3.106 In the absence of Alk3 the number of spindle
shaped osteoblastic cells expressing N-cadherin were increased.
Angiopoietin-like proteins Interestingly, this correlated with an increase in the number of
HSCs, which were found attached to N-cadherin⫹ osteoblasts.
Building on the fact that the fetal liver (FL) is a powerful site of Moreover, depletion of osteoblasts in vivo was associated with
HSC expansion during development, Lodish et al embarked on an decreased BM cellularity and extramedullary hematopoiesis,
approach to identify “stromal” cells and their expression pattern of further emphasizing the importance of osteoblasts for HSC
molecules responsible for stimulation of HSC self-renewal in the function.107 In addition, Arai et al identified Tie2/Angiopoietin1
murine FL. Ter119⫹CD3⫹ cells were recognized as a population of signaling to be critical for the maintenance of HSCs in a
cells derived from the FL that could support expansion of HSCs in quiescent state in the BM niche.108 Tie2 is a receptor tyrosine
vitro. Insulin-like growth factor 2 (IGF-2) was identified on the kinase expressed on endothelial cells and HSCs, whereas
basis of being expressed in CD3⫹ FL cells and was shown to Angiopoietin1 (Ang1) is expressed by osteoblasts. Tie2⫹ HSCs
modestly expand HSCs in vitro. More interestingly, they found that adhered to Ang1 expressing osteoblasts, supposedly through a
several angiopoietin-like (Angptl) proteins were expressed in these mechanism involving N-cadherin.
support cells for HSC growth. When highly enriched HSCs were Even though osteoblasts clearly constitute part of the HSC
cultured under serum free conditions in the presence of TPO, SCF, niche, they are probably not the only players. Apart from soluble
IGF-2, and Angptl protein 2 or 3, a robust increase in repopulating factors, matrix components and other cellular constituents are
HSCs was observed after 10 days of in vitro culture.101 More likely to also have important functions. One example is
BLOOD, 15 JANUARY 2008 䡠 VOLUME 111, NUMBER 2 SIGNALING PATHWAYS AND STEM CELL FATE 499

Figure 5. The HSC niche. HSCs are positioned in


close proximity to osteoblasts at the endosteal surface
of bone. Other factors contributing to the in vivo
microenvironment of HSCs include extracellular matrix
(ECM), soluble compounds, as well as other cellular
components.
Osteoblasts

ECM

Ca 2+
Ca 2+

Endosteal bone surface


Ca 2+

HSC

Downloaded from http://ashpublications.org/blood/article-pdf/111/2/492/1220086/zh800208000492.pdf by guest on 29 January 2024


BM cell

Soluble factors

osteopontin, a matrix glycoprotein synthesized by osteoblasts, which


has been associated with negative regulation of the HSC compart- It’s not all in the niche
ment.109,110 Furthermore, HSCs deficient in the calcium-sensing receptor
were profoundly defective in localizing anatomically to the endosteal Despite the plethora of evidence pointing at the importance of the BM
niche, a behavior that correlated with diminished adhesion to the milieu, it would be premature to assume that the microenvironment is
extracellular matrix protein collagen I.111 These data indicate that the the sole regulator of HSC fate options. For instance, during ontogeny
local calcium gradient is involved in retaining HSCs in close physical before the establishment of the BM niche, HSCs undergo extensive
proximity to the endosteal surface of bone. Furthermore, it was recently self-renewal in the fetal liver. Indeed, HSCs derived from fetal liver have
shown that a defective niche could result in HSC disorders further been shown to encompass a more efficient regenerative potential
emphasizing the regulatory function of the HSC niche in vivo.112,113 compared with their adult counterparts when transplanted into irradiated
recipients.118 Recent evidence uncovered by Bowie et al suggest the
Hypoxia and reactive oxygen species (ROS)
existence of an intrinsically regulated developmental switch, which
Another peculiar property of the BM microenvironment is its abruptly changes the self-renewing properties of HSCs at the age of 3 to
hypoxic nature, a fact recently highlighted by Parmar et al showing 4 weeks in the mouse.119 At this time the HSC compartment rapidly
that HSCs are distributed predominantly at the lowest end of an changes from a fully cycling population to a largely quiescent one.119
oxygen gradient within the BM.114 This observation bears impor- This change appears to be independent of the niche as it occurs well after
tance for several findings, which indicate that the level of oxidative the migration of HSCs to the BM and because it is not affected by
stress influences HSC function. For example, mice deficient of the transplantation into adult recipients.120 Interestingly, juvenile HSCs
cell-cycle regulator Atm developed early onset BM failure, a exhibit a greater sensitivity to SCF; thus, signaling events downstream
phenotype accompanied by elevated levels of ROS in HSCs.115 of the c-kit receptor may be developmentally and intrinsically regulated,
Interestingly, ROS appear to activate the p38/MAPK pathway ultimately determining symmetric versus asymmetric self-renewal.121
causing quiescent HSCs to cycle more frequently and eventually To summarize, HSC behavior is regulated by joint efforts from intrinsic
become exhausted.116 Moreover, members of the FoxO subfamily factors and environmental cues, both entities that are integrated in a
of forkhead transcription factors have been shown to protect HSCs more comprehensive and holistic view by the concept of the stem-cell
from oxidative stress by up-regulating genes involved in their niche (Figure 5).
detoxification. Triple knockout mice of FoxO1, FoxO3, and FoxO4
exhibited defective long-term repopulating activity that correlated
with increased cycling and apoptosis of HSCs as well as increased Stem-cell expansion for cell and gene therapy
levels of ROS.117 Similarly, the HSC compartment of FoxO3a null
mice suffers from augmented levels of ROS.146 As previously
reported for mice deficient of Atm, the HSC defect resulting from The process of finding donors that have compatible histocompatibil-
loss of FoxOs could be rescued by administration of the antioxidant ity antigens for patients who need blood and marrow transplanta-
N-acetyl-L-cysteine. In light of these observations, it is conceiv- tion is often a challenge of significant magnitude. As of today,
able that the hypoxic environment in which the HSCs reside may umbilical CB is being used increasingly as a source of HSCs because of
serve to protect them from oxygen radicals ultimately keeping the common availability of CB cells and the diversity of histocompatibil-
them quiescent. ity gene haplotypes that are available in banked CB samples.122 The
500 BLANK et al BLOOD, 15 JANUARY 2008 䡠 VOLUME 111, NUMBER 2

number of HSCs in each CB sample is limited, and it would therefore relatively short and safe culture period, suitable for GMP conditions that
greatly increase the applicability of this stem-cell source if the CB stem will be required for the clinic. Worthy of mention are also fibroblast
cells could be expanded ex vivo before transplantation. Stem-cell growth factors, which have been shown to sustain primitive murine
expansion can also theoretically be accomplished in vivo after engraft- hematopoietic cells in culture and to maintain their primitive pheno-
ment of the transplanted cells, but this approach may involve greater type.131,132 Interestingly, fibroblast growth factor-1 in conjunction with
risks compared with ex vivo expansion unless the growth stimuli used in SCF, TPO, and IGF-2 resulted in a 20-fold increase of LT-HSCs during a
vivo can be carefully controlled.123,124 Moreover, because culture 10-day culture period.133
conditions can be precisely defined and transient modulation of regula- It should be emphasized that most of the knowledge about stem- cell
tory pathways more easily used in vitro, stem-cell expansion ex vivo expansion has come from studies in mice. There is a difference between
presents an attractive approach. However, even this tactic has proven a mouse and human HSCs with regards to cytokine receptors, telomere
great challenge because a successful stem-cell expansion involves biology, and proliferative capacity, and there will therefore be both
symmetric self-renewal divisions of HSCs, where both daughter cells differences and similarities in the approaches used to expand mouse and
retain HSC properties.123 More commonly, HSCs grown in vitro human HSCs. Recently, the protein nephroblastoma overexpressed
undergo asymmetric divisions characterized by the production of one (NOV/CCN3) has been shown to expand primitive human HSCs, and
HSC and a more differentiated progenitor, or alternatively, a symmetric this protein should therefore be carefully evaluated for use in clinical

Downloaded from http://ashpublications.org/blood/article-pdf/111/2/492/1220086/zh800208000492.pdf by guest on 29 January 2024


division where both progeny cells have lost their HSC potential. stem-cell expansion protocols.134 As more detailed knowledge is un-
Positive and negative regulators ultimately balance the transi- earthed concerning the regulatory pathways that govern HSC self-
tion from quiescence to proliferation of HSCs. Thus, the strategies renewal, it may even be possible to modulate these pathways with small
for stem-cell expansion should involve activation of regulators that molecule drugs as has been shown in the modulation of Wnt signaling
encourage HSC self-renewal and/or inhibition of pathways that and the expression of p21 and HoxB4.54,135,136 As an example, chemicals
mediate quiescence, differentiation, or apoptosis of HSCs. Because that enhance prostaglandin E2 synthesis were recently reported to
of the recent advances in understanding the mechanisms that increase HSC numbers in zebrafish and mice.137 Undoubtedly, however,
control HSC self-renewal, novel methods to expand stem cells can a major challenge still ahead will be the integration of major signaling
now be developed.70,123,124 Some approaches require viral vector- pathways into networks of control mechanisms ultimately coupling
mediated gene transfer to HSCs for efficient expansion. Ideally, the extrinsic with intrinsic regulatory fate determinants. Such knowledge
vectors should be nonintegrating and mediate transient gene would open up new avenues for maintaining and expanding HSCs in
expression for safety reasons.125-127 However, the safest approaches vitro.
would involve soluble factors, for example, cytokines, developmen-
tal cues or factors like the Angptl proteins.
When the transcription factor HoxB4 was expressed in HSCs ex Acknowledgments
vivo, a more than 40-fold net expansion of murine HSCs was generated
This work was supported by the European Commission (INHERINET,
over a period of 10 to 14 days.128 Furthermore, modified HoxB4
Gene Therapy of Hematopoietic Stem Cells for Inherited Diseases, and
proteins have been used successfully to stimulate proliferation of human
CONSERT, Concerted Safety and Efficiency Evaluation of Retroviral
and murine HSCs ex vivo.129,130 The soluble form of the HoxB4 protein
Transgenesis for Gene Therapy of Inherited Diseases); Swedish Gene
could actively be taken up by HSCs and could be used without concerns
Therapy Program; Swedish Medical Research Council; Swedish Chil-
for insertional mutagenesis or the potential adverse effects that continu- dren Cancer Foundation; Clinical Research Award from Lund Univer-
ous production of HoxB4 in HSCs might generate in vivo. However, the sity Hospital; Joint Program on Stem Cell Research; Juvenile Diabetes
HSC expansion achieved by the soluble HoxB4 was quite modest Research Foundation; and Swedish Medical Research Council (S.K.),
compared with overexpression of HoxB4 within HSCs. Developmental the Wenner-Gren Foundations (U.B.), and Kungliga Fysiografiska
cues that activate Notch and Wnt signaling in HSCs may also be useful Sällskapet (G.K. and U.B.). The Lund Stem Cell Center is supported by
to increase HSC self-renewal ex vivo. As mentioned previously, Wnt3A a Center of Excellence grant in life sciences from the Swedish
has been shown to expand murine repopulating HSCs and Wnt5A Foundation for Strategic Research.
injection into NOD/SCID mice repopulated with human hematopoietic
cells increased the reconstitution and number of primitive hematopoietic
cells. Similarly, a soluble form of the Notch ligand, Jagged 1, was Authorship
reported to stimulate growth of human SRCs and may therefore be used
to expand stem cells ex vivo.35 However, the Angptl proteins are by far Contribution: All authors contributed to the writing of this
the most promising soluble factors identified to date for expansion of manuscript.
murine HSCs.101 If the angiopoietin-like proteins would prove to expand Conflict-of-interest disclosure: The authors declare no compet-
human HSCs as efficiently as in the murine system, they may become a ing financial interests.
promising tool for future cell and gene therapy approaches. Further- Correspondence: Stefan Karlsson, Molecular Medicine and
more, other positive regulatory factors, in combination with Angptl Gene Therapy, Lund University Hospital, BMC A12, 221 84 Lund,
proteins, may be able to expand human HSCs by a factor of 10 during a Sweden; e-mail: Stefan.Karlsson@med.lu.se.

References
1. Ogawa M. Differentiation and proliferation of he- Long-term lymphohematopoietic reconstitution by cell function of c-kit- and Sca-1-positive murine
matopoietic stem cells. Blood. 1993;81:2844- a single CD34-low/negative hematopoietic stem hematopoietic cells. Blood. 1992;80:3044-
2853. cell. Science. 1996;273:242-245. 3050.
2. Matsuzaki Y, Kinjo K, Mulligan RC, Okano H. Un- 4. Spangrude GJ, Heimfeld S, Weissman IL. Purifi- 6. Yang L, Bryder D, Adolfsson J, et al. Identification
expectedly efficient homing capacity of purified cation and characterization of mouse hematopoi- of Lin(-)Sca1(⫹)kit(⫹)CD34(⫹)Flt3- short-term
murine hematopoietic stem cells. Immunity. 2004; etic stem cells. Science. 1988;241:58-62. hematopoietic stem cells capable of rapidly re-
20:87-93. 5. Okada S, Nakauchi H, Nagayoshi K, Nishikawa constituting and rescuing myeloablated transplant
3. Osawa M, Hanada K, Hamada H, Nakauchi H. S, Miura Y, Suda T. In vivo and in vitro stem recipients. Blood. 2005;105:2717-2723.
BLOOD, 15 JANUARY 2008 䡠 VOLUME 111, NUMBER 2 SIGNALING PATHWAYS AND STEM CELL FATE 501

7. Adolfsson J, Borge OJ, Bryder D, et al. Upregula- suppression by inhibiting p53-dependent apopto- 45. Calvi LM, Adams GB, Weibrecht KW, et al. Os-
tion of Flt3 expression within the bone marrow sis. Blood. 2001;98:2084-2090. teoblastic cells regulate the haematopoietic stem
Lin(-)Sca1(⫹)c-kit(⫹) stem cell compartment is 27. Seita J, Ema H, Ooehara J, et al. Lnk negatively cell niche. Nature. 2003;425:841-846.
accompanied by loss of self-renewal capacity. regulates self-renewal of hematopoietic stem 46. Duncan AW, Rattis FM, DiMascio LN, et al. Inte-
Immunity. 2001;15:659-669. cells by modifying thrombopoietin-mediated sig- gration of Notch and Wnt signaling in hematopoi-
8. Morrison SJ, Weissman IL. The long-term re- nal transduction. Proc Natl Acad Sci U S A. 2007; etic stem cell maintenance. Nat Immunol. 2005;6:
populating subset of hematopoietic stem cells is 104:2349-2354. 314-322.
deterministic and isolatable by phenotype. Immu- 28. Takaki S, Sauer K, Iritani BM, et al. Control of B 47. Mancini SJ, Mantei N, Dumortier A, Suter U, Mac-
nity. 1994;1:661-673. cell production by the adaptor protein lnk: defini- Donald HR, Radtke F. Jagged1-dependent Notch
9. Wolf NS, Kone A, Priestley GV, Bartelmez SH. In tion of a conserved family of signal-modulating signaling is dispensable for hematopoietic stem
vivo and in vitro characterization of long-term re- proteins. Immunity. 2000;13:599-609. cell self-renewal and differentiation. Blood. 2005;
populating primitive hematopoietic cells isolated 29. Tong W, Zhang J, Lodish HF. Lnk inhibits erythro- 105:2340-2342.
by sequential Hoechst 33342-rhodamine 123 poiesis and Epo-dependent JAK2 activation and 48. Austin TW, Solar GP, Ziegler FC, Liem L, Mat-
FACS selection. Exp Hematol. 1993;21:614-622. downstream signaling pathways. Blood. 2005; thews W. A role for the Wnt gene family in hema-
10. Goodell MA, Brose K, Paradis G, Conner AS, 105:4604-4612. topoiesis: expansion of multilineage progenitor
Mulligan RC. Isolation and functional properties 30. Velazquez L, Cheng AM, Fleming HE, et al. Cyto- cells. Blood. 1997;89:3624-3635.
of murine hematopoietic stem cells that are repli- kine signaling and hematopoietic homeostasis 49. Willert K, Brown JD, Danenberg E, et al. Wnt pro-
cating in vivo. J Exp Med. 1996;183:1797-1806. are disrupted in Lnk-deficient mice. J Exp Med. teins are lipid-modified and can act as stem cell
11. Kiel MJ, Yilmaz OH, Iwashita T, Yilmaz OH, Ter- 2002;195:1599-1611. growth factors. Nature. 2003;423:448-452.

Downloaded from http://ashpublications.org/blood/article-pdf/111/2/492/1220086/zh800208000492.pdf by guest on 29 January 2024


horst C, Morrison SJ. SLAM family receptors dis- 31. Ema H, Sudo K, Seita J, et al. Quantification of 50. Van Den Berg DJ, Sharma AK, Bruno E, Hoffman
tinguish hematopoietic stem and progenitor cells self-renewal capacity in single hematopoietic R. Role of members of the Wnt gene family in hu-
and reveal endothelial niches for stem cells. Cell. stem cells from normal and Lnk-deficient mice. man hematopoiesis. Blood. 1998;92:3189-3202.
2005;121:1109-1121. Dev Cell. 2005;8:907-914. 51. Reya T, Duncan AW, Ailles L, et al. A role for Wnt
12. McKenzie JL, Takenaka K, Gan OI, Doedens M, 32. Ellisen LW, Bird J, West DC, et al. TAN-1, the hu- signalling in self-renewal of haematopoietic stem
Dick JE. Low rhodamine 123 retention identifies man homolog of the Drosophila notch gene, is cells. Nature. 2003;423:409-414.
long-term human hematopoietic stem cells within broken by chromosomal translocations in T lym- 52. Kirstetter P, Anderson K, Porse BT, Jacobsen SE,
the Lin-CD34⫹CD38- population. Blood. 2007; phoblastic neoplasms. Cell. 1991;66:649-661. Nerlov C. Activation of the canonical Wnt path-
109:543-545. 33. Radtke F, Wilson A, Mancini SJ, MacDonald HR. way leads to loss of hematopoietic stem cell re-
13. Dick JE, Bhatia M, Gan O, Kapp U, Wang JC. Notch regulation of lymphocyte development and population and multilineage differentiation block.
Assay of human stem cells by repopulation of function. Nat Immunol. 2004;5:247-253. Nat Immunol. 2006;7:1048-1056.
NOD/SCID mice. Stem Cells. 1997;15(Suppl 1): 34. Milner LA, Kopan R, Martin DI, Bernstein ID. A 53. Cobas M, Wilson A, Ernst B, et al. Beta-catenin is
199-203; discussion 204. human homologue of the Drosophila develop- dispensable for hematopoiesis and lymphopoi-
14. Till JE, McCulloch EA, Siminovitch L. A stochastic mental gene, Notch, is expressed in CD34⫹ he- esis. J Exp Med. 2004;199:221-229.
model of stem cell proliferation, based on the matopoietic precursors. Blood. 1994;83:2057- 54. Trowbridge JJ, Xenocostas A, Moon RT, Bhatia
growth of spleen colony-forming cells. Proc Natl 2062. M. Glycogen synthase kinase-3 is an in vivo regu-
Acad Sci U S A. 1964;51:29-36. 35. Karanu FN, Murdoch B, Gallacher L, et al. The lator of hematopoietic stem cell repopulation. Nat
15. Metcalf D. Hematopoietic regulators: redundancy notch ligand jagged-1 represents a novel growth Med. 2006;12:89-98.
or subtlety? Blood. 1993;82:3515-3523. factor of human hematopoietic stem cells. J Exp
55. Sitnicka E, Ruscetti FW, Priestley GV, Wolf NS,
16. Enver T, Heyworth CM, Dexter TM. Do stem cells Med. 2000;192:1365-1372.
Bartelmez SH. Transforming growth factor beta 1
play dice? Blood. 1998;92:348-351; discussion 36. Karanu FN, Murdoch B, Miyabayashi T, et al. Hu- directly and reversibly inhibits the initial cell divi-
352. man homologues of Delta-1 and Delta-4 function sions of long-term repopulating hematopoietic
17. Miller CL, Eaves CJ. Expansion in vitro of adult as mitogenic regulators of primitive human hema- stem cells. Blood. 1996;88:82-88.
murine hematopoietic stem cells with transplant- topoietic cells. Blood. 2001;97:1960-1967.
56. Batard P, Monier MN, Fortunel N, et al. TGF-
able lympho-myeloid reconstituting ability. Proc 37. Varnum-Finney B, Brashem-Stein C, Bernstein (beta)1 maintains hematopoietic immaturity by a
Natl Acad Sci U S A. 1997;94:13648-13653. ID. Combined effects of Notch signaling and cyto- reversible negative control of cell cycle and in-
kines induce a multiple log increase in precursors duces CD34 antigen up-modulation. J Cell Sci.
18. Christensen JL, Weissman IL. Flk-2 is a marker in
with lymphoid and myeloid reconstituting ability. 2000;113(Pt 3):383-390.
hematopoietic stem cell differentiation: a simple
Blood. 2003;101:1784-1789.
method to isolate long-term stem cells. Proc Natl 57. Garbe A, Spyridonidis A, Mobest D, Schmoor C,
Acad Sci U S A. 2001;98:14541-14546. 38. Vas V, Szilagyi L, Paloczi K, Uher F. Soluble Mertelsmann R, Henschler R. Transforming
Jagged-1 is able to inhibit the function of its multi- growth factor-beta 1 delays formation of granulo-
19. Audet J, Miller CL, Rose-John S, Piret JM, Eaves
valent form to induce hematopoietic stem cell cyte-macrophage colony-forming cells, but
CJ. Distinct role of gp130 activation in promoting
self-renewal in a surrogate in vitro assay. J Leu- spares more primitive progenitors during ex vivo
self-renewal divisions by mitogenically stimulated
koc Biol. 2004;75:714-720. expansion of CD34⫹ haemopoietic progenitor
murine hematopoietic stem cells. Proc Natl Acad
Sci U S A. 2001;98:1757-1762. 39. Ohishi K, Varnum-Finney B, Bernstein ID. Delta-1 cells. Br J Haematol. 1997;99:951-958.
enhances marrow and thymus repopulating abil- 58. Hatzfeld J, Li ML, Brown EL, et al. Release of
20. Nandurkar HH, Robb L, Tarlinton D, Barnett L,
ity of human CD34(⫹)CD38(-) cord blood cells. early human hematopoietic progenitors from qui-
Kontgen F, Begley CG. Adult mice with targeted
J Clin Invest. 2002;110:1165-1174. escence by antisense transforming growth factor
mutation of the interleukin-11 receptor (IL11Ra)
display normal hematopoiesis. Blood. 1997;90: 40. Delaney C, Varnum-Finney B, Aoyama K, beta 1 or Rb oligonucleotides. J Exp Med. 1991;
2148-2159. Brashem-Stein C, Bernstein ID. Dose-dependent 174:925-929.
effects of the Notch ligand Delta1 on ex vivo dif- 59. Fortunel N, Batard P, Hatzfeld A, et al. High prolif-
21. Kimura S, Roberts AW, Metcalf D, Alexander WS.
ferentiation and in vivo marrow repopulating abil- erative potential-quiescent cells: a working model
Hematopoietic stem cell deficiencies in mice lack-
ity of cord blood cells. Blood. 2005;106:2693- to study primitive quiescent hematopoietic cells.
ing c-Mpl, the receptor for thrombopoietin. Proc
2699. J Cell Sci. 1998;111(Pt 13):1867-1875.
Natl Acad Sci U S A. 1998;95:1195-1200.
41. Suzuki A, Raya A, Kawakami Y, et al. Nanog 60. Soma T, Yu JM, Dunbar CE. Maintenance of mu-
22. Solar GP, Kerr WG, Zeigler FC, et al. Role of c-
binds to Smad1 and blocks bone morphogenetic rine long-term repopulating stem cells in ex vivo
mpl in early hematopoiesis. Blood. 1998;92:4-10.
protein-induced differentiation of embryonic stem culture is affected by modulation of transforming
23. Ikuta K, Weissman IL. Evidence that hematopoi- cells. Proc Natl Acad Sci U S A. 2006;103:10294- growth factor-beta but not macrophage inflamma-
etic stem cells express mouse c-kit but do not 10299. tory protein-1 alpha activities. Blood. 1996;87:
depend on steel factor for their generation. Proc 42. Kunisato A, Chiba S, Nakagami-Yamaguchi E, et 4561-4567.
Natl Acad Sci U S A. 1992;89:1502-1506. al. HES-1 preserves purified hematopoietic stem 61. Jacobsen SE, Ruscetti FW, Dubois CM, Lee J,
24. Buza-Vidas N, Antonchuk J, Qian H, et al. Cyto- cells ex vivo and accumulates side population Boone TC, Keller JR. Transforming growth factor-
kines regulate postnatal hematopoietic stem cell cells in vivo. Blood. 2003;101:1777-1783. beta trans-modulates the expression of colony
expansion: opposing roles of thrombopoietin and 43. Stier S, Cheng T, Dombkowski D, Carlesso N, stimulating factor receptors on murine hemato-
LNK. Genes Dev. 2006;20:2018-2023. Scadden DT. Notch1 activation increases hema- poietic progenitor cell lines. Blood. 1991;77:1706-
25. Borge OJ, Ramsfjell V, Veiby OP, Murphy MJ Jr, topoietic stem cell self-renewal in vivo and favors 1716.
Lok S, Jacobsen SE. Thrombopoietin, but not lymphoid over myeloid lineage outcome. Blood. 62. Dubois CM, Ruscetti FW, Palaszynski EW, Falk
erythropoietin promotes viability and inhibits apo- 2002;99:2369-2378. LA, Oppenheim JJ, Keller JR. Transforming
ptosis of multipotent murine hematopoietic pro- 44. Varnum-Finney B, Xu L, Brashem-Stein C, et al. growth factor beta is a potent inhibitor of interleu-
genitor cells in vitro. Blood. 1996;88:2859-2870. Pluripotent, cytokine-dependent, hematopoietic kin 1 (IL-1) receptor expression: proposed
26. Pestina TI, Cleveland JL, Yang C, Zambetti GP, stem cells are immortalized by constitutive mechanism of inhibition of IL-1 action. J Exp Med.
Jackson CW. Mpl ligand prevents lethal myelo- Notch1 signaling. Nat Med. 2000;6:1278-1281. 1990;172:737-744.
502 BLANK et al BLOOD, 15 JANUARY 2008 䡠 VOLUME 111, NUMBER 2

63. Dubois CM, Ruscetti FW, Stankova J, Keller JR. tiation of hematopoietic mesoderm in Xenopus ity of Hoxa9 by repressing its DNA-binding ability.
Transforming growth factor-beta regulates c-kit laevis. Blood. 1996;88:1965-1972. EMBO J. 2006;25:1469-1480.
message stability and cell-surface protein expres- 81. Schmid B, Furthauer M, Connors SA, et al. 101. Zhang CC, Kaba M, Ge G, et al. Angiopoietin-like
sion in hematopoietic progenitors. Blood. 1994; Equivalent genetic roles for bmp7/snailhouse and proteins stimulate ex vivo expansion of hemato-
83:3138-3145. bmp2b/swirl in dorsoventral pattern formation. poietic stem cells. Nat Med. 2006;12:240-245.
64. Sansilvestri P, Cardoso AA, Batard P, et al. Early Development. 2000;127:957-967. 102. Lord BI, Testa NG, Hendry JH. The relative spa-
CD34high cells can be separated into KIThigh 82. Schmerer M, Evans T. Primitive erythropoiesis is tial distributions of CFUs and CFUc in the normal
cells in which transforming growth factor-beta regulated by Smad-dependent signaling in post- mouse femur. Blood.1975;46:65-72.
(TGF-beta) downmodulates c-kit and KITlow cells gastrulation mesoderm. Blood. 2003;102:3196-
in which anti-TGF-beta upmodulates c-kit. Blood. 103. Gong JK. Endosteal marrow: a rich source of he-
3205. matopoietic stem cells. Science. 1978;199:1443-
1995;86:1729-1735.
83. Winnier G, Blessing M, Labosky PA, Hogan BL. 1445.
65. Ducos K, Panterne B, Fortunel N, Hatzfeld A,
Bone morphogenetic protein-4 is required for me- 104. Schofield R. The relationship between the spleen
Monier MN, Hatzfeld J. p21(cip1) mRNA is con-
soderm formation and patterning in the mouse. colony-forming cell and the haemopoietic stem
trolled by endogenous transforming growth fac-
Genes Dev. 1995;9:2105-2116. cell. Blood Cells. 1978;4:7-25.
tor-beta1 in quiescent human hematopoietic
stem/progenitor cells. J Cell Physiol. 2000;184: 84. Bhatia M, Bonnet D, Wu D, et al. Bone morpho- 105. Wilson A, Trumpp A. Bone-marrow haematopoi-
80-85. genetic proteins regulate the developmental pro- etic-stem-cell niches. Nat Rev Immunol. 2006;6:
gram of human hematopoietic stem cells. J Exp 93-106.
66. Cheng T, Shen H, Rodrigues N, Stier S, Scadden
Med. 1999;189:1139-1148.
DT. Transforming growth factor beta 1 mediates 106. Zhang J, Niu C, Ye L, et al. Identification of the

Downloaded from http://ashpublications.org/blood/article-pdf/111/2/492/1220086/zh800208000492.pdf by guest on 29 January 2024


cell-cycle arrest of primitive hematopoietic cells 85. Bhardwaj G, Murdoch B, Wu D, et al. Sonic haematopoietic stem cell niche and control of the
independent of p21(Cip1/Waf1) or p27(Kip1). hedgehog induces the proliferation of primitive niche size. Nature. 2003;425:836-841.
Blood. 2001;98:3643-3649. human hematopoietic cells via BMP regulation.
Nat Immunol. 2001;2:172-180. 107. Visnjic D, Kalajzic Z, Rowe DW, Katavic V,
67. Dao MA, Taylor N, Nolta JA. Reduction in levels Lorenzo J, Aguila HL. Hematopoiesis is severely
of the cyclin-dependent kinase inhibitor 86. Utsugisawa T, Moody JL, Aspling M, Nilsson E, altered in mice with an induced osteoblast defi-
p27(kip-1) coupled with transforming growth fac- Carlsson L, Karlsson S. A road map towards de- ciency. Blood. 2004;103:3258-3264.
tor beta neutralization induces cell-cycle entry fining the role of Smad signaling in hematopoietic
stem cells. Stem Cells. 2006;24:1128-1136. 108. Arai F, Hirao A, Ohmura M, et al. Tie2/angiopoi-
and increases retroviral transduction of primitive
etin-1 signaling regulates hematopoietic stem cell
human hematopoietic cells. Proc Natl Acad Sci 87. Liu B, Sun Y, Jiang F, et al. Disruption of Smad5 quiescence in the bone marrow niche. Cell. 2004;
U S A. 1998;95:13006-13011. gene leads to enhanced proliferation of high-pro- 118:149-161.
68. Dao MA, Hwa J, Nolta JA. Molecular mechanism liferative potential precursors during embryonic
of transforming growth factor beta-mediated cell- hematopoiesis. Blood. 2003;101:124-133. 109. Stier S, Ko Y, Forkert R, et al. Osteopontin is a
cycle modulation in primary human CD34(⫹) pro- hematopoietic stem cell niche component that
88. Singbrant S, Moody JL, Blank U, et al. Smad5 is negatively regulates stem cell pool size. J Exp
genitors. Blood. 2002;99:499-506. dispensable for adult murine hematopoiesis. Med. 2005;201:1781-1791.
69. Scandura JM, Boccuni P, Massague J, Nimer SD. Blood. 2006;108:3707-3712.
Transforming growth factor beta-induced cell 110. Nilsson SK, Johnston HM, Whitty GA, et al. Os-
89. Blank U, Karlsson G, Moody JL, et al. Smad7 teopontin, a key component of the hematopoietic
cycle arrest of human hematopoietic cells re- promotes self-renewal of hematopoietic stem
quires p57KIP2 up-regulation. Proc Natl Acad Sci stem cell niche and regulator of primitive hemato-
cells in vivo. Blood. 2006;108:4246-4254. poietic progenitor cells. Blood. 2005;106:1232-
U S A. 2004;101:15231-15236.
90. Chadwick K, Shojaei F, Gallacher L, Bhatia M. 1239.
70. Larsson J, Karlsson S. The role of Smad signal-
Smad7 alters cell fate decisions of human hema- 111. Adams GB, Chabner KT, Alley IR, et al. Stem cell
ing in hematopoiesis. Oncogene. 2005;24:5676-
topoietic repopulating cells. Blood. 2005;105: engraftment at the endosteal niche is specified by
5692.
1905-1915. the calcium-sensing receptor. Nature. 2006;439:
71. Ruscetti FW, Akel S, Bartelmez SH. Autocrine
91. Moustakas A, Heldin CH. Non-Smad TGF-beta 599-603.
transforming growth factor-beta regulation of he-
signals. J Cell Sci. 2005;118:3573-3584. 112. Walkley CR, Olsen GH, Dworkin S, et al. A micro-
matopoiesis: many outcomes that depend on the
context. Oncogene. 2005;24:5751-5763. 92. Karlsson G, Blank U, Moody JL, et al. Smad4 is environment-induced myeloproliferative syn-
critical for self-renewal of hematopoietic stem drome caused by retinoic acid receptor gamma
72. Fortunel NO, Hatzfeld A, Hatzfeld JA. Transform-
cells. J Exp Med. 2007;204:467-474. deficiency. Cell. 2007;129:1097-1110.
ing growth factor-beta: pleiotropic role in the regu-
lation of hematopoiesis. Blood. 2000;96:2022- 93. Nishita M, Hashimoto MK, Ogata S, et al. Interac- 113. Walkley CR, Shea JM, Sims NA, Purton LE, Orkin
2036. tion between Wnt and TGF-beta signalling path- SH. Rb regulates interactions between hemato-
ways during formation of Spemann’s organizer. poietic stem cells and their bone marrow microen-
73. Yaswen L, Kulkarni AB, Fredrickson T, et al. Auto-
Nature. 2000;403:781-785. vironment. Cell. 2007;129:1081-1095.
immune manifestations in the transforming
growth factor-beta 1 knockout mouse. Blood. 94. Labbé E, Letamendia A, Attisano L. Association 114. Parmar K, Mauch P, Vergilio JA, Sackstein R,
1996;87:1439-1445. of Smads with lymphoid enhancer binding factor Down JD. Distribution of hematopoietic stem cells
74. Letterio JJ, Roberts AB. Regulation of immune 1/T cell-specific factor mediates cooperative sig- in the bone marrow according to regional hyp-
responses by TGF-beta. Annu Rev Immunol. naling by the transforming growth factor-beta and oxia. Proc Natl Acad Sci U S A. 2007;104:5431-
1998;16:137-161. wnt pathways. Proc Natl Acad Sci U S A. 2000; 5436.
97:8358-8363. 115. Ito K, Hirao A, Arai F, et al. Regulation of oxidative
75. Levéen P, Larsson J, Ehinger M, et al. Induced
disruption of the transforming growth factor beta 95. Itoh F, Itoh S, Goumans MJ, et al. Synergy and stress by ATM is required for self-renewal of
type II receptor gene in mice causes a lethal in- antagonism between Notch and BMP receptor haematopoietic stem cells. Nature. 2004;431:
flammatory disorder that is transplantable. Blood. signaling pathways in endothelial cells. EMBO J. 997-1002.
2002;100:560-568. 2004;23:541-551. 116. Ito K, Hirao A, Arai F, et al. Reactive oxygen spe-
76. Letterio JJ, Geiser AG, Kulkarni AB, et al. Autoim- 96. He W, Dorn DC, Erdjument-Bromage H, Tempst cies act through p38 MAPK to limit the lifespan of
munity associated with TGF-beta1-deficiency in P, Moore MA, Massague J. Hematopoiesis con- hematopoietic stem cells. Nat Med. 2006;12:446-
mice is dependent on MHC class II antigen ex- trolled by distinct TIF1gamma and Smad4 451.
pression. J Clin Invest. 1996;98:2109-2119. branches of the TGFbeta pathway. Cell. 2006; 117. Tothova Z, Kollipara R, Huntly BJ, et al. FoxOs
125:929-941. are critical mediators of hematopoietic stem cell
77. Larsson J, Blank U, Helgadottir H, et al. TGF-beta
signaling-deficient hematopoietic stem cells have 97. Ransom DG, Bahary N, Niss K, et al. The ze- resistance to physiologic oxidative stress. Cell.
normal self-renewal and regenerative ability in brafish moonshine gene encodes transcriptional 2007;128:325-339.
vivo despite increased proliferative capacity in intermediary factor 1gamma, an essential regula- 118. Rebel VI, Miller CL, Eaves CJ, Lansdorp PM. The
vitro. Blood. 2003;102:3129-3135. tor of hematopoiesis. PLoS Biol. 2004;2:E237. repopulation potential of fetal liver hematopoietic
78. Larsson J, Blank U, Klintman J, Magnusson M, 98. Dupont S, Zacchigna L, Cordenonsi M, et al. stem cells in mice exceeds that of their liver adult
Karlsson S. Quiescence of hematopoietic stem Germ-layer specification and control of cell bone marrow counterparts. Blood. 1996;87:3500-
cells and maintenance of the stem cell pool is not growth by Ectodermin, a Smad4 ubiquitin ligase. 3507.
dependent on TGF-beta signaling in vivo. Exp Cell. 2005;121:87-99. 119. Bowie MB, McKnight KD, Kent DG, McCaffrey L,
Hematol. 2005;33:592-596. 99. Kroon E, Krosl J, Thorsteinsdottir U, Baban S, Hoodless PA, Eaves CJ. Hematopoietic stem
79. Huber TL, Zhou Y, Mead PE, Zon LI. Cooperative Buchberg AM, Sauvageau G. Hoxa9 transforms cells proliferate until after birth and show a re-
effects of growth factors involved in the induction primary bone marrow cells through specific col- versible phase-specific engraftment defect. J Clin
of hematopoietic mesoderm. Blood. 1998;92: laboration with Meis1a but not Pbx1b. EMBO J. Invest. 2006;116:2808-2816.
4128-4137. 1998;17:3714-3725. 120. Bowie MB, Kent DG, Dykstra B, et al. Identifica-
80. Maeno M, Mead PE, Kelley C, et al. The role of 100. Wang N, Kim HG, Cotta CV, et al. TGFbeta/BMP tion of a new intrinsically timed developmental
BMP-4 and GATA-2 in the induction and differen- inhibits the bone marrow transformation capabil- checkpoint that reprograms key hematopoietic
BLOOD, 15 JANUARY 2008 䡠 VOLUME 111, NUMBER 2 SIGNALING PATHWAYS AND STEM CELL FATE 503

stem cell properties. Proc Natl Acad Sci U S A. expansion of human hematopoietic stem cells by taglandin E2 regulates vertebrate haematopoietic
2007;104:5878-5882. direct delivery of the HOXB4 homeoprotein. Nat stem cell homeostasis. Nature. 2007;447:1007-
121. Bowie MB, Kent DG, Copley MR, Eaves CJ. Steel Med. 2003;9:1423-1427. 1011.
factor responsiveness regulates the high self- 130. Krosl J, Austin P, Beslu N, Kroon E, Humphries 138. Chiba S. Normal and deregulated notch signaling
renewal phenotype of fetal hematopoietic stem RK, Sauvageau G. In vitro expansion of hemato- in regulation of hematopoietic stem cells and de-
cells. Blood. 2007;109:5043-5048. poietic stem cells by recombinant TAT-HOXB4 velopment of leukemia [in Japanese]. Rinsho Ket-
122. Gluckman E, Broxmeyer HA, Auerbach AD, et al. protein. Nat Med. 2003;9:1428-1432. sueki. 2006;47:371-378.
Hematopoietic reconstitution in a patient with 131. de Haan G, Weersing E, Dontje B, et al. In vitro 139. Barker N, Clevers H. Mining the Wnt pathway for
Fanconi’s anemia by means of umbilical cord generation of long-term repopulating hematopoi- cancer therapeutics. Nat Rev Drug Discov. 2006;
blood from an HLA-identical sibling. N Engl J etic stem cells by fibroblast growth factor-1. Dev 5:997-1014.
Med. 1989;321:1174-1178. Cell. 2003;4:241-251. 140. Clevers H. Wnt/beta-catenin signaling in develop-
123. Attar EC, Scadden DT. Regulation of hematopoi- 132. Yeoh JS, van Os R, Weersing E, et al. Fibroblast ment and disease. Cell. 2006;127:469-480.
etic stem cell growth. Leukemia. 2004;18:1760- growth factor-1 and -2 preserve long-term re- 141. Massagué J. TGF-beta signal transduction. Annu
1768. populating ability of hematopoietic stem cells in Rev Biochem. 1998;67:753-791.
124. Sorrentino BP. Clinical strategies for expansion of serum-free cultures. Stem Cells. 2006;24:1564- 142. Heldin CH, Miyazono K, ten Dijke P. TGF-beta
haematopoietic stem cells. Nat Rev Immunol. 1572. signalling from cell membrane to nucleus through
2004;4:878-888. 133. Zhang CC, Lodish HF. Murine hematopoietic SMAD proteins. Nature. 1997;390:465-471.
125. Baum C, Dullmann J, Li Z, et al. Side effects of stem cells change their surface phenotype during 143. Nakao A, Afrakhte M, Moren A, et al. Identification
retroviral gene transfer into hematopoietic stem ex vivo expansion. Blood. 2005;105:4314-4320. of Smad7, a TGFbeta-inducible antagonist of

Downloaded from http://ashpublications.org/blood/article-pdf/111/2/492/1220086/zh800208000492.pdf by guest on 29 January 2024


cells. Blood. 2003;101:2099-2114. 134. Gupta R, Hong D, Iborra F, Sarno S, Enver T. TGF-beta signalling. Nature. 1997;389:631-635.
126. Hacein-Bey-Abina S, Von Kalle C, Schmidt M, et NOV (CCN3) functions as a regulator of human 144. Hayashi H, Abdollah S, Qiu Y, et al. The MAD-
al. LMO2-associated clonal T cell proliferation in hematopoietic stem or progenitor cells. Science. related protein Smad7 associates with the TGF-
two patients after gene therapy for SCID-X1. Sci- 2007;316:590-593. beta receptor and functions as an antagonist of
ence. 2003;302:415-419. 135. Bug G, Gul H, Schwarz K, et al. Valproic acid TGFbeta signaling. Cell. 1997;89:1165-1173.
127. Woods NB, Muessig A, Schmidt M, et al. Lentivi- stimulates proliferation and self-renewal of hema- 145. Souchelnytskyi S, Nakayama T, Nakao A, et al.
ral vector transduction of NOD/SCID repopulating topoietic stem cells. Cancer Res. 2005;65:2537- Physical and functional interaction of murine and
cells results in multiple vector integrations per 2541. Xenopus Smad7 with bone morphogenetic pro-
transduced cell: risk of insertional mutagenesis. 136. De Felice L, Tatarelli C, Mascolo MG, et al. His- tein receptors and transforming growth factor-
Blood. 2003;101:1284-1289. tone deacetylase inhibitor valproic acid enhances beta receptors. J Biol Chem. 1998;273:25364-
128. Antonchuk J, Sauvageau G, Humphries RK. the cytokine-induced expansion of human hema- 25370.
HOXB4-induced expansion of adult hematopoi- topoietic stem cells. Cancer Res. 2005;65:1505- 146. Miyamoto K, Araki KY, Naka K, et al. Foxo3a is
etic stem cells ex vivo. Cell. 2002;109:39-45. 1513. essential for maintenance of the hematopoietic
129. Amsellem S, Pflumio F, Bardinet D, et al. Ex vivo 137. North TE, Goessling W, Walkley CR, et al. Pros- stem cell pool. Cell Stem. 2007;1:101-112.

You might also like