Abstract
Blood and bone are dynamic tissues that are continuously renewed through out life. Early observations based upon the proximity of bone and hematopoietic progenitor populations in marrow suggested that interactions between skeletal and hematopoietic elements are likely to be crucial in the development and function of each system. As a result of these morphologic observations, several groups have demonstrated that the osteoblasts play an important role in hematopoiesis by serving as a specific local microenvironment, or “niche”, for hematopoietic stem cells (HSC). Significant new developments in this area of active investigation have emerged since our last examination of this area in 2005. Here we discuss these new insights into the function and morphology of the HSC niche with a particular focus on cells of the osteoblastic lineage.
Introduction
During the 3rd week of gestation, hematopoiesis is first initiated in the yolk sac. By the fifth week of development, primitive hematopoietic cells may be detected in aorta-gonad-metanephros region, liver and spleen [1]. By the 11th week of gestation, hematopoietic activity begins to shift towards the bone marrow such that by the time of birth, the marrow remains the major location for hematopoiesis in humans [1]. Marrow-derived hematopoietic stem cells (HSCs), which are the top of the hematopoietic hierarchy, have the potential to differentiate into all mature blood cells while maintaining the capacity for self-renewal. To provide a constant supply of mature blood elements, HSCs must maintain a balance between differentiation and self-renewal. The site that regulates self-renewal, proliferation and differentiation of HSCs has been termed either the HSC microenvironment or the ‘HSC niche’ [2]. Within the HSC niche, HSCs are likely to be exposed to both support/maintenance/renewal and growth signals. Most evidence indicates that combinations of cell intrinsic and extrinsic factors influence the decision of HSCs to self-renew or differentiate.
In the early 1970’s, studies examining the interactions between bone marrow stromal cells and HSCs provided clues that osteoblasts may play a central role in hematopoiesis [3-5]. In 1994, we hypothesized that osteoblasts play an important role in hematopoiesis in human system and demonstrated that human osteoblasts support maintenance of HSCs and expansion of progenitors in vitro [6]. In late 2003 and early 2004, several reports demonstrated that HSCs were closely localized to osteoblasts based upon morphology and the expression of N-cadherin and Ang-1 using murine models [7-9]. In addition, the essential roles of the osteoblastic niche in hematopoiesis were also confirmed by depleting the osteoblastic niche with ganciclovir in Col2.3Δ-TK transgenic mice [10, 11].
More recently, sinusoidal endothelium [12, 13], CXCL12 highly expressing reticular cells [14], adipocytes [15], and mesenchymal stem cells [16, 17] have been described as components of the HSC niche. However, what the function of these cells are, and how they control primitive hematopoiesis are poorly understood and remain controversial. In fact, only scant evidence exists as to what are the support/maintenance/renewal and growth signals produced by these cells that regulate HSC function. Recent controversy centers on where the niche is itself – is it located close to the endosteal surfaces or near vascular surfaces? Likely both endosteal and vascular components participate in the generation of the HSC niche. Yet how the functions of these two HSCs niches are synchronized is not clear. In this review, we will discuss the current understanding as to the various cells that participate in the function of the HSC niche, with a particular focus on what is known regarding the osteoblastic HSC niche.
HSC homing to the HSC niche
Beginning in the 11th week of fetal life, HSCs begin to migrate from the liver and spleen into the bone marrow where the majority of hematopoietic activity occurs after birth [1]. Later, under basal conditions, the majority of HSCs reside in the marrow. However a small but significant number of HSCs can be found in the circulation. The number of circulating HSCs may increase dramatically during infection or in response to recombinant growth factors. It is likely that these HSCs play a significant role in regulating and coordinating HSC activities across the organism where hematopoietic activities are widely dispersed.
Yet the abilities of circulating and donor HSCs to establish hematopoietic activities during transplantation are dependent on their ability to home and lodge in the marrow. The chemokine stromal derived factor-1 (SDF-1/CXCL12) is perhaps the best characterized molecule that regulates HSC homing. Two receptors CXCR4 and CXCR7 are known to bind CXCL12 and both CXCR4 and CXCR7 are expressed by HSCs [18]. Considerably more is known regarding the role that CXCR4 plays in regulating HSC homing and engraftment [19, 20], where competitive inhibitors have entered therapeutic trials as HSC mobilization agents [21]. Several cell types have been identified as sources of CXCL12 in normal marrow. Osteoblasts appear to be a major source of CXCL12 [22] where synthesis is regulated by several inflammatory stimuli, DNA damaging agents and hormones. Moreover, osteoblastic production of CXCL12 appears to be regulated by circadian rhythms [23, 24]. Importantly, CXCL12 production appears to be an early feature of the osteoblastic lineage, while CXCL12 production is down regulated upon maturation and matrix synthesis [20, 25].
Vascular endothelial cells represent other cellular source of CXCL12 in the marrow [26]. Less is understood regarding regulation and production of CXCL12 by these cells. However, a unique function for CXCR4 expressed on marrow endothelial cells has been described. Endothelial CXCR4 internalizes circulating CXCL12 to translocate the protein into the bone marrow [27]. By this mechanism, CXCL12 has been demonstrated to increase the homing of transplanted human CD34+ hematopoietic progenitors into the bone marrow. The transporter function of CXCR4 appears to be cell type specific where endothelial and other stromal cells share in the capacity, but not of hematopoietic cells. Whether osteoblasts share in this capacity has not been determined and what the physiologic significance of such a mechanism to place CXCL12 in the bone matrix is uncertain, but CXCL12 could serve as a mechanism to localize osteoclasts into areas requiring active bone turnover [28].
Additional molecular regulators of HSC homing have also been recently identified in addition to CXCL12. For example, under basal conditions, the normal physiologic oxygen tension in marrow is hypoxic particularly at the level of the endosteal/osteoblastic niche. One thought suggests that a low O2 environment is necessary for the ability of HSCs to maintain their stemness [29]. In addition, hypoxia induces both CXCL12 secretion [30] and CXCR4 expression [31]. Together, these observations suggest that hypoxia may be involved in the recruitment of HSCs to the niche through the regulation of the CXCR4/CXCL12 axis. Just as low oxygen levels are thought to increase CXCL12, degradation of CXCL12 is also likely to play a major role in regulating HSC levels in the marrow. In particular, a report suggesting that CD26 plays a major role in degrading CXCL12 is of considerable interest. As blocking CD26 activity lead to enhanced engraftment of HSCs into marrow niches [32], although this has recently been called into question [33].
Over the last several years, CXCL12-independent mechanisms of HSC homing have also been identified. For example, expression of the calcium-sensing receptor (CaR) and the guanine-nucleotide-binding protein stimulatory α subunit (Gαs) expressed by HSCs plays a crucial role in their homing to osteoblastic niche [34, 35]. CaRs are expressed mostly by HSC in the marrow and are thought to be related to the localization of HSCs to the endosteal surfaces [34]. Consequently, HSCs derived from CaR−/− mice fail to engraft into wild-type marrow [34]. Likewise, HSCs from Gαs−/− mice have lost the ability of homing to the marrow [35]. In fact, when Gαs was conditional deleted in HSCs after engraftment to wild-type bone marrow microenvironment, the migratory ability of Gαs−/− HSCs in response to granulocyte colony-stimulating factor (G-CSF) treatment was impaired compare to Gαs+/+ HSCs [35]. In contrast, when Gαs activity in HSCs was stimulated using cholera toxin (a compound known to constitutively activate Gαs by preventing GTP hydrolysis from the ADP-ribose-Gαs-GTP complex), the ability to engraft and home to the marrow is enhanced [35].
As maintaining chemokine gradients are critical for HSC homing, occupancy of established niches also appears to regulate the ability of transplanted HSCs to engraft into the niche. Without available or “vacant niches”, exogenous (or donor) HSCs are not able to lodge in the HSC niche occupied by endogenous (or recipient) HSCs. Thus, pre-treatment with the CXCR4 antagonist AMD3100 prior to transplantation facilitates stem cell engraftment [36, 37]. Likewise, antibody to c-kit (ACK2) which is expressed by HSCs has been shown to increase the level of circulating HSCs by opening up the HSC niches to prepare niche space for donor HSCs to engraft into the marrow of recipient animals [38]. Clearly, these observations along with studies on how to prevent CXCL12 degradation offer significant hope as either pre-conditioning or HSC mobilization regimes during stem cell transplantation.
Adhesion interactions between HSC and HSC niche
Direct cell-to-cell and/or cell-to-extra cellular matrix contacts between HSCs and their niche are thought to play a crucial role in the regulation of HSC fate, although what adhesion molecules maintain HSCs in the marrow remains unclear [20]. Mouse models reveal that adhesion interactions between HSCs and the osteoblastic niche are partially regulated by Tie2/Ang-1 and N-cadherin/N-cadherin interactions [8, 9]. However, controversies as to the roles of N-cadherin in the early hematopoiesis have also been appreciated [39]. Earlier work by Zhang et al in 2003 suggested that N-cadherin expressing “spindle shape osteoblasts” are critical components of the HSC niche, and approximately 10% of HSCs express N-cadherin [9]. Homotypic binding between HSCs and their niche via N-cadherin appeared to be of great significance [9]. Nonetheless, recent studies have revealed that primitive HSCs express low or no N-cadherin on their surface [40, 41], whereas mature blood cells express high levels of N-cadherin [41]. A more recent study using N-cadherin knock-out animals failed to demonstrate the effects of N-cadherin on HSC maintenance [42]. Moreover, although the N-cadherin in osteoblasts was conditionally deleted using Cre/lox system, the function of HSCs, the engraftment capacity of wild-type cells, and the responses to the G-CSF treatment in those animals are not impaired [43]. Thus the role of N-cadherin homodimers regulating HSC interactions within the niche remains unclear. N-cadherin may however regulate niche interactions of either more mature progenitor cells or via heterodimers with other receptor molecules expressed by HSCs.
The α4β1 integrin, or very late antigen-4 (VLA-4), is also widely known as one of the adhesion molecules involved in HSCs fate. Vascular cell adhesion molecule-1 (VCAM-1), a ligand for VLA-4, is known to be expressed by stromal reticular cells [44], endothelial cells lining bone marrow sinusoids [44] and osteoblasts in the marrow [10]. When serial transplantation assays were performed using VLA-4 conditional deleted HSCs, the HSC homing and self-renewal are diminished [45]. Strikingly, more hematopoietic progenitor cells (HPCs) were found by colony-forming-unit assays in the peripheral blood recovered from VLA-4−/− chimeras compared with the animals transplanted with wild-type marrow [45]. Intriguingly, one-third of the animals survived when VLA-4−/− peripheral blood cells were transplanted into wild-type animals, whereas no animals survived when transplanted with wild-type peripheral blood [45].
Another molecule of great interest to our group that is involved in the HSC homing and binding to the niche is annexin II (Anxa2). Moreover, it has been demonstrated that Anxa2 expressed by osteoblastic niche serves as an anchor for CXCL12 to support the HSCs homing to the niche [46]. Anxa2 is also known to regulate osteoclastic activity [47] and participate in plasminogen activation, cell adhesion and tumor metastasis and invasion [48-51]. High levels of Anxa2 are expressed by the marrow osteoblasts and endothelial cells and subsequently Anxa2 is involved in the adhesion between HSCs and osteoblasts [52]. By blocking Anxa2 using monoclonal antibodies and competing peptides, both short- and long-term HSC repopulation are impaired [52]. Importantly, less HSCs are found in the Anxa2−/− mice [52]. In prostate cancer [51] and multiple myeloma [53], the receptor for Anxa2 is identified and Anxa2/its receptor interaction has been know as a regulator of tumor growth in the marrow. Although further study will be needed, these data suggest that HSCs also interacts with the niche through the Anxa2/Anxa2 receptor axis.
HSC mobilization from HSC niche
HSCs continue to circulate between the marrow and the bloodstream throughout life to maintain homeostasis. Under steady-state conditions, few HSCs are circulating in the peripheral blood to respond to the stress conditions such as injury and inflammation. G-CSF and granulocyte/macrophage colony-stimulating factor (GM-CSF) have been largely known as HSC mobilizing agents during hematopoietic stem cell transplantation [54]. As well as the hematopoietic growth factors, chemotherapeutic agents (cyclophosphamide), the blockade of cytokine-receptor interactions (CXCL12/CXCR4; CXCL2/CXCR2), and the degradation of adhesive connections (VLA-4/VCAM-1; CD44/HA) have been also utilized to mobilize HSCs from the marrow [54]. Interestingly, both CXCL2/CXCR2 [55] and CD44/HA [56] regulate the mobilization of HSCs by synergizing with the CXCL12/CXCR4 axis. One potential mechanism for mobilization induced by G-CSF is the activation of protease activities, including neutrophil serine proteases, metalloproteinases (MMPs) and CD26 [54], which results in the proteolytic cleavage or degradation of CXCL12 in the marrow. G-CSF may also induce mobilization by decreasing CXCL12 levels by reducing the production and the number of osteoblastic cells capable of producing CXCL12 [22, 57]. The ability to suppress CXCL12 production following the suppression of osteoblastic differentiation or numbers appears to be a common pathway for agents that mobilize HSCs including Flt3L and SCF [57]. Recent studies also revealed that G-CSF reduces monocytic cells/macrophages, leading to the inhibition of the osteoblast formation and the HSC mobilization, suggesting that monocytic cells/macrophages are involved in the osteoblastic niche formation and HSC mobilization [58-60]. On the other hand, osteoclasts have been directly involved in G-CSF-induced mobilization by activating CXCL12 degradation in conjunction with increased levels of MMP9 and cathepsin K [28]. However, it was also demonstrated that osteoclasts might not be involved in HSC mobilization [61].
AMD3100 has been approved as a HSC mobilizing agent [62]. The mechanism whereby AMD3100 stimulates mobilization appears to differ from G-CSF and other agents which reduce CXCL12 expression directly or indirectly by blocking CXCL12 interactions with its receptors. Moreover, cells mobilized by AMD3100 including CD34+ cells appear to be different than G-CSF-mobilized CD34+ cells in that they are at a different stage of the cell cycle (G1 phase vs. G0 phase) [63]. In fact, partially due to enhanced levels of CXCR4 and VLA-4, AMD3100-mobilized CD34+ cells are more potent in their ability to home when compared to G-CSF-mobilized cells [63, 64]. In addition, G-CSF suppresses the macrophage and osteoblastic niche formation to mobilize HSCs, while AMD3100 does not appear to do so [65]. Further evidence that the mode of action differs comes from studies that demonstrate that when AMD3100 and GCSF are administered together, enhanced mobilization is seen compared to either agent alone [21]. What is more, the combination of VLA-4 blockade with AMD3100 and/or G-CSF mobilized HSC dramatically better than either of these agents alone [66, 67]. Thus several mechanisms may regulate HSC mobilization such that combination of agents may prove to be superior and may yield different profiles of HSC and progenitors when engraftment is examined.
Circadian rhythm and the HSC niche
Among the more interesting findings in the past several years are observations that suggest HSC numbers in the peripheral blood is controlled by the central nervous system. In fact, CXCL12 expression by osteoblasts is regulated by signals generated locally in marrow from the sympathetic nervous system (SNS) [23]. Moreover, the number of HSC mobilized in response to cytokine stimulation depends upon the circadian rhythms [24, 68]. Reductions in CXCL12 levels are due in part to reduced Sp1 transcription factor levels because of noradrenaline secretion by sympathetic nerves in the marrow [24]. Part of the mechanism involved in the circadian ossilations of HSCs for Murine and human bone marrow CD34+ cells is dependent on the expression of the clock genes Per1, Per2, Bmal1, Cry1, Cry2, Clock and Rev-erb α [69-71]. The clock gene expression in murine whole bone marrow cells show circadian rhythmic expression, but most of clock gene expression in murine HSCs is not circadian rhythmic [70]. Yet subtle differences exist between murine and human HSCs where all but Clock shows circadian rhythms [71]. As a result, the pattern of HSC mobilization in humans is opposite to what is found in mice suggesting considerable heterogeneity between the two species [68]. At present it remains less clear whether circadian rhythms also play a major role in engraftment [72].
Circadian oscillations also appear to affect the HSC niche through actions mediated by osteoclastic bone remodeling. One molecule thought to play a major role is leptin. Leptin is known to regulate bone formation by binding to its receptor located in hypothalamus rather than acting directly on osteoblasts [73-77]. Leptin-dependent SNS promotes osteoblast proliferation through transcriptional activation of AP-1. This leads to up-regulation of c-myc and cyclin D1 [73]. Conversely, leptin-dependent SNS prevents osteoblast proliferation trough the clock genes probably by down-regulating c-myc and cyclin D1 [73]. Neuromedin U is also thought to be another downstream target of lepin signaling to inhibit osteoblast proliferation [75]. In addition, SNS induces bone loss by enhancing osteoclastic activity and osteoclast formation [76, 77]. Leptin-dependent SNS regulates bone resorption by inducing RANKL expression of osteoblasts [77]. For this sympathetic function, phosphorylation of transcription factor ATF4 in osteoblasts expressing β2-andrenergic receptors is indispensable [77]. The bone remodeling is controlled by not only the balance between the ostoblastogenesis and the osteoclastogenesis but also the osteoblastic differentiation from mescenchymal stem cells (MSCs). It has been recently demonstrated that MSCs express the clock genes, and that the potential roles of MSC in bone metabolism and HSC biology as HSC niche in response to circadian mechanisms [78]. Taken together, circadian clocks play an important role in the HSC physiology directly and indirectly by stimulating osteoblastic niche components and by targeting CXCL12 levels produced in the niche.
Signaling pathways and the HSC niche
During the last several years a number of signaling molecules including Wnt [9, 79-81], Notch [7, 82], Hedgehog [83, 84] and bone morphogenic proteins (BMPs) [9] have been implicated in HSC proliferation and quiescence. Recent work also suggests that Tie2 (expressed by HSC) and Ang-1 (expressed by osteoblast) regulate HSC quiescence in the marrow [8]. Likewise, osteopontin, a matrix glycoprotein expressed by osteoblasts has been shown to inhibit HSCs entering into replicative cycles [85, 86]. Accumulating evidence also suggests that hyper activation of mTOR signaling pathways that enhances lineage commitment of HSCs may ultimately lead to HSC exhaustion and effect which may be reversed by rapamycin [87-89].
It has also recently been appreciated that thrombopoietin (THPO) and its receptor Mpl participate in the maintenance of HSC quiescence [90, 91]. Morphologic studies demonstrated that Mpl expressing HSCs were located close to THPO expressing osteoblastic niche cells in the marrow [90]. When animals are treated with neutralizing anti-Mpl antibody, the numbers of quiescent HSCs are reduced [90]. In addition, the number of HSCs in Thpo knockout mice are less than wild-type mice and the cell cycle of Thpo−/− HSCs is accelerated [91]. Part of the proposed mechanisms for these observations is that cycling-dependent kinase inhibitors p57Kip2 and p19INK4D are down-regulated during THPO/Mpl signaling [90, 91]. Importantly, Thpo deficiency on both HSCs and niche cells results in limited engraftment after transplantation [91]. Moreover, neutralizing anti-Mpl antibody is able to open the HSC niche to facilitate the ability of transplanted-HSCs to engraft into “vacant niche” without irradiation [90]. Alternatively, the adaptor protein Lnk known as an inhibitor of THPO/Mpl signaling pathway [92], negatively regulates self-renewal and quiescence of HSCs [93]. HSCs recovered from Lnk−/− animals enter into a quiescent state relative to HSCs derived from wild-type animals, whereas HSCs from Mpl−/− animals lost a quiescent state [93]. However, when animals are generated with a double-knock out of both Mpl and Lnk, the HSC phenotype resembles wild-type HSCs in terms of quiescence [93]. It was also revealed that JAK2 was involved in regulating the balance between Lnk and Mpl in response to THPO [93]. Thus, the signaling axis THPO/Mpl/JAK2/Lnk may be one of the major pathways regulating self-renewal and quiescence of HSCs [93].
Regulation of the niche by HSCs
Crosstalk between HSCs and the HSC niche is thought to be important for hematopoiesis. However, the mechanisms of this process are largely unknown. The BMP signaling pathway is a good model for exploring the interactions between HSC and HSC niche, as BMPs affect both HSCs and niche cells. For example, BMP4 is thought to be essential for mesoderm commitment of embryonic stem cells to the hematopoietic lineage during embryogenesis [94, 95], suggesting that BMPs play important roles in deciding HSC fate. However, it has been demonstrated that the enhancement of the spindle shape N-cadherin expressing osteoblastic HSC niche size results in an increase in the HSC numbers in BMP receptor type IA knock down animals [9]. In addition, BMP4 deficient animals decrease the number of HSCs and the engraftment of normal HSCs is impaired within BMP4 deficient microenvironment [96]. These data suggest that the effect of BMPs effect on the HSC microenvironment is also relevant to how BMPs regulate hematopoiesis.
An additional mechanism whereby HSCs participate in regulation of the niche has come from work by our group where it was demonstrated that HSC are able to directly regulate bone formation [97]. Part of the mechanisms appears to be the expression of BMP2 and BMP6 by HSCs in response to signals that are received following acute bleeding or other marrow damaging agents [97]. In addition, erythropoietin (Epo) supports BMP production in HSCs through JAK2/STAT3 signaling pathway [98]. Importantly, HSCs-recovered from stressed-animals induced osteoblastic differentiation from bone marrow stromal cells in vitro and in vivo significantly more than those from control animals [97]. Yet HSCs recovered from aged animals and under conditions of experimentally induced osteoporosis, HSCs are less efficient in generating a BMP response following acute blood loss [97]. These findings suggest that HSCs do not only rest passively in their niche but also directly participate in development of an osteoblastic niche [97]. However, it is less clear if BMP production by HSCs regulates endothelial niche expansion.
Deeply quiescent HSCs
Hypoxia is thought to contribute to the ability of HSC to become quiescence (or dormant) and self-renewal [99]. It has been also demonstrated that oxidative stress reverses HSC dormancy [100, 101]. In the marrow, the osteoblastic niche is thought to represent a lower O2 environment compared with the vascular niche [29]. For this reason, it has been hypothesized that the osteoblastic niche maintains quiescence and self-renewal of HSCs, while the vascular niche may regulate the proliferation and differentiation of HSCs [29]. In other words, HSCs reside near the osteoblastic nice (dormant HSCs) are in slow-dividing state and maintain the cell compartment in the marrow, whereas HSCs near the vascular niche (active HSCs) are in cell-cycling state and are ready to egress from the marrow in response to stress including infection, injury or chemotherapy. It has also been recently revealed that not only active HSCs respond to stress to maintain homeostasis but so do dormant HSCs [102]. The switch from an actively cycling cell to dormancy and back to an active state appears to be quite rapid as would be required in order to respond to physiologic and non-physiologic stressors on the hematopoietic system [102]. These data support the idea that osteoblastic niche and vascular niche, including perivascular cells [13] and nestin-expressing mesenchymal stem cells [17] that are located near the endothelial cells, cooperate with each other in constructing and maintaining the HSC niche.
Future Challenges
Several lines of evidence has demonstrated that cells of the osteoblastic lineage participates in the development of the HSC niche in murine [7-9, 12, 24, 52, 68] and human models [6] (Figure 1). More recent findings have expanded these observations suggesting that the central nervous system is involved in the regulation of HSC movement in and out of the niche with direct effects on osteoblast function [24, 68] (Figure 1). Yet the precise location of the HSC niche and whether it is endosteal-osteoblastic or endothelial-vascular or both remain unclear. It has recently been suggested that endochondral ossification is essential for osteoblastic niche formation [103], and that pre-osteoblasts may play an important role in normal hematopoiesis [104]. However, which cell in the osteoblastic lineage is directly responsible for establishing/participating in the niche, and what is its relationship to other cells in the marrow are unclear. Recent advances in in vivo real-time imaging have begun to address these issues [105-107]. However until actual markers emerge that denote the “HSC” niche as the niche this area is likely to serve as a source for future controversy.
Figure 1. The model of ecological hematopoietic stem cell niche.
Hematopoietic stem cell (HSC) homing, maintenance and differentiation depend upon their specific microenvironment (or niche). In the bone marrow there are at least two major HSC niches. The vascular niche, which is a gateway for HSCs between the periphery and marrow that likely facilitates the proliferation and differentiation of HSCs, while the osteoblastic niche likely maintains self-renewal and quiescence of HSCs. Osteoclasts are also known to participate in niche function partially by regulating HSC mobilization. In addition, there are growing indications that other types of cells including fibroblasts, CXCL12-expressing reticular cells, nestin-expressing mesenchymal stem cell (MSC) and adipocytes, also serve as the HSC niche in the marrow. Homing of stem cells is mainly regulated thorough CXCL12 expressed by the HSC niche. Interestingly, circadian rhythm is involved in these relationships by regulating CXCL12 expression in the marrow. As such, variety of the marrow cells may dynamically affect each other and participate in hematopoiesis.
Just as important are observations that not all niches are the same. Several investigators have suggested that niches that regulate HSC quiescence in marrow may differ in either location or quality from niches that participate in HSC maturation events (Figure 1). For example, adipocytes negatively regulate hematopoiesis in the marrow [15, 108] by up-regulating neuropilin-1 [108] and nestin positive MSCs are involved in the HSC niche formation [17]. Adding further complexity to our understanding is that HSCs probably migrate between niches at low levels that can fluctuate in response to physiologic stimuli and in response to circadian rhythms. Nor is it known how or when the endosteal-osteoblastic or endothelial-vascular niches, or any other HSC niches, cross-talk to one another. It also remains uncertain whether there are times or conditions in which one niche takes priority over the other. If so, what are the signals that participate in setting priority?
Stem cell transplantation is the treatment of choice where restoration of normal hematopoiesis is required including several blood neoplasms, immune deficiencies and during the treatment of solid tumors following chemotherapy [109]. Because of limited number of HSCs and the potential for delayed engraftment [110], it is critical to develop improved techniques for ex vivo HSC expansion while maintaining HSC stemness. However, most of the ex vivo HSC expansion protocols lead to exhaustion of the critical target cells [110]. Interestingly, it has been appreciated that the niche itself may be targetable - such that manipulating the microenvironment for HSCs (e.g. expanding HSC niche by PTH treatment) may support HSC engraftment during transplantation [111]. Just as critical is the necessity for a greater understanding of the HSC-niche interactions to facilitate targeting of the region to improve mobilization of HSCs out of the niche and to facilitate enhancements in engrafting capabilities.
Adding another layer of complexity to our understanding of the niche is the notion that hematologic malignancies [112, 113] and solid tumor [114] target the HSC niche. Intriguingly, metastatic prostate cancer cells compete for occupancy of the endosteal niche with HSCs [114]. Once there, the occult malignant cells may become dormant, parasitizing the molecular signals that regulate HSCs dormancy. Under these conditions, tumor cells may be able to escape immune surveillance mechanisms by resembling HSCs in the niche and evade conventional radiation and/or chemotherapy that target rapidly dividing cells. Moreover, the effects of the bone marrow microenvironment on the development of malignancy have been revealed. Myeloproliferative syndromes (MPS) develope in the animals when the retinoic acid receptor γ (RARγ) [115] or retinoblastoma protein (RB) [116] was deleted from the bone marrow microenvironment. However, RARγ- or RB-deficient HSCs function normally in the wild-type microenvironment [115, 116]. Indeed, whether malignancies create their own specific niche or use normal HSC niche as their niche remains an open question. A recent study by Raaijmakers et al has begun to address this question [104]. Disfunctional osteoprogenitor cells were established by deleting Dicer1 which lead to secondary hematopoietic malignancies including myelodysplastic syndrome, myeloid sarcoma and acute myelogenous leukemia [104]. Strikingly, Raaijmakers et al also provide the linkage between their findings and human disease (e.g. Shwachman–Diamond–Bodian syndrome) [104]. Although further study is clearly needed, this intriguing concept could suggest that the HSC niche serve as a new potential target therapy for malignancies [111, 113].
Over the past five years it is clear that a variety of the bone marrow components are intricately intertwined and support many aspects of hematopoiesis in a collaborative fashion. Osteoblasts have been shown to be a major compartment of HSC niche (Table 1), although clearly not the only cell that has ‘niche’ capacity. Although our knowledge of HSC niche has made dramatically progress, critical questions are still unanswered since our previous review [20]. i) Does only one single component of the marrow cells serve as HSC niche? - Which is the HSC niche (e.g. osteoblasts vs endothelial cells)? ii) If there are several different HSC niche components, how do they interact? iii) How does the HSC niche balance self-renewal and the differentiation of HSCs? iv) Is the HSC niche involved in tumorigenesis? - Are the normal HSC niche and leukemia/cancer niche different? Although further studies will be needed, these questions have begun to be addressed. If successful, the HSC niche could be a potential target during the bone marrow transplantation as well as the leukemia/cancer treatment.
Table 1. Milestones of the osteoblastic niche.
Year | Authors | Major discoveries associated with osteoblastic niche | Reference |
---|---|---|---|
1972 | Patt HM et al. | Osteogenesis and hematopoiesis | 3 |
1975 | Lord BI et al. | Endosteal surface and hematopoiesis | 4 |
1978 | Gong JK et al. | Endosteal surface and hematopoiesis | 5 |
1978 | Schofield R | Stem cell niche concept | 2 |
1994 | Taichman RS &Emerson SG | Human osteoblast and hematopoiesis | 6 |
2003 | Calvi LM et al. | PTH | 7 |
Zhang J et al. | BMP | 9 | |
2004 | Arai F et al. | Tie2/Ang1 | 8 |
Visnjic D et al. | Osteoblast KO mouse | 11 | |
2005 | Nilsson SK et al. | Osteopontin | 86 |
Stier S et al. | Osteopontin | 85 | |
2006 | Adams GB et al. | Calcium-sensing receptor | 34 |
Kollet O et al. | Osteoclast | 28 | |
2007 | Zhu J et al. | B-cell | 10 |
Qian H et al. | Thrombopoietin | 91 | |
Yoshihara T et al. | Thrombopoietin | 90 | |
Walkley CR et al. | Myeloproliferative syndrome and retinoic scid receptor γ | 115 | |
Walkley CR et al. | Myeloproliferative syndrome and retinoblastma protein | 116 | |
2008 | Mendez-Ferrer S et al. | Circadian oscillations | 24 |
2009 | Adams GB et al. | Guanine-nucleotide-binding protein stimulatory α subunit | 35 |
Lo Celso C et al. | In vivo real-time imaging | 105 | |
Xie Y et al. | In vivo real-time imaging | 106 | |
2010 | Raaijmakers MH et al. | Leukemogenesis | 104 |
Winkler IG et al. | Macrophage and HSC mobilization | 58 | |
2011 | Christopher MJ et al. | Monocytic cells and HSC mobilization | 59 |
Chow A et al. | Macrophage and HSC mobilization | 60 | |
Shiozawa Y et al. | Niche competition between HSC and metastatic prostate cancer | 114 |
Acknowledgements
This work was directly supported by a Pediatric Oncology Research Fellowship (Y.S), the Department of Defense (Y.S. and R.S.T.) and National Institutes of health (DK082481 R.S.T., DE020721 R.S.T., 1RC1DE020721 R.S.T., CA141426 R.S.T., CA093900 R.S.T. and CA163124, Y.S. and R.S.T.), and the Prostate Cancer Foundation (Y.S. and R.S.T.).
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Reference Lists
- [1].Tavian M, Peault B. Embryonic development of the human hematopoietic system. Int J Dev Biol. 2005;49:243–250. doi: 10.1387/ijdb.041957mt. [DOI] [PubMed] [Google Scholar]
- [2].Schofield R. The relationship between the spleen colony-forming cell and the haemopoietic stem cell. Blood Cells. 1978;4:7–25. [PubMed] [Google Scholar]
- [3].Patt HM, Maloney MA. Bone formation and resorption as a requirement for marrow development. Proc Soc Exp Biol Med. 1972;140:205–207. doi: 10.3181/00379727-140-36426. [DOI] [PubMed] [Google Scholar]
- [4].Lord BI, Hendry JH. The distribution of haemopoietic colony-forming units in the mouse femur, and its modification by x rays. Br J Radiol. 1972;45:110–115. doi: 10.1259/0007-1285-45-530-110. [DOI] [PubMed] [Google Scholar]
- [5].Gong JK. Endosteal marrow: a rich source of hematopoietic stem cells. Science. 1978;199:1443–1445. doi: 10.1126/science.75570. [DOI] [PubMed] [Google Scholar]
- [6].Taichman RS, Emerson SG. Human osteoblasts support hematopoiesis through the production of granulocyte colony-stimulating factor. J Exp Med. 1994;179:1677–1682. doi: 10.1084/jem.179.5.1677. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [7].Calvi LM, Adams GB, Weibrecht KW, et al. Osteoblastic cells regulate the haematopoietic stem cell niche. Nature. 2003;425:841–846. doi: 10.1038/nature02040. [DOI] [PubMed] [Google Scholar]
- [8].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. doi: 10.1016/j.cell.2004.07.004. [DOI] [PubMed] [Google Scholar]
- [9].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. doi: 10.1038/nature02041. [DOI] [PubMed] [Google Scholar]
- [10].Zhu J, Garrett R, Jung Y, et al. Osteoblasts support B-lymphocyte commitment and differentiation from hematopoietic stem cells. Blood. 2007;109:3706–3712. doi: 10.1182/blood-2006-08-041384. [DOI] [PubMed] [Google Scholar]
- [11].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. doi: 10.1182/blood-2003-11-4011. [DOI] [PubMed] [Google Scholar]
- [12].Kiel MJ, Yilmaz OH, Iwashita T, 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. doi: 10.1016/j.cell.2005.05.026. [DOI] [PubMed] [Google Scholar]
- [13].Ding L, Saunders TL, Enikolopov G, Morrison SJ. Endothelial and perivascular cells maintain haematopoietic stem cells. Nature. 2012;481:457–462. doi: 10.1038/nature10783. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [14].Sugiyama T, Kohara H, Noda M, Nagasawa T. Maintenance of the hematopoietic stem cell pool by CXCL12-CXCR4 chemokine signaling in bone marrow stromal cell niches. Immunity. 2006;25:977–988. doi: 10.1016/j.immuni.2006.10.016. [DOI] [PubMed] [Google Scholar]
- [15].Naveiras O, Nardi V, Wenzel PL, Hauschka PV, Fahey F, Daley GQ. Bone-marrow adipocytes as negative regulators of the haematopoietic microenvironment. Nature. 2009;460:259–263. doi: 10.1038/nature08099. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [16].Morikawa S, Mabuchi Y, Kubota Y, et al. Prospective identification, isolation, and systemic transplantation of multipotent mesenchymal stem cells in murine bone marrow. J Exp Med. 2009;206:2483–2496. doi: 10.1084/jem.20091046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [17].Mendez-Ferrer S, Michurina TV, Ferraro F, et al. Mesenchymal and haematopoietic stem cells form a unique bone marrow niche. Nature. 2010;466:829–834. doi: 10.1038/nature09262. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [18].Watt SM, Forde SP. The central role of the chemokine receptor, CXCR4, in haemopoietic stem cell transplantation: will CXCR4 antagonists contribute to the treatment of blood disorders? Vox Sang. 2008;94:18–32. doi: 10.1111/j.1423-0410.2007.00995.x. [DOI] [PubMed] [Google Scholar]
- [19].Peled A, Petit I, Kollet O, et al. Dependence of human stem cell engraftment and repopulation of NOD/SCID mice on CXCR4. Science. 1999;283:845–848. doi: 10.1126/science.283.5403.845. [DOI] [PubMed] [Google Scholar]
- [20].Taichman RS. Blood and bone: two tissues whose fates are intertwined to create the hematopoietic stem-cell niche. Blood. 2005;105:2631–2639. doi: 10.1182/blood-2004-06-2480. [DOI] [PubMed] [Google Scholar]
- [21].Broxmeyer HE, Orschell CM, Clapp DW, et al. Rapid mobilization of murine and human hematopoietic stem and progenitor cells with AMD3100, a CXCR4 antagonist. J Exp Med. 2005;201:1307–1318. doi: 10.1084/jem.20041385. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [22].Semerad CL, Christopher MJ, Liu F, et al. G-CSF potently inhibits osteoblast activity and CXCL12 mRNA expression in the bone marrow. Blood. 2005;106:3020–3027. doi: 10.1182/blood-2004-01-0272. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [23].Katayama Y, Battista M, Kao WM, et al. Signals from the sympathetic nervous system regulate hematopoietic stem cell egress from bone marrow. Cell. 2006;124:407–421. doi: 10.1016/j.cell.2005.10.041. [DOI] [PubMed] [Google Scholar]
- [24].Mendez-Ferrer S, Lucas D, Battista M, Frenette PS. Haematopoietic stem cell release is regulated by circadian oscillations. Nature. 2008;452:442–447. doi: 10.1038/nature06685. [DOI] [PubMed] [Google Scholar]
- [25].Jung Y, Wang J, Schneider A, et al. Regulation of SDF-1 (CXCL12) production by osteoblasts; a possible mechanism for stem cell homing. Bone. 2006;38:497–508. doi: 10.1016/j.bone.2005.10.003. [DOI] [PubMed] [Google Scholar]
- [26].Ponomaryov T, Peled A, Petit I, et al. Induction of the chemokine stromal-derived factor-1 following DNA damage improves human stem cell function. J Clin Invest. 2000;106:1331–1339. doi: 10.1172/JCI10329. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [27].Dar A, Goichberg P, Shinder V, et al. Chemokine receptor CXCR4-dependent internalization and resecretion of functional chemokine SDF-1 by bone marrow endothelial and stromal cells. Nat Immunol. 2005;6:1038–1046. doi: 10.1038/ni1251. [DOI] [PubMed] [Google Scholar]
- [28].Kollet O, Dar A, Shivtiel S, et al. Osteoclasts degrade endosteal components and promote mobilization of hematopoietic progenitor cells. Nat Med. 2006;12:657–664. doi: 10.1038/nm1417. [DOI] [PubMed] [Google Scholar]
- [29].Yin T, Li L. The stem cell niches in bone. J Clin Invest. 2006;116:1195–1201. doi: 10.1172/JCI28568. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [30].Ceradini DJ, Kulkarni AR, Callaghan MJ, et al. Progenitor cell trafficking is regulated by hypoxic gradients through HIF-1 induction of SDF-1. Nat Med. 2004;10:858–864. doi: 10.1038/nm1075. [DOI] [PubMed] [Google Scholar]
- [31].Staller P, Sulitkova J, Lisztwan J, Moch H, Oakeley EJ, Krek W. Chemokine receptor CXCR4 down-regulated by von Hippel-Lindau tumour suppressor pVHL. Nature. 2003;425:307–311. doi: 10.1038/nature01874. [DOI] [PubMed] [Google Scholar]
- [32].Christopherson KW, 2nd, Hangoc G, Mantel CR, Broxmeyer HE. Modulation of hematopoietic stem cell homing and engraftment by CD26. Science. 2004;305:1000–1003. doi: 10.1126/science.1097071. [DOI] [PubMed] [Google Scholar]
- [33].Schwaiger E, Klaus C, Matheeussen V, et al. Dipeptidyl peptidase IV (DPPIV/CD26) inhibition does not improve engraftment of unfractionated syngeneic or allogeneic bone marrow after nonmyeloablative conditioning. Exp Hematol. 2012;40:97–106. doi: 10.1016/j.exphem.2011.10.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [34].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. doi: 10.1038/nature04247. [DOI] [PubMed] [Google Scholar]
- [35].Adams GB, Alley IR, Chung UI, et al. Haematopoietic stem cells depend on Galpha(s)-mediated signalling to engraft bone marrow. Nature. 2009;459:103–107. doi: 10.1038/nature07859. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [36].Abkowitz JL, Chen J. Studies of c-Mpl function distinguish the replication of hematopoietic stem cells from the expansion of differentiating clones. Blood. 2007;109:5186–5190. doi: 10.1182/blood-2006-08-044503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [37].Chen J, Larochelle A, Fricker S, Bridger G, Dunbar CE, Abkowitz JL. Mobilization as a preparative regimen for hematopoietic stem cell transplantation. Blood. 2006;107:3764–3771. doi: 10.1182/blood-2005-09-3593. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [38].Czechowicz A, Kraft D, Weissman IL, Bhattacharya D. Efficient transplantation via antibody-based clearance of hematopoietic stem cell niches. Science. 2007;318:1296–1299. doi: 10.1126/science.1149726. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [39].Li P, Zon LI. Resolving the controversy about N-cadherin and hematopoietic stem cells. Cell Stem Cell. 6:199–202. doi: 10.1016/j.stem.2010.02.007. [DOI] [PubMed] [Google Scholar]
- [40].Kiel MJ, Radice GL, Morrison SJ. Lack of evidence that hematopoietic stem cells depend on N-cadherin-mediated adhesion to osteoblasts for their maintenance. Cell Stem Cell. 2007;1:204–217. doi: 10.1016/j.stem.2007.06.001. [DOI] [PubMed] [Google Scholar]
- [41].Haug JS, He XC, Grindley JC, et al. N-cadherin expression level distinguishes reserved versus primed states of hematopoietic stem cells. Cell Stem Cell. 2008;2:367–379. doi: 10.1016/j.stem.2008.01.017. [DOI] [PubMed] [Google Scholar]
- [42].Kiel MJ, Acar M, Radice GL, Morrison SJ. Hematopoietic stem cells do not depend on N-cadherin to regulate their maintenance. Cell Stem Cell. 2009;4:170–179. doi: 10.1016/j.stem.2008.10.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [43].Greenbaum AM, Revollo LD, Woloszynek JR, Civitelli R, Link DC. N-cadherin in osteolineage cells is not required for maintenance of hematopoietic stem cells. Blood. 2012 doi: 10.1182/blood-2011-09-377457. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [44].Jacobsen K, Kravitz J, Kincade PW, Osmond DG. Adhesion receptors on bone marrow stromal cells: in vivo expression of vascular cell adhesion molecule-1 by reticular cells and sinusoidal endothelium in normal and gamma-irradiated mice. Blood. 1996;87:73–82. [PubMed] [Google Scholar]
- [45].Priestley GV, Scott LM, Ulyanova T, Papayannopoulou T. Lack of alpha4 integrin expression in stem cells restricts competitive function and self-renewal activity. Blood. 2006;107:2959–2967. doi: 10.1182/blood-2005-07-2670. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [46].Jung Y, Shiozawa Y, Wang J, et al. Annexin-2 is a regulator of stromal cell-derived factor-1/CXCL12 function in the hematopoietic stem cell endosteal niche. Exp Hematol. 2011;39:151–166. e151. doi: 10.1016/j.exphem.2010.11.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [47].Takahashi S, Reddy SV, Chirgwin JM, et al. Cloning and identification of annexin II as an autocrine/paracrine factor that increases osteoclast formation and bone resorption. J Biol Chem. 1994;269:28696–28701. [PubMed] [Google Scholar]
- [48].Siever DA, Erickson HP. Extracellular annexin II. Int J Biochem Cell Biol. 1997;29:1219–1223. doi: 10.1016/s1357-2725(97)00057-5. [DOI] [PubMed] [Google Scholar]
- [49].Kwon M, MacLeod TJ, Zhang Y, Waisman DM. S100A10, annexin A2, and annexin a2 heterotetramer as candidate plasminogen receptors. Front Biosci. 2005;10:300–325. doi: 10.2741/1529. [DOI] [PubMed] [Google Scholar]
- [50].Kang HM, Choi KS, Kassam G, Fitzpatrick SL, Kwon M, Waisman DM. Role of annexin II tetramer in plasminogen activation. Trends Cardiovasc Med. 1999;9:92–102. doi: 10.1016/s1050-1738(99)00012-2. [DOI] [PubMed] [Google Scholar]
- [51].Shiozawa Y, Havens AM, Jung Y, et al. Annexin II/annexin II receptor axis regulates adhesion, migration, homing, and growth of prostate cancer. J Cell Biochem. 2008;105:370–380. doi: 10.1002/jcb.21835. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [52].Jung Y, Wang J, Song J, et al. Annexin II expressed by osteoblasts and endothelial cells regulates stem cell adhesion, homing, and engraftment following transplantation. Blood. 2007;110:82–90. doi: 10.1182/blood-2006-05-021352. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [53].D’Souza S, Kurihara N, Shiozawa Y, et al. Annexin II interactions with the annexin II receptor enhance multiple myeloma cell adhesion and growth in the bone marrow microenvironment. Blood. 2012;119:1888–1896. doi: 10.1182/blood-2011-11-393348. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [54].Nervi B, Link DC, DiPersio JF. Cytokines and hematopoietic stem cell mobilization. J Cell Biochem. 2006;99:690–705. doi: 10.1002/jcb.21043. [DOI] [PubMed] [Google Scholar]
- [55].Eash KJ, Greenbaum AM, Gopalan PK, Link DC. CXCR2 and CXCR4 antagonistically regulate neutrophil trafficking from murine bone marrow. J Clin Invest. 2010;120:2423–2431. doi: 10.1172/JCI41649. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [56].Avigdor A, Goichberg P, Shivtiel S, et al. CD44 and hyaluronic acid cooperate with SDF-1 in the trafficking of human CD34+ stem/progenitor cells to bone marrow. Blood. 2004;103:2981–2989. doi: 10.1182/blood-2003-10-3611. [DOI] [PubMed] [Google Scholar]
- [57].Christopher MJ, Liu F, Hilton MJ, Long F, Link DC. Suppression of CXCL12 production by bone marrow osteoblasts is a common and critical pathway for cytokine-induced mobilization. Blood. 2009;114:1331–1339. doi: 10.1182/blood-2008-10-184754. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [58].Winkler IG, Sims NA, Pettit AR, et al. Bone marrow macrophages maintain hematopoietic stem cell (HSC) niches and their depletion mobilizes HSCs. Blood. 2010;116:4815–4828. doi: 10.1182/blood-2009-11-253534. [DOI] [PubMed] [Google Scholar]
- [59].Christopher MJ, Rao M, Liu F, Woloszynek JR, Link DC. Expression of the G-CSF receptor in monocytic cells is sufficient to mediate hematopoietic progenitor mobilization by G-CSF in mice. J Exp Med. 2011;208:251–260. doi: 10.1084/jem.20101700. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [60].Chow A, Lucas D, Hidalgo A, et al. Bone marrow CD169+ macrophages promote the retention of hematopoietic stem and progenitor cells in the mesenchymal stem cell niche. J Exp Med. 2011;208:261–271. doi: 10.1084/jem.20101688. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [61].Miyamoto K, Yoshida S, Kawasumi M, et al. Osteoclasts are dispensable for hematopoietic stem cell maintenance and mobilization. J Exp Med. 2011;208:2175–2181. doi: 10.1084/jem.20101890. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [62].Uy GL, Rettig MP, Cashen AF. Plerixafor, a CXCR4 antagonist for the mobilization of hematopoietic stem cells. Expert Opin Biol Ther. 2008;8:1797–1804. doi: 10.1517/14712598.8.11.1797. [DOI] [PubMed] [Google Scholar]
- [63].Larochelle A, Krouse A, Metzger M, et al. AMD3100 mobilizes hematopoietic stem cells with long-term repopulating capacity in nonhuman primates. Blood. 2006;107:3772–3778. doi: 10.1182/blood-2005-09-3592. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [64].Hess DA, Bonde J, Craft TP, et al. Human progenitor cells rapidly mobilized by AMD3100 repopulate NOD/SCID mice with increased frequency in comparison to cells from the same donor mobilized by granulocyte colony stimulating factor. Biol Blood Marrow Transplant. 2007;13:398–411. doi: 10.1016/j.bbmt.2006.12.445. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [65].Winkler IG, Pettit AR, Raggatt LJ, et al. Hematopoietic stem cell mobilizing agents G-CSF, cyclophosphamide or AMD3100 have distinct mechanisms of action on bone marrow HSC niches and bone formation. Leukemia. 2012 doi: 10.1038/leu.2012.17. In Press. [DOI] [PubMed] [Google Scholar]
- [66].Ramirez P, Rettig MP, Uy GL, et al. BIO5192, a small molecule inhibitor of VLA-4, mobilizes hematopoietic stem and progenitor cells. Blood. 2009;114:1340–1343. doi: 10.1182/blood-2008-10-184721. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [67].Bonig H, Watts KL, Chang KH, Kiem HP, Papayannopoulou T. Concurrent blockade of alpha4-integrin and CXCR4 in hematopoietic stem/progenitor cell mobilization. Stem Cells. 2009;27:836–837. doi: 10.1002/stem.9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [68].Lucas D, Battista M, Shi PA, Isola L, Frenette PS. Mobilized hematopoietic stem cell yield depends on species-specific circadian timing. Cell Stem Cell. 2008;3:364–366. doi: 10.1016/j.stem.2008.09.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [69].Tsinkalovsky O, Rosenlund B, Laerum OD, Eiken HG. Clock gene expression in purified mouse hematopoietic stem cells. Exp Hematol. 2005;33:100–107. doi: 10.1016/j.exphem.2004.09.007. [DOI] [PubMed] [Google Scholar]
- [70].Tsinkalovsky O, Filipski E, Rosenlund B, et al. Circadian expression of clock genes in purified hematopoietic stem cells is developmentally regulated in mouse bone marrow. Exp Hematol. 2006;34:1249–1261. doi: 10.1016/j.exphem.2006.05.008. [DOI] [PubMed] [Google Scholar]
- [71].Tsinkalovsky O, Smaaland R, Rosenlund B, et al. Circadian variations in clock gene expression of human bone marrow CD34+ cells. J Biol Rhythms. 2007;22:140–150. doi: 10.1177/0748730406299078. [DOI] [PubMed] [Google Scholar]
- [72].D’Hondt L, McAuliffe C, Damon J, et al. Circadian variations of bone marrow engraftability. J Cell Physiol. 2004;200:63–70. doi: 10.1002/jcp.20032. [DOI] [PubMed] [Google Scholar]
- [73].Fu L, Patel MS, Bradley A, Wagner EF, Karsenty G. The molecular clock mediates leptin-regulated bone formation. Cell. 2005;122:803–815. doi: 10.1016/j.cell.2005.06.028. [DOI] [PubMed] [Google Scholar]
- [74].Takeda S, Elefteriou F, Levasseur R, et al. Leptin regulates bone formation via the sympathetic nervous system. Cell. 2002;111:305–317. doi: 10.1016/s0092-8674(02)01049-8. [DOI] [PubMed] [Google Scholar]
- [75].Sato S, Hanada R, Kimura A, et al. Central control of bone remodeling by neuromedin U. Nat Med. 2007;13:1234–1240. doi: 10.1038/nm1640. [DOI] [PubMed] [Google Scholar]
- [76].Kondo H, Nifuji A, Takeda S, et al. Unloading induces osteoblastic cell suppression and osteoclastic cell activation to lead to bone loss via sympathetic nervous system. J Biol Chem. 2005;280:30192–30200. doi: 10.1074/jbc.M504179200. [DOI] [PubMed] [Google Scholar]
- [77].Elefteriou F, Ahn JD, Takeda S, et al. Leptin regulation of bone resorption by the sympathetic nervous system and CART. Nature. 2005;434:514–520. doi: 10.1038/nature03398. [DOI] [PubMed] [Google Scholar]
- [78].Wu X, Yu G, Parks H, et al. Circadian mechanisms in murine and human bone marrow mesenchymal stem cells following dexamethasone exposure. Bone. 2008;42:861–870. doi: 10.1016/j.bone.2007.12.226. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [79].Nemeth MJ, Topol L, Anderson SM, Yang Y, Bodine DM. Wnt5a inhibits canonical Wnt signaling in hematopoietic stem cells and enhances repopulation. Proc Natl Acad Sci U S A. 2007;104:15436–15441. doi: 10.1073/pnas.0704747104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [80].Fleming HE, Janzen V, Lo Celso C, et al. Wnt signaling in the niche enforces hematopoietic stem cell quiescence and is necessary to preserve self-renewal in vivo. Cell Stem Cell. 2008;2:274–283. doi: 10.1016/j.stem.2008.01.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [81].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. doi: 10.1038/nature01593. [DOI] [PubMed] [Google Scholar]
- [82].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. doi: 10.1038/ni1164. [DOI] [PubMed] [Google Scholar]
- [83].Gao J, Graves S, Koch U, et al. Hedgehog signaling is dispensable for adult hematopoietic stem cell function. Cell Stem Cell. 2009;4:548–558. doi: 10.1016/j.stem.2009.03.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [84].Hofmann I, Stover EH, Cullen DE, et al. Hedgehog signaling is dispensable for adult murine hematopoietic stem cell function and hematopoiesis. Cell Stem Cell. 2009;4:559–567. doi: 10.1016/j.stem.2009.03.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [85].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. doi: 10.1084/jem.20041992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [86].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. doi: 10.1182/blood-2004-11-4422. [DOI] [PubMed] [Google Scholar]
- [87].Yilmaz OH, Valdez R, Theisen BK, et al. Pten dependence distinguishes haematopoietic stem cells from leukaemia-initiating cells. Nature. 2006;441:475–482. doi: 10.1038/nature04703. [DOI] [PubMed] [Google Scholar]
- [88].Huang J, Zhang Y, Bersenev A, et al. Pivotal role for glycogen synthase kinase-3 in hematopoietic stem cell homeostasis in mice. J Clin Invest. 2009;119:3519–3529. doi: 10.1172/JCI40572. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [89].Campbell TB, Basu S, Hangoc G, Tao W, Broxmeyer HE. Overexpression of Rheb2 enhances mouse hematopoietic progenitor cell growth while impairing stem cell repopulation. Blood. 2009;114:3392–3401. doi: 10.1182/blood-2008-12-195214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [90].Yoshihara H, Arai F, Hosokawa K, et al. Thrombopoietin/MPL signaling regulates hematopoietic stem cell quiescence and interaction with the osteoblastic niche. Cell Stem Cell. 2007;1:685–697. doi: 10.1016/j.stem.2007.10.020. [DOI] [PubMed] [Google Scholar]
- [91].Qian H, Buza-Vidas N, Hyland CD, et al. Critical role of thrombopoietin in maintaining adult quiescent hematopoietic stem cells. Cell Stem Cell. 2007;1:671–684. doi: 10.1016/j.stem.2007.10.008. [DOI] [PubMed] [Google Scholar]
- [92].Tong W, Lodish HF. Lnk inhibits Tpo-mpl signaling and Tpo-mediated megakaryocytopoiesis. J Exp Med. 2004;200:569–580. doi: 10.1084/jem.20040762. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [93].Bersenev A, Wu C, Balcerek J, Tong W. Lnk controls mouse hematopoietic stem cell self-renewal and quiescence through direct interactions with JAK2. J Clin Invest. 2008;118:2832–2844. doi: 10.1172/JCI35808. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [94].Nostro MC, Cheng X, Keller GM, Gadue P. Wnt, activin, and BMP signaling regulate distinct stages in the developmental pathway from embryonic stem cells to blood. Cell Stem Cell. 2008;2:60–71. doi: 10.1016/j.stem.2007.10.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [95].Lengerke C, Schmitt S, Bowman TV, et al. BMP and Wnt specify hematopoietic fate by activation of the Cdx-Hox pathway. Cell Stem Cell. 2008;2:72–82. doi: 10.1016/j.stem.2007.10.022. [DOI] [PubMed] [Google Scholar]
- [96].Goldman DC, Bailey AS, Pfaffle DL, Al Masri A, Christian JL, Fleming WH. BMP4 regulates the hematopoietic stem cell niche. Blood. 2009;114:4393–4401. doi: 10.1182/blood-2009-02-206433. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [97].Jung Y, Song J, Shiozawa Y, et al. Hematopoietic stem cells regulate mesenchymal stromal cell induction into osteoblasts thereby participating in the formation of the stem cell niche. Stem Cells. 2008;26:2042–2051. doi: 10.1634/stemcells.2008-0149. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [98].Shiozawa Y, Jung Y, Ziegler AM, et al. Erythropoietin Couples Hematopoiesis with Bone Formation. PLoS One. 2010;5:e10853. doi: 10.1371/journal.pone.0010853. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [99].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. doi: 10.1073/pnas.0701152104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [100].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. doi: 10.1038/nm1388. [DOI] [PubMed] [Google Scholar]
- [101].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. doi: 10.1038/nature02989. [DOI] [PubMed] [Google Scholar]
- [102].Wilson A, Laurenti E, Oser G, et al. Hematopoietic stem cells reversibly switch from dormancy to self-renewal during homeostasis and repair. Cell. 2008;135:1118–1129. doi: 10.1016/j.cell.2008.10.048. [DOI] [PubMed] [Google Scholar]
- [103].Chan CK, Chen CC, Luppen CA, et al. Endochondral ossification is required for haematopoietic stem-cell niche formation. Nature. 2009;457:490–494. doi: 10.1038/nature07547. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [104].Raaijmakers MH, Mukherjee S, Guo S, et al. Bone progenitor dysfunction induces myelodysplasia and secondary leukaemia. Nature. 464:852–857. doi: 10.1038/nature08851. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [105].Lo Celso C, Fleming HE, Wu JW, et al. Live-animal tracking of individual haematopoietic stem/progenitor cells in their niche. Nature. 2009;457:92–96. doi: 10.1038/nature07434. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [106].Xie Y, Yin T, Wiegraebe W, et al. Detection of functional haematopoietic stem cell niche using real-time imaging. Nature. 2009;457:97–101. doi: 10.1038/nature07639. [DOI] [PubMed] [Google Scholar]
- [107].Fujisaki J, Wu J, Carlson AL, et al. In vivo imaging of Treg cells providing immune privilege to the haematopoietic stem-cell niche. Nature. 2011;474:216–219. doi: 10.1038/nature10160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [108].Chitteti BR, Cheng YH, Poteat B, et al. Impact of interactions of cellular components of the bone marrow microenvironment on hematopoietic stem and progenitor cell function. Blood. 115:3239–3248. doi: 10.1182/blood-2009-09-246173. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [109].Myers KC, Davies SM. Hematopoietic stem cell transplantation for bone marrow failure syndromes in children. Biol Blood Marrow Transplant. 2009;15:279–292. doi: 10.1016/j.bbmt.2008.11.037. [DOI] [PubMed] [Google Scholar]
- [110].Hofmeister CC, Zhang J, Knight KL, Le P, Stiff PJ. Ex vivo expansion of umbilical cord blood stem cells for transplantation: growing knowledge from the hematopoietic niche. Bone Marrow Transplant. 2007;39:11–23. doi: 10.1038/sj.bmt.1705538. [DOI] [PubMed] [Google Scholar]
- [111].Adams GB, Scadden DT. A niche opportunity for stem cell therapeutics. Gene Ther. 2008;15:96–99. doi: 10.1038/sj.gt.3303063. [DOI] [PubMed] [Google Scholar]
- [112].Lane SW, Scadden DT, Gilliland DG. The leukemic stem cell niche: current concepts and therapeutic opportunities. Blood. 2009;114:1150–1157. doi: 10.1182/blood-2009-01-202606. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [113].Shiozawa Y, Havens AM, Pienta KJ, Taichman RS. The bone marrow niche: habitat to hematopoietic and mesenchymal stem cells, and unwitting host to molecular parasites. Leukemia. 2008;22:941–950. doi: 10.1038/leu.2008.48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [114].Shiozawa Y, Pedersen EA, Havens AM, et al. Human prostate cancer metastases target the hematopoietic stem cell niche to establish footholds in mouse bone marrow. J Clin Invest. 2011;121:1298–1312. doi: 10.1172/JCI43414. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [115].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. doi: 10.1016/j.cell.2007.05.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [116].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. doi: 10.1016/j.cell.2007.03.055. [DOI] [PMC free article] [PubMed] [Google Scholar]