Hematopoietic stem cells (HSCs) are historically the most thoroughly characterized type of adult stem cell, and the hematopoietic system has served as a principal model structure of stem-cell biology for several decades. However, paradoxically, although HSCs can be defined by function and even purified to near-homogeneity, the intricate molecular machinery and the signaling mechanisms regulating fate events, such as self-renewal and differentiation, have remained elusive. Recently, several developmentally conserved signaling pathways have emerged as important control devices of HSC fate, including Notch, Wingless-type (Wnt), Sonic hedgehog (Shh), and Smad pathways. HSCs reside in a complex environment in the bone marrow, providing a niche that optimally balances signals that control self-renewal and differentiation. These signaling circuits provide a valuable structure for our understanding of how HSC regulation occurs, concomitantly with providing information of how the bone marrow microenvironment couples and integrates extrinsic with intrinsic HSC fate determinants. It is the focus of this review to highlight some of the most recent developments concerning signaling pathways governing HSC fate.

In tissues with a high cellular turnover, stem-cell populations are essential for lifelong maintenance of organ function. At present, somatic stem cells have been identified in several self-renewing organs, including intestinal and epidermal epithelia as well as the blood cell system. Residing in the bone marrow (BM) of adults, hematopoietic stem cells (HSCs) are ultimately responsible for the continuous daily production of all mature blood cell lineages.1  HSCs are functionally defined at the single-cell level by their dual capacity for self-renewal and multipotential differentiation. Self-renewal pertains to the process whereby at least one daughter cell of a dividing HSC retains stem-cell fate and may be either symmetric or asymmetric in its nature (Figure 1). A symmetric self-renewing division refers to the process whereby both daughter cells retain stem cell properties. Theoretically, this type of cell division expands the stem-cell pool and is therefore thought to be important after transplantation or after hematopoietic injury. In an asymmetric self-renewing division, the 2 daughter cells adopt different fates, resulting in one cell maintaining stem-cell properties. On a population basis, this allows for steady-state main-tenance of the HSC compartment. Self-renewal is critical for sustaining the HSC compartment and thus is a prerequisite for lifelong hematopoiesis.

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.

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.

Close modal

The hematopoietic system is hierarchically organized, with a rare population of HSCs giving rise to progeny that progressively lose self-renewal potential and successively become more and more restricted in their differentiation capacity, finally generating functionally mature cells (Figure 2). Phenotypic characterization of HSCs has allowed for their prospective isolation, and several groups have reported the successful regeneration of all blood cell lineages after transplantation of a single HSC into lethally irradiated mice.2,3  All functional HSCs are associated with lack of expression of a range of cell surface markers normally found on differentiating or mature blood cells while displaying high levels of Sca1 and c-kit.4,5  This population of cells is commonly referred to as the LSK compartment (lineage/Sca1+/c-kit+) and 100 LSK cells have been shown to be sufficient for long-term multilineage repopulation of lethally irradiated hosts.5  Based on their ability to self-renew, HSCs can be divided into long-term and short-term reconstituting HSCs (LT-HSCs and ST-HSCs, respectively). LT-HSCs have extensive self-renewal capacity and can sustain lifelong hematopoiesis, whereas ST-HSCs are restricted in self-renewal potential, sustaining hematopoiesis only for a limited time in vivo.6,8  With respect to self-renewal, the LSK compartment is heterogeneous, containing a mixture of LT-HSCs, ST-HSCs, and multipotent progenitor cells (MPPs). Further fractionation based on differential expression of CD34 and Flt3 has been shown to correlate with distinct functional properties, with LT-HSCs being negative 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 side population defines a small and distinct subset of cells with LT-HSC characteristics.10  More recently, Kiel et al identified the SLAM family of cell surface receptors that were shown to be differentially expressed among stem and progenitor populations.11 

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.

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.

Close modal

Efficient isolation of human HSCs has proven more difficult, primarily as a consequence of lack of appropriate in vivo assays. As of today, the rhodaminelow fraction of the LinCD34+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 human HSCs can be functionally analyzed in nonobese diabetic/severe combined immune deficient (NOD/SCID) mouse strains.13  SCID repopulating cells (SRCs) represent the most primitive hematopoietic population assessable, but NOD/SCID assays are hampered by the generally low engraftment of human cells and the difficulty in keeping NOD/SCID mice for long-term analyses. This predicament can be overcome by serial NOD/SCID transplantations, allowing for analysis of even more primitive populations. However, such assays keep HSCs under constant stress, which is why caution should be taken when interpreting obtained data.

To meet the need of a variety of situations, ranging from normal homeostasis to acute blood loss or infection, hematopoiesis must be rapidly yet meticulously controlled. Whether regarded as stochastic mainly regulating probabilities or deterministic providing more precise instructions, both intrinsic and extrinsic signals undoubtedly participate in the regulation of HSCs.1,14,16  Through reductionistic strategies, a wide variety of factors critical for HSC regulation have been identified, including cytokines, growth factors, transcription factors, chromatin modifiers, and cell-cycle regulators. Delineating the signaling pathways that integrate these diverse components remains an enduring challenge of foremost importance, which would have tremendous impact on regenerative medicine. During the course of development, a handful of signaling pathways shape the structures of the embryo and determine cell fate and identity. In the adult organism, these signaling pathways are reiterated, and it is becoming increasingly clear that most somatic stem-cell compartments are under the influence of developmentally conserved signals.

Numerous attempts have been made to use classic hematopoietic cytokines for the purpose of expanding HSCs in vitro. Many of the interleukins, particularly, interleukin (IL)-3, IL-6, and IL-11, as well as Flt-3 ligand, thrombopoietin (TPO) and stem-cell factor (SCF) have been investigated. In most cases, efforts to expand HSCs have failed because of differentiation of HSCs and subsequent loss of reconstitution capacity. In one study, a modest expansion of repopulating murine HSCs was observed using a 10-day culture protocol in the presence of IL-11, Flt-3 ligand, and SCF.17  However, the same conditions failed to expand HSCs derived from the fetal liver and later studies have found that the Flt-3 receptor is not expressed on LT-HSCs.7,18  Furthermore, the gp130 protein, which is a part of the receptor for IL-6 and IL-11 was suggested to play a role in the self-renewal of HSCs.19  However, elimination of the IL-11 receptor does not affect 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 c-kit, respectively, are both expressed on repopulating HSCs,6,7,21,,24  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 reported to support viability25  and suppressed apoptosis of HSCs.26  Thus, the main function of TPO may be to counteract apoptosis of HSCs rather than promote their expansion.24 

A further role for TPO in regulating HSC self-renewal was recently suggested through the association with Lnk, an intracellular adaptor molecule functioning as a broad inhibitor of several cytokine signaling pathways, including TPO, SCF, erythropoietin, IL-3, and IL-7.24,27,,30  Mice deficient in Lnk have been reported to exhibit increased HSC numbers and their repopulation function was elevated, a phenotype attributable to augmented proliferation and self-renewal.24,27,31  Interestingly, the increased self-renewal capacity of Lnk−/− HSCs was dependent on TPO, and Lnk-negative HSCs were shown to be hypersensitive to stimulation by TPO.24,27  Thus, TPO and Lnk serve opposing roles in the regulation of HSC self-renewal and Lnk acts as a powerful inhibitor of TPO signaling in HSCs. To conclude, of the classic hematopoietic cytokines, the most important positive regulators of HSCs are TPO and SCF.

In humans, Notch was discovered 15 years ago as an oncogene that, after chromosomal translocation, gave rise to T-cell leukemia.32  Since then, several loss- and gain-of-function studies have demonstrated a critical role for Notch in lineage specification of lymphopoiesis, where active Notch signaling (Figure 3A) drives differentiation toward T cells.33  It was soon suggested that Notch could have a role in HSC biology as primitive hematopoietic cells as well as cells in the putative HSC microenvironment expressed both Notch and Notch-ligand mRNA.34,35  Indeed, through various approaches, several groups have shown that members of the Notch ligand families, Delta and Jagged, can expand both murine and human hematopoietic progenitors in vitro as measured by reconstitution assays.35,,,,,41  Importantly, enforced activation of the Notch signaling pathway or the downstream target Hes-1, have been demonstrated to increase the self-renewal capacity of long-term in vivo repopulating HSCs42,43  or even to immortalize primitive hematopoietic progenitor cells.44  When Calvi et al established osteoblasts as putative members of the HSC niche, they demonstrated how constitutively active parathyroid hormone receptor expanded these cells and stimulated their expression of Jagged-1.45  The activated osteoblasts were able to increase self-renewal of primitive hematopoietic cells as measured by LTC-ICs and 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 normalized with a γ-secretase inhibitor.45  Supporting these data were the observation by Duncan et al that a Notch reporter was active in c-kit+ hematopoietic cells in the trabecular bone and isolated LSK and side population cells.46 

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 γ-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 β-catenin is trapped by adenomatous polyposis coli (APC) and Axin in a destruction complex, where it is phosphorylated by casein-kinase 1α (CK1α) and glycogen synthase kinase (GSK3β). Phosphorylation ultimately leads to ubiquitination and degradation of β-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 β-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 

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 γ-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 β-catenin is trapped by adenomatous polyposis coli (APC) and Axin in a destruction complex, where it is phosphorylated by casein-kinase 1α (CK1α) and glycogen synthase kinase (GSK3β). Phosphorylation ultimately leads to ubiquitination and degradation of β-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 β-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 

Close modal

Together these findings indicate not only that osteoblasts are part of the so-called HSC niche but also that Jagged/Notch signaling activated by these cells might be important in the extracellular regulation of HSC self-renewal.45  However, HSCs deficient in Notch-1 could reconstitute Jagged-1 deficient hosts in a normal fashion, suggesting that Notch signaling through mechanisms including these molecules is dispensable for in vivo HSC function.47  It is probable that the lack of phenotype observed in this setting is the result of compensatory mechanisms mediated by other Notch receptors and ligands, which is why approaches to broadly eliminate Notch signaling would be informative. Regardless of what the case may be, manipulation of the Notch signaling pathway holds great promise for ex vivo expansion of HSCs in future clinical protocols.

A role for Wnt signaling in the regulation of murine HSCs was suggested a decade ago when expression of different Wnts was discovered at sites of fetal hematopoiesis and Wnt5a was shown to stimulate proliferation and self-renewal of fetal HSCs/progenitors in vitro.48  Adult HSCs also appear to respond to Wnt treatment because exposure to purified Wnt3a was recently shown to increase self-renewal of murine HSCs in vitro, as determined by in vivo reconstituting assays.49  Moreover, Van den Berg et al demonstrated that the positive effects on HSCs were not exclusive to the murine setting because human LinCD34+ cells exposed to WNTs also expanded in vitro as measured by immunophenotype and 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 proteins have also been reported to be expressed in both human fetal BM stroma and adult BM, where WNT5A has been shown to be expressed in the primitive LinCD34+ population.50  This would suggest that Wnts are physiologically important and act on HSCs through both paracrine and autocrine mechanisms. In support of the latter statement was the finding that exposure of HSCs in vitro to a soluble Wnt binding antagonist reduced their proliferation capacity.51  However, the intracellular mechanisms through which Wnts exercise their effect on HSCs are somewhat inconclusive. Canonical Wnt signaling (Figure 3B) occurs through mechanisms involving the stabilization and nuclear translocation of β-catenin, followed by transcriptional regulation of target genes by a β-catenin/T-cell factor complex. Reya et al detected active canonical Wnt signaling, as measured by a T-cell factor reporter construct, in a large proportion of the HSC enriched LSK compartment in adult BM.51  Intriguingly, retroviral–mediated enforced expression of β-catenin resulted in a 102 to 103 fold increase in HSCs during long-term cultures of KTLS cells as defined by both phenotype and function.51  In accordance with the observed phenotype resulting from forced expression of β-catenin was an up-regulation in expression of the HSC self-renewal stimulating genes Notch1 and HoxB4.51  However, when β-catenin was conditionally expressed in vivo under the control of the ROSA26 locus, no increase in HSC self-renewal was observed.52  Adding to the contradiction, deletion of the β-catenin gene in HSCs failed to result in an in vivo phenotype.53 

The reasons for these discrepancies are unclear but may reflect a stronger role for Wnt signaling in in vitro expansion of HSCs as opposed to its function in vivo. The more complex and enduring in vivo setting might allow for redundant mechanisms to develop. Alternatively, the HSC expansion observed when β-catenin was retrovirally overexpressed might be affected by the use of Bcl-2 transgenic HSCs in that particular study.51  Thus, even though Wnts may possibly be useful for expanding HSCs in vitro, the unresolved mechanisms behind the effect and the question pertaining to the role of canonical Wnt signaling remains to be determined.

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 fraction of cells in the trabecular bone.46  Even though Notch- and Wnt signaling both are implied to stimulate self-renewal of HSCs, Duncan et al propose that they do so through different mechanisms.46  As Wnt signaling seems to induce proliferation and support viability of LSK cells, inhibition of the Notch pathway had no effect on these functions.46  Instead, Notch signaling was demonstrated to be imperative in maintaining HSCs in an undifferentiated state, even if Wnt3a was present in the culture.46  These findings suggest that Wnt and Notch signaling together could play a role in self-renewal of HSCs. Moreover, observations that Wnt3a regulate expression of established Notch target genes46  and that inhibition of the Wnt signaling component GSK-3 affect HSC fate options through mechanisms involving regulation of both Wnt and Notch target genes54  suggest that the 2 pathways belong to a network of regulatory circuits controlling the HSC pool.

The Smad signaling pathway embodies an evolutionary ancient signaling circuitry, which functions to transduce signals downstream of the transforming growth factor-β (TGF-β) family of ligands (Figure 4). Apart from TGF-β, activins and bone morphogenetic proteins (BMPs) also belong to this superfamily, which regulate a bewildering array of fundamental biologic processes during development and postnatally. Because of the highly redundant nature of the Smad pathway and early embryonic lethality of most knockout mouse models, the precise role of the Smad circuitry in hematopoiesis has remained nebulous. However, recently several reports have shed new light suggesting that this pathway plays a pivotal role in the regulation of HSC fate decisions.

Figure 4

Smad signaling pathway. Smad pathway: TGF-β 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 proteins are activated through phosphorylation by type I receptors.141,142  Three groups of Smads have been identified: receptor-activated Smads (R-Smads), common- partner Smads (Co-Smads), and inhibitory Smads (I-Smads). In general, TGF-β and activin signal via R-Smad2 and 3, whereas BMP signals are transduced 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 Co-Smads, the I-Smads, Smad6 and Smad7, function in a negative feedback loop to prevent activation of R-Smads.143,145  Divergence and convergence of the Smad signaling circuitry are depicted. Commonly used alternative names include: ALK2/Activin type I receptor, ALK3/BMP type IA receptor, ALK4/Activin type IB receptor, ALK5/TGF-β type I receptor, and ALK6/BMP type IB receptor. The shaded portion indicates the nucleus. P indicates phosphorylation.

Figure 4

Smad signaling pathway. Smad pathway: TGF-β 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 proteins are activated through phosphorylation by type I receptors.141,142  Three groups of Smads have been identified: receptor-activated Smads (R-Smads), common- partner Smads (Co-Smads), and inhibitory Smads (I-Smads). In general, TGF-β and activin signal via R-Smad2 and 3, whereas BMP signals are transduced 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 Co-Smads, the I-Smads, Smad6 and Smad7, function in a negative feedback loop to prevent activation of R-Smads.143,145  Divergence and convergence of the Smad signaling circuitry are depicted. Commonly used alternative names include: ALK2/Activin type I receptor, ALK3/BMP type IA receptor, ALK4/Activin type IB receptor, ALK5/TGF-β type I receptor, and ALK6/BMP type IB receptor. The shaded portion indicates the nucleus. P indicates phosphorylation.

Close modal

TGF-β, key negative modulator of HSCs in vitro

TGF-β is generally catalogued as one of the most potent inhibitors of HSC growth in vitro, and ample evidence from a variety of culture systems exist to support this fact.55,57  A hallmark feature of HSCs is their relative quiescence, and given the strong growth inhibitory properties of TGF-β, it has naturally been hypothesized to be a cardinal regulator of HSC quiescence in vivo. In keeping with this, neutralization of TGF-β in vitro was shown to release early hematopoietic progenitor cells from quiescence.58,60  Several molecular mechanisms have been proposed to account for TGF-β–mediated growth inhibition, including alterations in cytokine receptor expression and up-regulation of cyclin-dependent kinase inhibitors, such as p21 and p57.59,61,,,,,,,69 

TGF-β can affect most cell types throughout the hematopoietic hierarchy and depending on the context and differentiation stage of the target cell different biologic responses are ultimately elicited.70,72  In vivo, TGF-β plays a principal role as regulator of immune cell homeostasis and function, as unambiguously evidenced by the development of a lethal inflammatory disorder of both TGF-β1- ligand and receptor knockout mice.73,75  Furthermore, Tgf-β1 null mice exhibited enhanced myelopoiesis, suggesting that TGF-β acts as a negative regulator of myelopoiesis in vivo.76  However, despite the pronounced role of TGF-β in vitro, mice deficient of the TGF-β type I receptor displayed normal HSC self-renewal and regenerative capacity in vivo, even under extreme hematopoietic stress.77,78  The apparent discrepancy between in vitro and in vivo findings may reflect mechanisms of redundancy between other type I receptors or alternatively other ligands such as activin, which signals through the same R-Smad pathway.

BMP promotes maintenance of HSCs in culture

BMPs have been implicated as key regulators of hematopoietic development in a variety of species, from zebrafish to mouse.79,,,83  In the context of adult hematopoiesis, BMP-4 was shown to promote maintenance of HSCs in culture, whereas lower concentrations of BMP-4 induced proliferation and differentiation of human hematopoietic progenitors.84  Furthermore, Shh induced proliferation of primitive human hematopoietic progenitors in vitro, apparently through a BMP-4–dependent mechanism.85  However, although BMP-4 has been shown to maintain human NOD/SCID repopulating cells in culture, it does not seem to cause an expansion, suggesting that Shh may act through additional mechanisms. In the murine system, BMP-4 does not appear to affect proliferation of purified HSCs in vitro, although it is currently unclear whether BMP-4 can extend the maintenance of murine HSCs in culture.86  Furthermore, BMPs function to regulate the HSC niche, thus indirectly controlling HSC numbers as discussed in “HSC niche.”

A role for Smad5, traditionally characterized as a mediator of BMP signals, in self-renewal was implicated through a series of in vitro studies. Interestingly, the number of colonies, derived from yolk sacs or ES cell differentiated embryoid bodies deficient of Smad5, were found to be increased.87 Smad5 deficient colonies also had an enhanced regenerative potential when secondary colony formation was assessed, indicative of increased self-renewal. However, conditional deletion of Smad5 in adult mice does not affect hematopoiesis, and HSCs lacking Smad5 have a normal differentiation and regenerative potential in vivo.88  It is possible that BMP–activated Smad signaling plays a more pronounced role early in hematopoietic development, as opposed to adult hematopoiesis. Alternatively, redundant mechanisms within the Smad pathway may explain the lack of phenotype in the adult setting.

Smad signaling: toward a deeper understanding

To block the entire Smad signaling pathway, thus sidestepping redundant mechanisms, the inhibitory Smad7 was overexpressed in murine HSCs using a retroviral gene transfer approach.89  Forced expression of Smad7 significantly increased the self-renewal capacity of HSCs in vivo, indicating that the Smad pathway negatively regulates self-renewal in vivo. In a similar approach using human SCID repopulating cells (SRCs), overexpression of Smad7 resulted in a shift from lymphoid dominant engraftment toward increased myeloid contribution.90  Thus, in the xenograft model system, forced expression of Smad7 modulates cell fate decisions of primitive multipotent human SRCs.

It is beyond doubt that the Smad signaling pathway lies at the very core of mediating signals from the TGF-β family of ligands. However, a considerably more diversified picture is now emerging, which indicates that the Smad circuitry is more varied than previously thought.91  Using a conditional knockout mouse model, disruption of the entire Smad pathway at the level of Smad4 was recently investigated. Intriguingly, Smad4 deficient HSCs displayed a significantly reduced repopulative capacity of primary and secondary recipients, indicating that Smad4 is critical for HSC self-renewal in vivo.92  Because overexpression of Smad7 versus deletion of Smad4 would be anticipated to yield similar hematopoietic phenotypes, it is conceivable that Smad4 functions as a positive regulator of self-renewal independently of its role as a central mediator of the canonical Smad pathway. The precise molecular mechanism for this is currently unknown, but it is possible that Smad4 participates in other signaling cascades such as Wnt or Notch.93,95  Moreover, He et al proposed an elegant model for how TGF-β mediates erythroid differentiation concomitantly with balancing growth inhibition in human hematopoietic stem/progenitor cells.96  According to this model, transcriptional intermediary factor1γ (TIF1γ) selectively binds Smad2 and Smad3 in competition with Smad4. In response to TGF-β, the TIF1γ/Smad2/3 complex was shown to stimulate erythroid differentiation of human hematopoietic progenitors, whereas Smad2/3 in association with Smad4 inhibited proliferation of the same cells. Thus, the relative abundance of Smad4 and TIF1γ is likely to be important for determining the effect of TGF-β stimulation. Interestingly, the zebrafish homolog of TIF1γ, encoded by moonshine, has been shown to be essential for blood formation with mutants displaying severe red cell aplasia, indicating that the function of TIF1γ may be preserved across species.97 

However, the mechanism through which TIF1γ cooperates with Smads is not entirely clear. Evidence from Xenopus has suggested that TIF1γ, known as Ectodermin, acts as a ubiquitin ligase for Smad4, thus functioning as a direct inhibitor of Smad4 downstream of TGF-β and BMP signaling.98  This dichotomy may reflect differences in cellular context or alternatively species or temporal differences. Another interesting observation is the association between Smad4 and Hox proteins. Homeobox (hox) genes encode transcription factors that function as paramount regulators of hematopoiesis and are frequently dysregulated in human leukemia, particularly acute myeloid leukemia. Enforced expression of HoxA9 immortalizes and blocks myeloid differentiation, eventually causing acute myeloid leukemia.99  Recently, Wang et al described a mechanism whereby TGF-β/BMP inhibited the bone marrow transformation capacity of HoxA9 and HoxA9-Nup98 fusion protein through a Smad4-dependent mechanism. Accordingly, Smad4 was shown to interact directly with HoxA9 and Nup98-HoxA9 fusion protein, thus precluding their DNA binding capacity and subsequent transcriptional activity.100  Whether Smad4 mutations facilitate leukemogenesis through increased Hox activity remains an unanswered question.

Building on the fact that the fetal liver (FL) is a powerful site of HSC expansion during development, Lodish et al embarked on an approach to identify “stromal” cells and their expression pattern of molecules responsible for stimulation of HSC self-renewal in the murine FL. Ter119+CD3+ cells were recognized as a population of cells derived from the FL that could support expansion of HSCs in vitro. Insulin-like growth factor 2 (IGF-2) was identified on the basis of being expressed in CD3+ FL cells and was shown to modestly expand HSCs in vitro. More interestingly, they found that several angiopoietin-like (Angptl) proteins were expressed in these support cells for HSC growth. When highly enriched HSCs were cultured under serum free conditions in the presence of TPO, SCF, IGF-2, and Angptl protein 2 or 3, a robust increase in repopulating HSCs was observed after 10 days of in vitro culture.101  More specifically, a 24- to 30-fold increase in HSC number was observed. Angptl proteins 5 and 7 were also reported to expand murine HSCs, whereas Angptl protein 4 failed to show a similar effect. The molecular mechanisms and the signaling events downstream of Angptl proteins remain unidentified and their receptors are currently unknown, which is why it is not clear how the Angptl proteins expand HSCs. However, Angptl protein 2 has been shown to bind to a highly enriched population of HSCs. Therefore, the expansion effect attributable to these proteins is likely to be a result of a direct action on HSCs. It will be exciting to see whether the Angptl proteins can also be used for robust expansion of human HSCs, particularly HSCs derived from cord blood.

These findings are important especially in light of the convenience of adding these factors to cultures of HSCs in vitro, and they can therefore be used in future attempts to expand HSCs for the clinic and possibly also to increase the efficiency of gene delivery to HSCs.

On the role of osteoblasts

The spatial organization of hematopoietic cells within the BM, with primitive cells distributed closer to the bone surface and more differentiated cells positioned closer to the BM longitudinal axis of the femur, has long been recognized.102,103  As early as 1978, Schofield proposed the niche hypothesis of a physiologically limited microenvironment supporting stem cells in vivo.104  Because of the anatomic complexity of mammalian stem cell niches, the precise location as well as the cellular and molecular basis for these structures has only recently begun to be uncovered. Several lines of evidence now converge to suggest that the osteoblasts, cells of mesenchymal origin positioned at the endosteal surface of bone, are essential components of the HSC niche.105  Using a transgenic mouse model expressing a constitutively active Parathyroid hormone receptor under an osteoblast specific promoter (α1(I) collagen), Calvi et al demonstrated that an increase in trabecular osteoblasts was accompanied by an increase in HSC numbers.45  Importantly, Calvi et al45  propose a model in which increased parathyroid hormone signaling in osteoblasts results in elevated levels of the Notch1 ligand, Jagged1, ultimately generating increased activation of Notch1 signaling in HSCs. An independent study investigated the role of the BMP signaling pathway to regulate HSC 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. Interestingly, this correlated with an increase in the number of HSCs, which were found attached to N-cadherin+ osteoblasts. Moreover, depletion of osteoblasts in vivo was associated with decreased BM cellularity and extramedullary hematopoiesis, further emphasizing the importance of osteoblasts for HSC function.107  In addition, Arai et al identified Tie2/Angiopoietin1 signaling to be critical for the maintenance of HSCs in a quiescent state in the BM niche.108  Tie2 is a receptor tyrosine kinase expressed on endothelial cells and HSCs, whereas Angiopoietin1 (Ang1) is expressed by osteoblasts. Tie2+ HSCs adhered to Ang1 expressing osteoblasts, supposedly through a mechanism involving N-cadherin.

Even though osteoblasts clearly constitute part of the HSC niche, they are probably not the only players. Apart from soluble factors, matrix components and other cellular constituents are likely to also have important functions. One example is osteopontin, a matrix glycoprotein synthesized by osteoblasts, which has been associated with negative regulation of the HSC compartment.109,110  Furthermore, HSCs deficient in the calcium-sensing receptor were profoundly defective in localizing anatomically to the endosteal niche, a behavior that correlated with diminished adhesion to the extracellular matrix protein collagen I.111  These data indicate that the local calcium gradient is involved in retaining HSCs in close physical proximity to the endosteal surface of bone. Furthermore, it was recently shown that a defective niche could result in HSC disorders further emphasizing the regulatory function of the HSC niche in vivo.112,113 

Hypoxia and reactive oxygen species (ROS)

Another peculiar property of the BM microenvironment is its hypoxic nature, a fact recently highlighted by Parmar et al showing that HSCs are distributed predominantly at the lowest end of an oxygen gradient within the BM.114  This observation bears importance for several findings, which indicate that the level of oxidative stress influences HSC function. For example, mice deficient of the cell-cycle regulator Atm developed early onset BM failure, a phenotype accompanied by elevated levels of ROS in HSCs.115  Interestingly, ROS appear to activate the p38/MAPK pathway causing quiescent HSCs to cycle more frequently and eventually become exhausted.116  Moreover, members of the FoxO subfamily of forkhead transcription factors have been shown to protect HSCs from oxidative stress by up-regulating genes involved in their 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 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 loss of FoxOs could be rescued by administration of the antioxidant N-acetyl-L-cysteine. In light of these observations, it is conceivable that the hypoxic environment in which the HSCs reside may serve to protect them from oxygen radicals ultimately keeping them quiescent.

Despite the plethora of evidence pointing at the importance of the BM milieu, it would be premature to assume that the microenvironment is the sole regulator of HSC fate options. For instance, during ontogeny before the establishment of the BM niche, HSCs undergo extensive self-renewal in the fetal liver. Indeed, HSCs derived from fetal liver have been shown to encompass a more efficient regenerative potential compared with their adult counterparts when transplanted into irradiated recipients.118  Recent evidence uncovered by Bowie et al suggest the existence of an intrinsically regulated developmental switch, which abruptly changes the self-renewing properties of HSCs at the age of 3 to 4 weeks in the mouse.119  At this time the HSC compartment rapidly changes from a fully cycling population to a largely quiescent one.119  This change appears to be independent of the niche as it occurs well after the migration of HSCs to the BM and because it is not affected by transplantation into adult recipients.120  Interestingly, juvenile HSCs exhibit a greater sensitivity to SCF; thus, signaling events downstream of the c-kit receptor may be developmentally and intrinsically regulated, ultimately determining symmetric versus asymmetric self-renewal.121  To summarize, HSC behavior is regulated by joint efforts from intrinsic factors and environmental cues, both entities that are integrated in a more comprehensive and holistic view by the concept of the stem-cell niche (Figure 5).

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.

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.

Close modal

The process of finding donors that have compatible histocompatibility antigens for patients who need blood and marrow transplantation is often a challenge of significant magnitude. As of today, umbilical CB is being used increasingly as a source of HSCs because of the common availability of CB cells and the diversity of histocompatibility gene haplotypes that are available in banked CB samples.122  The number of HSCs in each CB sample is limited, and it would therefore greatly increase the applicability of this stem-cell source if the CB stem cells could be expanded ex vivo before transplantation. Stem-cell expansion can also theoretically be accomplished in vivo after engraftment of the transplanted cells, but this approach may involve greater risks compared with ex vivo expansion unless the growth stimuli used in vivo can be carefully controlled.123,124  Moreover, because culture conditions can be precisely defined and transient modulation of regulatory pathways more easily used in vitro, stem-cell expansion ex vivo presents an attractive approach. However, even this tactic has proven a great challenge because a successful stem-cell expansion involves symmetric self-renewal divisions of HSCs, where both daughter cells retain HSC properties.123  More commonly, HSCs grown in vitro undergo asymmetric divisions characterized by the production of one HSC and a more differentiated progenitor, or alternatively, a symmetric division where both progeny cells have lost their HSC potential.

Positive and negative regulators ultimately balance the transition from quiescence to proliferation of HSCs. Thus, the strategies for stem-cell expansion should involve activation of regulators that encourage HSC self-renewal and/or inhibition of pathways that mediate quiescence, differentiation, or apoptosis of HSCs. Because of the recent advances in understanding the mechanisms that control HSC self-renewal, novel methods to expand stem cells can now be developed.70,123,124  Some approaches require viral vector-mediated gene transfer to HSCs for efficient expansion. Ideally, the vectors should be nonintegrating and mediate transient gene expression for safety reasons.125,127  However, the safest approaches would involve soluble factors, for example, cytokines, developmental cues or factors like the Angptl proteins.

When the transcription factor HoxB4 was expressed in HSCs ex vivo, a more than 40-fold net expansion of murine HSCs was generated over a period of 10 to 14 days.128  Furthermore, modified HoxB4 proteins have been used successfully to stimulate proliferation of human and murine HSCs ex vivo.129,130  The soluble form of the HoxB4 protein could actively be taken up by HSCs and could be used without concerns for insertional mutagenesis or the potential adverse effects that continuous production of HoxB4 in HSCs might generate in vivo. However, the HSC expansion achieved by the soluble HoxB4 was quite modest compared with overexpression of HoxB4 within HSCs. Developmental cues that activate Notch and Wnt signaling in HSCs may also be useful to increase HSC self-renewal ex vivo. As mentioned previously, Wnt3A has been shown to expand murine repopulating HSCs and Wnt5A 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 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 the most promising soluble factors identified to date for expansion of murine HSCs.101  If the angiopoietin-like proteins would prove to expand human HSCs as efficiently as in the murine system, they may become a promising tool for future cell and gene therapy approaches. Furthermore, other positive regulatory factors, in combination with Angptl proteins, may be able to expand human HSCs by a factor of 10 during a relatively short and safe culture period, suitable for GMP conditions that will be required for the clinic. Worthy of mention are also fibroblast growth factors, which have been shown to sustain primitive murine hematopoietic cells in culture and to maintain their primitive phenotype.131,132  Interestingly, fibroblast growth factor-1 in conjunction with SCF, TPO, and IGF-2 resulted in a 20-fold increase of LT-HSCs during a 10-day culture period.133 

It should be emphasized that most of the knowledge about stem- cell expansion has come from studies in mice. There is a difference between mouse and human HSCs with regards to cytokine receptors, telomere biology, and proliferative capacity, and there will therefore be both differences and similarities in the approaches used to expand mouse and human HSCs. Recently, the protein nephroblastoma overexpressed (NOV/CCN3) has been shown to expand primitive human HSCs, and this protein should therefore be carefully evaluated for use in clinical stem-cell expansion protocols.134  As more detailed knowledge is unearthed concerning the regulatory pathways that govern HSC self-renewal, it may even be possible to modulate these pathways with small molecule drugs as has been shown in the modulation of Wnt signaling and the expression of p21 and HoxB4.54,135,136  As an example, chemicals that enhance prostaglandin E2 synthesis were recently reported to increase HSC numbers in zebrafish and mice.137  Undoubtedly, however, a major challenge still ahead will be the integration of major signaling pathways into networks of control mechanisms ultimately coupling extrinsic with intrinsic regulatory fate determinants. Such knowledge would open up new avenues for maintaining and expanding HSCs in vitro.

This work was supported by the European Commission (INHERINET, Gene Therapy of Hematopoietic Stem Cells for Inherited Diseases, and CONSERT, Concerted Safety and Efficiency Evaluation of Retroviral Transgenesis for Gene Therapy of Inherited Diseases); Swedish Gene Therapy Program; Swedish Medical Research Council; Swedish Children Cancer Foundation; Clinical Research Award from Lund University Hospital; Joint Program on Stem Cell Research; Juvenile Diabetes Research Foundation; and Swedish Medical Research Council (S.K.), the Wenner-Gren Foundations (U.B.), and Kungliga Fysiografiska Sällskapet (G.K. and U.B.). The Lund Stem Cell Center is supported by a Center of Excellence grant in life sciences from the Swedish Foundation for Strategic Research.

Contribution: All authors contributed to the writing of this manuscript.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Stefan Karlsson, Molecular Medicine and Gene Therapy, Lund University Hospital, BMC A12, 221 84 Lund, Sweden; e-mail: Stefan.Karlsson@med.lu.se.

1
Ogawa
M
Differentiation and proliferation of hematopoietic stem cells.
Blood
1993
81
2844
2853
2
Matsuzaki
Y
Kinjo
K
Mulligan
RC
Okano
H
Unexpectedly efficient homing capacity of purified murine hematopoietic stem cells.
Immunity
2004
20
87
93
3
Osawa
M
Hanada
K
Hamada
H
Nakauchi
H
Long-term lymphohematopoietic reconstitution by a single CD34-low/negative hematopoietic stem cell.
Science
1996
273
242
245
4
Spangrude
GJ
Heimfeld
S
Weissman
IL
Purification and characterization of mouse hematopoietic stem cells.
Science
1988
241
58
62
5
Okada
S
Nakauchi
H
Nagayoshi
K
Nishikawa
S
Miura
Y
Suda
T
In vivo and in vitro stem cell function of c-kit- and Sca-1-positive murine hematopoietic cells.
Blood
1992
80
3044
3050
6
Yang
L
Bryder
D
Adolfsson
J
et al
Identification of Lin(-)Sca1(+)kit(+)CD34(+)Flt3- short-term hematopoietic stem cells capable of rapidly reconstituting and rescuing myeloablated transplant recipients.
Blood
2005
105
2717
2723
7
Adolfsson
J
Borge
OJ
Bryder
D
et al
Upregulation of Flt3 expression within the bone marrow Lin(-)Sca1(+)c-kit(+) stem cell compartment is accompanied by loss of self-renewal capacity.
Immunity
2001
15
659
669
8
Morrison
SJ
Weissman
IL
The long-term repopulating subset of hematopoietic stem cells is deterministic and isolatable by phenotype.
Immunity
1994
1
661
673
9
Wolf
NS
Kone
A
Priestley
GV
Bartelmez
SH
In vivo and in vitro characterization of long-term repopulating primitive hematopoietic cells isolated by sequential Hoechst 33342-rhodamine 123 FACS selection.
Exp Hematol
1993
21
614
622
10
Goodell
MA
Brose
K
Paradis
G
Conner
AS
Mulligan
RC
Isolation and functional properties of murine hematopoietic stem cells that are replicating in vivo.
J Exp Med
1996
183
1797
1806
11
Kiel
MJ
Yilmaz
OH
Iwashita
T
Yilmaz
OH
Terhorst
C
Morrison
SJ
SLAM family receptors distinguish hematopoietic stem and progenitor cells and reveal endothelial niches for stem cells.
Cell
2005
121
1109
1121
12
McKenzie
JL
Takenaka
K
Gan
OI
Doedens
M
Dick
JE
Low rhodamine 123 retention identifies long-term human hematopoietic stem cells within the Lin-CD34+CD38- population.
Blood
2007
109
543
545
13
Dick
JE
Bhatia
M
Gan
O
Kapp
U
Wang
JC
Assay of human stem cells by repopulation of NOD/SCID mice.
Stem Cells
1997
15
Suppl 1
199
203
discussion 204
14
Till
JE
McCulloch
EA
Siminovitch
L
A stochastic model of stem cell proliferation, based on the growth of spleen colony-forming cells.
Proc Natl Acad Sci U S A
1964
51
29
36
15
Metcalf
D
Hematopoietic regulators: redundancy or subtlety?
Blood
1993
82
3515
3523
16
Enver
T
Heyworth
CM
Dexter
TM
Do stem cells play dice?
Blood
1998
92
348
351
discussion 352
17
Miller
CL
Eaves
CJ
Expansion in vitro of adult murine hematopoietic stem cells with transplantable lympho-myeloid reconstituting ability.
Proc Natl Acad Sci U S A
1997
94
13648
13653
18
Christensen
JL
Weissman
IL
Flk-2 is a marker in hematopoietic stem cell differentiation: a simple method to isolate long-term stem cells.
Proc Natl Acad Sci U S A
2001
98
14541
14546
19
Audet
J
Miller
CL
Rose-John
S
Piret
JM
Eaves
CJ
Distinct role of gp130 activation in promoting self-renewal divisions by mitogenically stimulated murine hematopoietic stem cells.
Proc Natl Acad Sci U S A
2001
98
1757
1762
20
Nandurkar
HH
Robb
L
Tarlinton
D
Barnett
L
Kontgen
F
Begley
CG
Adult mice with targeted mutation of the interleukin-11 receptor (IL11Ra) display normal hematopoiesis.
Blood
1997
90
2148
2159
21
Kimura
S
Roberts
AW
Metcalf
D
Alexander
WS
Hematopoietic stem cell deficiencies in mice lacking c-Mpl, the receptor for thrombopoietin.
Proc Natl Acad Sci U S A
1998
95
1195
1200
22
Solar
GP
Kerr
WG
Zeigler
FC
et al
Role of c-mpl in early hematopoiesis.
Blood
1998
92
4
10
23
Ikuta
K
Weissman
IL
Evidence that hematopoietic stem cells express mouse c-kit but do not depend on steel factor for their generation.
Proc Natl Acad Sci U S A
1992
89
1502
1506
24
Buza-Vidas
N
Antonchuk
J
Qian
H
et al
Cytokines regulate postnatal hematopoietic stem cell expansion: opposing roles of thrombopoietin and LNK.
Genes Dev
2006
20
2018
2023
25
Borge
OJ
Ramsfjell
V
Veiby
OP
Murphy
MJ
Jr
Lok
S
Jacobsen
SE
Thrombopoietin, but not erythropoietin promotes viability and inhibits apoptosis of multipotent murine hematopoietic progenitor cells in vitro.
Blood
1996
88
2859
2870
26
Pestina
TI
Cleveland
JL
Yang
C
Zambetti
GP
Jackson
CW
Mpl ligand prevents lethal myelosuppression by inhibiting p53-dependent apoptosis.
Blood
2001
98
2084
2090
27
Seita
J
Ema
H
Ooehara
J
et al
Lnk negatively regulates self-renewal of hematopoietic stem cells by modifying thrombopoietin-mediated signal transduction.
Proc Natl Acad Sci U S A
2007
104
2349
2354
28
Takaki
S
Sauer
K
Iritani
BM
et al
Control of B cell production by the adaptor protein lnk: definition of a conserved family of signal-modulating proteins.
Immunity
2000
13
599
609
29
Tong
W
Zhang
J
Lodish
HF
Lnk inhibits erythropoiesis and Epo-dependent JAK2 activation and downstream signaling pathways.
Blood
2005
105
4604
4612
30
Velazquez
L
Cheng
AM
Fleming
HE
et al
Cytokine signaling and hematopoietic homeostasis are disrupted in Lnk-deficient mice.
J Exp Med
2002
195
1599
1611
31
Ema
H
Sudo
K
Seita
J
et al
Quantification of self-renewal capacity in single hematopoietic stem cells from normal and Lnk-deficient mice.
Dev Cell
2005
8
907
914
32
Ellisen
LW
Bird
J
West
DC
et al
TAN-1, the human homolog of the Drosophila notch gene, is broken by chromosomal translocations in T lymphoblastic neoplasms.
Cell
1991
66
649
661
33
Radtke
F
Wilson
A
Mancini
SJ
MacDonald
HR
Notch regulation of lymphocyte development and function.
Nat Immunol
2004
5
247
253
34
Milner
LA
Kopan
R
Martin
DI
Bernstein
ID
A human homologue of the Drosophila developmental gene, Notch, is expressed in CD34+ hematopoietic precursors.
Blood
1994
83
2057
2062
35
Karanu
FN
Murdoch
B
Gallacher
L
et al
The notch ligand jagged-1 represents a novel growth factor of human hematopoietic stem cells.
J Exp Med
2000
192
1365
1372
36
Karanu
FN
Murdoch
B
Miyabayashi
T
et al
Human homologues of Delta-1 and Delta-4 function as mitogenic regulators of primitive human hematopoietic cells.
Blood
2001
97
1960
1967
37
Varnum-Finney
B
Brashem-Stein
C
Bernstein
ID
Combined effects of Notch signaling and cytokines induce a multiple log increase in precursors with lymphoid and myeloid reconstituting ability.
Blood
2003
101
1784
1789
38
Vas
V
Szilagyi
L
Paloczi
K
Uher
F
Soluble Jagged-1 is able to inhibit the function of its multivalent form to induce hematopoietic stem cell self-renewal in a surrogate in vitro assay.
J Leukoc Biol
2004
75
714
720
39
Ohishi
K
Varnum-Finney
B
Bernstein
ID
Delta-1 enhances marrow and thymus repopulating ability of human CD34(+)CD38(-) cord blood cells.
J Clin Invest
2002
110
1165
1174
40
Delaney
C
Varnum-Finney
B
Aoyama
K
Brashem-Stein
C
Bernstein
ID
Dose-dependent effects of the Notch ligand Delta1 on ex vivo differentiation and in vivo marrow repopulating ability of cord blood cells.
Blood
2005
106
2693
2699
41
Suzuki
A
Raya
A
Kawakami
Y
et al
Nanog binds to Smad1 and blocks bone morphogenetic protein-induced differentiation of embryonic stem cells.
Proc Natl Acad Sci U S A
2006
103
10294
10299
42
Kunisato
A
Chiba
S
Nakagami-Yamaguchi
E
et al
HES-1 preserves purified hematopoietic stem cells ex vivo and accumulates side population cells in vivo.
Blood
2003
101
1777
1783
43
Stier
S
Cheng
T
Dombkowski
D
Carlesso
N
Scadden
DT
Notch1 activation increases hematopoietic stem cell self-renewal in vivo and favors lymphoid over myeloid lineage outcome.
Blood
2002
99
2369
2378
44
Varnum-Finney
B
Xu
L
Brashem-Stein
C
et al
Pluripotent, cytokine-dependent, hematopoietic stem cells are immortalized by constitutive Notch1 signaling.
Nat Med
2000
6
1278
1281
45
Calvi
LM
Adams
GB
Weibrecht
KW
et al
Osteoblastic cells regulate the haematopoietic stem cell niche.
Nature
2003
425
841
846
46
Duncan
AW
Rattis
FM
DiMascio
LN
et al
Integration of Notch and Wnt signaling in hematopoietic stem cell maintenance.
Nat Immunol
2005
6
314
322
47
Mancini
SJ
Mantei
N
Dumortier
A
Suter
U
MacDonald
HR
Radtke
F
Jagged1-dependent Notch signaling is dispensable for hematopoietic stem cell self-renewal and differentiation.
Blood
2005
105
2340
2342
48
Austin
TW
Solar
GP
Ziegler
FC
Liem
L
Matthews
W
A role for the Wnt gene family in hematopoiesis: expansion of multilineage progenitor cells.
Blood
1997
89
3624
3635
49
Willert
K
Brown
JD
Danenberg
E
et al
Wnt proteins are lipid-modified and can act as stem cell growth factors.
Nature
2003
423
448
452
50
Van Den Berg
DJ
Sharma
AK
Bruno
E
Hoffman
R
Role of members of the Wnt gene family in human hematopoiesis.
Blood
1998
92
3189
3202
51
Reya
T
Duncan
AW
Ailles
L
et al
A role for Wnt signalling in self-renewal of haematopoietic stem cells.
Nature
2003
423
409
414
52
Kirstetter
P
Anderson
K
Porse
BT
Jacobsen
SE
Nerlov
C
Activation of the canonical Wnt pathway leads to loss of hematopoietic stem cell repopulation and multilineage differentiation block.
Nat Immunol
2006
7
1048
1056
53
Cobas
M
Wilson
A
Ernst
B
et al
Beta-catenin is dispensable for hematopoiesis and lymphopoiesis.
J Exp Med
2004
199
221
229
54
Trowbridge
JJ
Xenocostas
A
Moon
RT
Bhatia
M
Glycogen synthase kinase-3 is an in vivo regulator of hematopoietic stem cell repopulation.
Nat Med
2006
12
89
98
55
Sitnicka
E
Ruscetti
FW
Priestley
GV
Wolf
NS
Bartelmez
SH
Transforming growth factor beta 1 directly and reversibly inhibits the initial cell divisions of long-term repopulating hematopoietic stem cells.
Blood
1996
88
82
88
56
Batard
P
Monier
MN
Fortunel
N
et al
TGF-(beta)1 maintains hematopoietic immaturity by a reversible negative control of cell cycle and induces CD34 antigen up-modulation.
J Cell Sci
2000
113
Pt 3
383
390
57
Garbe
A
Spyridonidis
A
Mobest
D
Schmoor
C
Mertelsmann
R
Henschler
R
Transforming growth factor-beta 1 delays formation of granulocyte-macrophage colony-forming cells, but spares more primitive progenitors during ex vivo expansion of CD34+ haemopoietic progenitor cells.
Br J Haematol
1997
99
951
958
58
Hatzfeld
J
Li
ML
Brown
EL
et al
Release of early human hematopoietic progenitors from quiescence by antisense transforming growth factor beta 1 or Rb oligonucleotides.
J Exp Med
1991
174
925
929
59
Fortunel
N
Batard
P
Hatzfeld
A
et al
High proliferative potential-quiescent cells: a working model to study primitive quiescent hematopoietic cells.
J Cell Sci
1998
111
Pt 13
1867
1875
60
Soma
T
Yu
JM
Dunbar
CE
Maintenance of murine long-term repopulating stem cells in ex vivo culture is affected by modulation of transforming growth factor-beta but not macrophage inflammatory protein-1 alpha activities.
Blood
1996
87
4561
4567
61
Jacobsen
SE
Ruscetti
FW
Dubois
CM
Lee
J
Boone
TC
Keller
JR
Transforming growth factor-beta trans-modulates the expression of colony stimulating factor receptors on murine hematopoietic progenitor cell lines.
Blood
1991
77
1706
1716
62
Dubois
CM
Ruscetti
FW
Palaszynski
EW
Falk
LA
Oppenheim
JJ
Keller
JR
Transforming growth factor beta is a potent inhibitor of interleukin 1 (IL-1) receptor expression: proposed mechanism of inhibition of IL-1 action.
J Exp Med
1990
172
737
744
63
Dubois
CM
Ruscetti
FW
Stankova
J
Keller
JR
Transforming growth factor-beta regulates c-kit message stability and cell-surface protein expression in hematopoietic progenitors.
Blood
1994
83
3138
3145
64
Sansilvestri
P
Cardoso
AA
Batard
P
et al
Early CD34high cells can be separated into KIThigh cells in which transforming growth factor-beta (TGF-beta) downmodulates c-kit and KITlow cells in which anti-TGF-beta upmodulates c-kit.
Blood
1995
86
1729
1735
65
Ducos
K
Panterne
B
Fortunel
N
Hatzfeld
A
Monier
MN
Hatzfeld
J
p21(cip1) mRNA is controlled by endogenous transforming growth factor-beta1 in quiescent human hematopoietic stem/progenitor cells.
J Cell Physiol
2000
184
80
85
66
Cheng
T
Shen
H
Rodrigues
N
Stier
S
Scadden
DT
Transforming growth factor beta 1 mediates cell-cycle arrest of primitive hematopoietic cells independent of p21(Cip1/Waf1) or p27(Kip1).
Blood
2001
98
3643
3649
67
Dao
MA
Taylor
N
Nolta
JA
Reduction in levels of the cyclin-dependent kinase inhibitor p27(kip-1) coupled with transforming growth factor beta neutralization induces cell-cycle entry and increases retroviral transduction of primitive human hematopoietic cells.
Proc Natl Acad Sci U S A
1998
95
13006
13011
68
Dao
MA
Hwa
J
Nolta
JA
Molecular mechanism of transforming growth factor beta-mediated cell-cycle modulation in primary human CD34(+) progenitors.
Blood
2002
99
499
506
69
Scandura
JM
Boccuni
P
Massague
J
Nimer
SD
Transforming growth factor beta-induced cell cycle arrest of human hematopoietic cells requires p57KIP2 up-regulation.
Proc Natl Acad Sci U S A
2004
101
15231
15236
70
Larsson
J
Karlsson
S
The role of Smad signaling in hematopoiesis.
Oncogene
2005
24
5676
5692
71
Ruscetti
FW
Akel
S
Bartelmez
SH
Autocrine transforming growth factor-beta regulation of hematopoiesis: many outcomes that depend on the context.
Oncogene
2005
24
5751
5763
72
Fortunel
NO
Hatzfeld
A
Hatzfeld
JA
Transforming growth factor-beta: pleiotropic role in the regulation of hematopoiesis.
Blood
2000
96
2022
2036
73
Yaswen
L
Kulkarni
AB
Fredrickson
T
et al
Autoimmune manifestations in the transforming growth factor-beta 1 knockout mouse.
Blood
1996
87
1439
1445
74
Letterio
JJ
Roberts
AB
Regulation of immune responses by TGF-beta.
Annu Rev Immunol
1998
16
137
161
75
Levéen
P
Larsson
J
Ehinger
M
et al
Induced disruption of the transforming growth factor beta type II receptor gene in mice causes a lethal inflammatory disorder that is transplantable.
Blood
2002
100
560
568
76
Letterio
JJ
Geiser
AG
Kulkarni
AB
et al
Autoimmunity associated with TGF-beta1-deficiency in mice is dependent on MHC class II antigen expression.
J Clin Invest
1996
98
2109
2119
77
Larsson
J
Blank
U
Helgadottir
H
et al
TGF-beta signaling-deficient hematopoietic stem cells have normal self-renewal and regenerative ability in vivo despite increased proliferative capacity in vitro.
Blood
2003
102
3129
3135
78
Larsson
J
Blank
U
Klintman
J
Magnusson
M
Karlsson
S
Quiescence of hematopoietic stem cells and maintenance of the stem cell pool is not dependent on TGF-beta signaling in vivo.
Exp Hematol
2005
33
592
596
79
Huber
TL
Zhou
Y
Mead
PE
Zon
LI
Cooperative effects of growth factors involved in the induction of hematopoietic mesoderm.
Blood
1998
92
4128
4137
80
Maeno
M
Mead
PE
Kelley
C
et al
The role of BMP-4 and GATA-2 in the induction and differentiation of hematopoietic mesoderm in Xenopus laevis.
Blood
1996
88
1965
1972
81
Schmid
B
Furthauer
M
Connors
SA
et al
Equivalent genetic roles for bmp7/snailhouse and bmp2b/swirl in dorsoventral pattern formation.
Development
2000
127
957
967
82
Schmerer
M
Evans
T
Primitive erythropoiesis is regulated by Smad-dependent signaling in postgastrulation mesoderm.
Blood
2003
102
3196
3205
83
Winnier
G
Blessing
M
Labosky
PA
Hogan
BL
Bone morphogenetic protein-4 is required for mesoderm formation and patterning in the mouse.
Genes Dev
1995
9
2105
2116
84
Bhatia
M
Bonnet
D
Wu
D
et al
Bone morphogenetic proteins regulate the developmental program of human hematopoietic stem cells.
J Exp Med
1999
189
1139
1148
85
Bhardwaj
G
Murdoch
B
Wu
D
et al
Sonic hedgehog induces the proliferation of primitive human hematopoietic cells via BMP regulation.
Nat Immunol
2001
2
172
180
86
Utsugisawa
T
Moody
JL
Aspling
M
Nilsson
E
Carlsson
L
Karlsson
S
A road map towards defining the role of Smad signaling in hematopoietic stem cells.
Stem Cells
2006
24
1128
1136
87
Liu
B
Sun
Y
Jiang
F
et al
Disruption of Smad5 gene leads to enhanced proliferation of high-proliferative potential precursors during embryonic hematopoiesis.
Blood
2003
101
124
133
88
Singbrant
S
Moody
JL
Blank
U
et al
Smad5 is dispensable for adult murine hematopoiesis.
Blood
2006
108
3707
3712
89
Blank
U
Karlsson
G
Moody
JL
et al
Smad7 promotes self-renewal of hematopoietic stem cells in vivo.
Blood
2006
108
4246
4254
90
Chadwick
K
Shojaei
F
Gallacher
L
Bhatia
M
Smad7 alters cell fate decisions of human hematopoietic repopulating cells.
Blood
2005
105
1905
1915
91
Moustakas
A
Heldin
CH
Non-Smad TGF-beta signals.
J Cell Sci
2005
118
3573
3584
92
Karlsson
G
Blank
U
Moody
JL
et al
Smad4 is critical for self-renewal of hematopoietic stem cells.
J Exp Med
2007
204
467
474
93
Nishita
M
Hashimoto
MK
Ogata
S
et al
Interaction between Wnt and TGF-beta signalling pathways during formation of Spemann's organizer.
Nature
2000
403
781
785
94
Labbé
E
Letamendia
A
Attisano
L
Association of Smads with lymphoid enhancer binding factor 1/T cell-specific factor mediates cooperative signaling by the transforming growth factor-beta and wnt pathways.
Proc Natl Acad Sci U S A
2000
97
8358
8363
95
Itoh
F
Itoh
S
Goumans
MJ
et al
Synergy and antagonism between Notch and BMP receptor signaling pathways in endothelial cells.
EMBO J
2004
23
541
551
96
He
W
Dorn
DC
Erdjument-Bromage
H
Tempst
P
Moore
MA
Massague
J
Hematopoiesis controlled by distinct TIF1gamma and Smad4 branches of the TGFbeta pathway.
Cell
2006
125
929
941
97
Ransom
DG
Bahary
N
Niss
K
et al
The zebrafish moonshine gene encodes transcriptional intermediary factor 1gamma, an essential regulator of hematopoiesis.
PLoS Biol
2004
2
E237
98
Dupont
S
Zacchigna
L
Cordenonsi
M
et al
Germ-layer specification and control of cell growth by Ectodermin, a Smad4 ubiquitin ligase.
Cell
2005
121
87
99
99
Kroon
E
Krosl
J
Thorsteinsdottir
U
Baban
S
Buchberg
AM
Sauvageau
G
Hoxa9 transforms primary bone marrow cells through specific collaboration with Meis1a but not Pbx1b.
EMBO J
1998
17
3714
3725
100
Wang
N
Kim
HG
Cotta
CV
et al
TGFbeta/BMP inhibits the bone marrow transformation capability of Hoxa9 by repressing its DNA-binding ability.
EMBO J
2006
25
1469
1480
101
Zhang
CC
Kaba
M
Ge
G
et al
Angiopoietin-like proteins stimulate ex vivo expansion of hematopoietic stem cells.
Nat Med
2006
12
240
245
102
Lord
BI
Testa
NG
Hendry
JH
The relative spatial distributions of CFUs and CFUc in the normal mouse femur.
Blood
1975
46
65
72
103
Gong
JK
Endosteal marrow: a rich source of hematopoietic stem cells.
Science
1978
199
1443
1445
104
Schofield
R
The relationship between the spleen colony-forming cell and the haemopoietic stem cell.
Blood Cells
1978
4
7
25
105
Wilson
A
Trumpp
A
Bone-marrow haematopoietic-stem-cell niches.
Nat Rev Immunol
2006
6
93
106
106
Zhang
J
Niu
C
Ye
L
et al
Identification of the haematopoietic stem cell niche and control of the niche size.
Nature
2003
425
836
841
107
Visnjic
D
Kalajzic
Z
Rowe
DW
Katavic
V
Lorenzo
J
Aguila
HL
Hematopoiesis is severely altered in mice with an induced osteoblast deficiency.
Blood
2004
103
3258
3264
108
Arai
F
Hirao
A
Ohmura
M
et al
Tie2/angiopoietin-1 signaling regulates hematopoietic stem cell quiescence in the bone marrow niche.
Cell
2004
118
149
161
109
Stier
S
Ko
Y
Forkert
R
et al
Osteopontin is a hematopoietic stem cell niche component that negatively regulates stem cell pool size.
J Exp Med
2005
201
1781
1791
110
Nilsson
SK
Johnston
HM
Whitty
GA
et al
Osteopontin, a key component of the hematopoietic stem cell niche and regulator of primitive hematopoietic progenitor cells.
Blood
2005
106
1232
1239
111
Adams
GB
Chabner
KT
Alley
IR
et al
Stem cell engraftment at the endosteal niche is specified by the calcium-sensing receptor.
Nature
2006
439
599
603
112
Walkley
CR
Olsen
GH
Dworkin
S
et al
A microenvironment-induced myeloproliferative syndrome caused by retinoic acid receptor gamma deficiency.
Cell
2007
129
1097
1110
113
Walkley
CR
Shea
JM
Sims
NA
Purton
LE
Orkin
SH
Rb regulates interactions between hematopoietic stem cells and their bone marrow microenvironment.
Cell
2007
129
1081
1095
114
Parmar
K
Mauch
P
Vergilio
JA
Sackstein
R
Down
JD
Distribution of hematopoietic stem cells in the bone marrow according to regional hypoxia.
Proc Natl Acad Sci U S A
2007
104
5431
5436
115
Ito
K
Hirao
A
Arai
F
et al
Regulation of oxidative stress by ATM is required for self-renewal of haematopoietic stem cells.
Nature
2004
431
997
1002
116
Ito
K
Hirao
A
Arai
F
et al
Reactive oxygen species act through p38 MAPK to limit the lifespan of hematopoietic stem cells.
Nat Med
2006
12
446
451
117
Tothova
Z
Kollipara
R
Huntly
BJ
et al
FoxOs are critical mediators of hematopoietic stem cell resistance to physiologic oxidative stress.
Cell
2007
128
325
339
118
Rebel
VI
Miller
CL
Eaves
CJ
Lansdorp
PM
The repopulation potential of fetal liver hematopoietic stem cells in mice exceeds that of their liver adult bone marrow counterparts.
Blood
1996
87
3500
3507
119
Bowie
MB
McKnight
KD
Kent
DG
McCaffrey
L
Hoodless
PA
Eaves
CJ
Hematopoietic stem cells proliferate until after birth and show a reversible phase-specific engraftment defect.
J Clin Invest
2006
116
2808
2816
120
Bowie
MB
Kent
DG
Dykstra
B
et al
Identification of a new intrinsically timed developmental checkpoint that reprograms key hematopoietic stem cell properties.
Proc Natl Acad Sci U S A
2007
104
5878
5882
121
Bowie
MB
Kent
DG
Copley
MR
Eaves
CJ
Steel factor responsiveness regulates the high self-renewal phenotype of fetal hematopoietic stem cells.
Blood
2007
109
5043
5048
122
Gluckman
E
Broxmeyer
HA
Auerbach
AD
et al
Hematopoietic reconstitution in a patient with Fanconi's anemia by means of umbilical cord blood from an HLA-identical sibling.
N Engl J Med
1989
321
1174
1178
123
Attar
EC
Scadden
DT
Regulation of hematopoietic stem cell growth.
Leukemia
2004
18
1760
1768
124
Sorrentino
BP
Clinical strategies for expansion of haematopoietic stem cells.
Nat Rev Immunol
2004
4
878
888
125
Baum
C
Dullmann
J
Li
Z
et al
Side effects of retroviral gene transfer into hematopoietic stem cells.
Blood
2003
101
2099
2114
126
Hacein-Bey-Abina
S
Von Kalle
C
Schmidt
M
et al
LMO2-associated clonal T cell proliferation in two patients after gene therapy for SCID-X1.
Science
2003
302
415
419
127
Woods
NB
Muessig
A
Schmidt
M
et al
Lentiviral vector transduction of NOD/SCID repopulating cells results in multiple vector integrations per transduced cell: risk of insertional mutagenesis.
Blood
2003
101
1284
1289
128
Antonchuk
J
Sauvageau
G
Humphries
RK
HOXB4-induced expansion of adult hematopoietic stem cells ex vivo.
Cell
2002
109
39
45
129
Amsellem
S
Pflumio
F
Bardinet
D
et al
Ex vivo expansion of human hematopoietic stem cells by direct delivery of the HOXB4 homeoprotein.
Nat Med
2003
9
1423
1427
130
Krosl
J
Austin
P
Beslu
N
Kroon
E
Humphries
RK
Sauvageau
G
In vitro expansion of hematopoietic stem cells by recombinant TAT-HOXB4 protein.
Nat Med
2003
9
1428
1432
131
de Haan
G
Weersing
E
Dontje
B
et al
In vitro generation of long-term repopulating hematopoietic stem cells by fibroblast growth factor-1.
Dev Cell
2003
4
241
251
132
Yeoh
JS
van Os
R
Weersing
E
et al
Fibroblast growth factor-1 and -2 preserve long-term repopulating ability of hematopoietic stem cells in serum-free cultures.
Stem Cells
2006
24
1564
1572
133
Zhang
CC
Lodish
HF
Murine hematopoietic stem cells change their surface phenotype during ex vivo expansion.
Blood
2005
105
4314
4320
134
Gupta
R
Hong
D
Iborra
F
Sarno
S
Enver
T
NOV (CCN3) functions as a regulator of human hematopoietic stem or progenitor cells.
Science
2007
316
590
593
135
Bug
G
Gul
H
Schwarz
K
et al
Valproic acid stimulates proliferation and self-renewal of hematopoietic stem cells.
Cancer Res
2005
65
2537
2541
136
De Felice
L
Tatarelli
C
Mascolo
MG
et al
Histone deacetylase inhibitor valproic acid enhances the cytokine-induced expansion of human hematopoietic stem cells.
Cancer Res
2005
65
1505
1513
137
North
TE
Goessling
W
Walkley
CR
et al
Prostaglandin E2 regulates vertebrate haematopoietic stem cell homeostasis.
Nature
2007
447
1007
1011
138
Chiba
S
Normal and deregulated notch signaling in regulation of hematopoietic stem cells and development of leukemia [in Japanese].
Rinsho Ketsueki
2006
47
371
378
139
Barker
N
Clevers
H
Mining the Wnt pathway for cancer therapeutics.
Nat Rev Drug Discov
2006
5
997
1014
140
Clevers
H
Wnt/beta-catenin signaling in development and disease.
Cell
2006
127
469
480
141
Massagué
J
TGF-beta signal transduction.
Annu Rev Biochem
1998
67
753
791
142
Heldin
CH
Miyazono
K
ten Dijke
P
TGF-beta signalling from cell membrane to nucleus through SMAD proteins.
Nature
1997
390
465
471
143
Nakao
A
Afrakhte
M
Moren
A
et al
Identification of Smad7, a TGFbeta-inducible antagonist of TGF-beta signalling.
Nature
1997
389
631
635
144
Hayashi
H
Abdollah
S
Qiu
Y
et al
The MAD-related protein Smad7 associates with the TGFbeta receptor and functions as an antagonist of TGFbeta signaling.
Cell
1997
89
1165
1173
145
Souchelnytskyi
S
Nakayama
T
Nakao
A
et al
Physical and functional interaction of murine and Xenopus Smad7 with bone morphogenetic protein receptors and transforming growth factor-beta receptors.
J Biol Chem
1998
273
25364
25370
146
Miyamoto
K
Araki
KY
Naka
K
et al
Foxo3a is essential for maintenance of the hematopoietic stem cell pool.
Cell Stem
2007
1
101
112
Sign in via your Institution