The hematopoietic stem cell niche

  • Louise E. Purton1,2,,
  • David T. Scadden3,4,5,

1St. Vincent's Institute, Fitzroy, Vic., 3065, Australia

2Department of Medicine at St. Vincent's Hospital, The University of Melbourne, Fitzroy, Vic., 3065, Australia

3Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA

4Harvard Stem Cell Institute

5Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA

The hematopoietic stem cell (HSC) niche is the anatomical location in which HSCs reside and self-renew. The HSCs outside the niche do not self-renew and commence the process of differentiation to ultimately produce mature blood cells. In recent years there have been a large number of studies performed to identify the cell types that comprise the hematopoietic stem cell niche, and to determine what factors are important contributions to HSC niche function. These studies are important not only for our understanding of how hematopoietic stem cells are regulated in vivo, but are also likely to be critical for designing stem cell targeted therapies. Here we summarize the history, recent studies, concepts and controversies in these analyses of the hematopoietic stem cell niche, and their implications for the future.

1. Hematopoietic inductive microenvironments and the hematopoietic stem cell niche: a historical perspective

Blood cell production (hematopoiesis) is a dynamic process that requires the replenishment of more than 7 × 109 blood cells (leukocytes, erythrocytes and platelets) per kg body weight per day. Homeostasis of the hematopoietic system is considered to occur by the capacity of hematopoietic stem cells (HSCs) to undergo differentiation decisions to sustain mature blood cell production and self-replication (self-renewal) processes to replenish the HSC pool throughout life. Note however that self-renewal has only been shown under conditions of transplantation, not steady-state, and proof of a single cell undergoing both differentiation and self-renewal (asymmetrically dividing) has not been accomplished. In adult mammals, hematopoiesis predominantly occurs within the bone marrow, which is situated inside the bone cavity.

It should be noted that the majority of studies of hematopoiesis have utilized the rodent model, which has provided us with a large amount of knowledge of the hematopoietic system that has generally translated well into the human. There are, however, some major differences in the regulation of hematopoiesis in human vs rodents, especially the anatomical location of hematopoiesis throughout life. In humans, the spleen does not support hematopoiesis after birth, although extramedullary splenic hematopoiesis can occur in times of hematopoietic stress (O’Malley et al., 2005). In contrast, the spleen remains hematopoietically active in rodents throughout life (Weiss, 1974). Furthermore, all bones of the rodent support hematopoiesis, and the long bones (especially femurs and tibiae) are the major sites in which hematopoiesis is studied. In contrast, hematopoiesis ceases in the long bones of humans between 5 and 7 years of age (with the exception of the proximal regions of these bones), with the red (hematopoietically active) bone marrow being replaced by yellow (hematopoietically inactive) adipose tissue in these bones (Kricun, 1985; Williams, 1995). In adult humans the major sites of hematopoiesis are bones of the axial skeleton (the cranium, sternum, ribs and vertebrae) in addition to the ilium (Williams, 1995). This chapter discusses studies that have predominantly been made in the rodent model- we have yet to learn if the same findings also apply to humans.

The bone marrow consists of the hematopoietic cells and the non-hematopoietic (stromal) cells. Forty years ago, studies performed in the laboratory of Dr John Trentin demonstrated that stromal cells had an active role in the regulation of the differentiation of hematopoietic stem cells (HSCs) into all blood cell lineage types. Trentin proposed that this was an inductive event involving the interaction of stromal cells and HSCs, and the hematopoietic organ stroma were thereby termed hematopoietic inductive microenvironments (HIMs; Trentin, 1971; Wolf and Trentin, 1968).

The first evidence that there were distinct HIMs in different hematopoietic organs was provided in an elegant study using trocar implantation of plugs of bone marrow stroma, a major site of granulopoiesis, within the spleen, predominantly supportive of erythropoiesis (Wolf and Trentin, 1968). In this experiment, primary recipient mice were irradiated and injected with bone marrow cells to reseed the depleted hematopoietic progenitor cells. The mice were euthanized 18–24 hours post-transplantation and plugs of their intact marrow (approximately ¼ to ½ of a femoral content) were implanted by trocar directly into spleens of irradiated secondary recipients. The spleens were then harvested 7 days after transplantation and sections of the transplanted spleens were visualized by light microscopy.

The trocar-implanted bone marrow stroma could clearly be distinguished from the spleen stroma by differences in connective tissue and the presence of bone spicules in the bone marrow stroma (Wolf and Trentin, 1968). Interestingly, the types of colonies visualized in the sections of the transplanted spleens were dependent upon the stromal microenvironment in which they were developing. Within the bone marrow stroma, the colonies were predominantly granulocytic in nature, whereas in the spleen stroma the colonies were predominantly erythroid. In contrast, colonies overlapping the borders of the two stroma types were of mixed lineage (erythrocytes and granulocytes). The stroma of the different organs therefore appeared to induce the differentiation of specific blood cell lineages from the progenitors, and were hence termed HIMs (Trentin, 1971; Wolf and Trentin, 1968). Various HIMs within a hematopoietic organ have since been identified (Avecilla et al., 2004; Tokoyoda et al., 2004). Examples of these include roles of cells of the osteoblast lineage in B lymphopoiesis (Tokoyoda et al., 2004; Zhu et al., 2007) and the regulation of megakaryocytopoiesis by vascular cells (Avecilla et al., 2004).

In 1978 Schofield expanded on the observations of Trentin and colleagues to predict that in addition to differentiation-inducing microenvironments, there was also a specific hematopoietic stem cell niche, which “fixed” the stem cells in place and prevented their maturation, allowing the stem cell to proliferate and retain its stemness. Once the stem cell progeny left the stem cell niche they proceeded to differentiate (Schofield, 1978). These predictions were based on his observations that HSCs needed to reside in the bone marrow to retain their “infinite” potential, whereas those that homed to the spleen and formed colonies (CFU-S) were more restricted in their capacity to sustain hematopoiesis (Schofield, 1978).

During the next 25 years, studies on the bone marrow microenvironment were predominantly based on either microscopy studies (light and ultrastructural; Lichtman, 1981) or ex vivo culture systems, especially the Dexter long-term bone marrow cultures, which were capable of long-term support of hematopoiesis (Dexter et al., 1977; Dexter et al., 1973). In these studies, the bone marrow stromal cells were defined as fibroblasts, reticular cells, endothelial cells, adipocytes and osteoblasts. However the exact identity of the cell type(s) comprising the hematopoietic stem cell niche remained unknown.

In 2003, two reports utilizing mouse models demonstrated that the bone-forming osteoblast was capable of influencing the size of the HSC pool in vivo, implying that the osteoblast was a critical component of the HSC niche (Calvi et al., 2003; Zhang et al., 2003). However, in 2005 a separate study suggested that the sinusoidal endothelial cells within the bone marrow were the HSC niche (Kiel et al., 2005). There is now considerable debate in the field regarding which of these cells form the true niche for hematopoietic stem cells. It is currently not clear if the osteoblasts and endothelial cells represent distinct or overlapping niches, and if separate niches, if the HSCs occupying the niches are the same or have different properties. We will address some of these issues below.

2. Cell types that have been identified to form prospective HSC niches

2.1. Osteoblasts

In adult humans, steady-state hematopoiesis occurs in the bone marrow, which is situated within the bone cavity. Rather than being an inert structure, bone tissue undergoes a constant process of remodeling via a tight coupling between bone formation from osteoblasts (derived from mesenchymal stem cells) and bone resorption by osteoclasts (which are hematopoietic in origin; Martin and Sims, 2005). A number of different cell types of the osteoblast lineage are present in bone and bone marrow including (from most primitive to most mature) mesenchymal stem cells, osteoprogenitor cells, osteoblasts and osteocytes (Mackie, 2003).

Osteoblasts are usually found in a layer along the endosteum at the interface between bone and marrow and periosteum, which comprise the internal and external surfaces of bone, respectively. The major functions of osteoblasts in bone remodeling are the secretion of unmineralized bone matrix proteins (collectively termed osteoid) and cells of the osteoblast lineage regulate osteoclast differentiation (Lian, 2003; Martin and Sims, 2005).

Two complementary and simultaneously published studies identified that osteoblasts play an active role as part of the regulating microenvironment or niche for HSCs (Calvi et al., 2003; Zhang et al., 2003). In one report, the bone morphogenetic protein receptor 1A (BMPR1A) was conditionally deleted from mice using Mx1-Cre (Zhang et al., 2003). This resulted in ectopic formation of a trabecular bone-like area (TBLA), and a significant increase in the number of N-cadherin-positive osteoblasts (SNO cells) in the TBLA (Zhang et al., 2003). The increased TBLA correlated with increased numbers of HSCs (measured by functional as well as phenotypical properties), and label-retaining cells thought to be HSC were found to be attached to the SNO cells in association with N-cadherin (Zhang et al., 2003). It should be noted however, that the promoter driving the Cre recombinase and therefore the excision of BMPR1a was not specific for osteoblasts therefore other cell types might also have been involved.

In contrast, Calvi et al. examined the effects of the constitutively activated parathyroid hormone/parathyroid hormone-related peptide receptor (PPR) under the control of the osteoblast-specific 2.3kB α1(1) collagen promoter (Calvi et al., 2003). The PPR transgenic mice had significantly increased HSCs in conjunction with increased trabecular bone and elevated numbers of trabecular osteoblasts which expressed the Notch ligand Jagged 1. HSC-enriched cells from PPR transgenic mice had more activated Notch 1 than wildtype HSCs. Moreover, contact between the HSCs and Jagged 1-expressing osteoblasts were required for the HSC-potentiating effects of the osteoblasts, and the addition of a γ secretase inhibitor (which inhibits Notch activation) to stromal cell cultures prevented this effect (Calvi et al., 2003). Finally, treatment of wildtype stromal cultures or wildtype mice with parathyroid hormone (PTH) recapitulated the phenotypes observed in PPR transgenic mice (Calvi et al., 2003). PTH has also been shown to have therapeutic potential for both HSC mobilization and hematopoietic recovery post-transplantation via regulation of the size of the osteoblast HSC niche in mouse models (Adams et al., 2007; Calvi et al., 2003).

It should be noted that these in vivo models were initiated largely because of prior in vitro work in which it was noted that osteoblasts or osteoblastic cell lines could support hematopoietic cells in culture. Human osteoblasts have been shown to support HSCs in ex vivo culture systems (Taichman and Emerson, 1994; Taichman and Emerson, 1998; Taichman et al., 1996; Taichman et al., 2000). While studies of osteoblast expansion have demonstrated an increase in HSCs, studies of osteoblast decrease have not consistently shown a reduction in HSCs. Visnjic et al. showed that deletion of osteoblasts by gancyclovir in mice transgenic for thymidine kinase expressed in osteoblasts caused reduced marrow and increased extrameduallary hematopoiesis (Visnjic et al., 2004). This was accompanied by a reduction in absolute numbers of primitive hematopoietic cells in the bone marrow and increased numbers in spleen and liver (Visnjic et al., 2004). However, osteoblast dysfunction in the biglycan deficient mouse did not result in a change in marrow HSC (Kiel et al., 2007). Therefore, either specific subsets of osteoblasts contribute to niche or osteoblasts are not required for niche function and may be supplanted by other cell types under conditions of more gradual decline in osteoblast number. Products of osteoblasts that have been shown to be positive regulators of HSCs include angiopoietin-1, thrombopoietin and Jagged-1 (under conditions of PPR activation; Arai et al., 2004; Calvi et al., 2003; Wilson et al., 2004; Yoshihara et al., 2007), whereas osteoblast-associated negative regulators of the HSC niche include osteopontin and dikkopf1 (Fleming et al., 2008; Nilsson et al., 2005; Stier et al., 2005; reviewed in detail in (Adams and Scadden, 2006; Kiel and Morrison, 2008; Yin and Li, 2006)). While Jagged-1 appears to be critical for PPR activated osteoblast increase in HSC, it does not participate in HSC homeostasis (Mancini et al., 2005). Osteoblasts also abundantly express CXCL12, a chemokine that has been implicated to have roles in chemotaxis, homing, survival of HSCs and in the retention of HSCs in the bone marrow (Broxmeyer et al., 2005). It should be noted, however, that other non-osteoblast “reticular” cells in the marrow also express high levels of CXCL12 and may participate in the niche (Papayannopoulou and Scadden, 2008; Sugiyama et al., 2006). While osteoblasts are contributors to HSC regulation in the marrow, it remains unclear whether direct contact with HSCs is required. What is clear is that other cell types besides the osteoblast participate in HSC regulation in the marrow.

2.2. Endothelial cells

Endothelial cells line all blood vessels in the body. In the bone marrow they form a barrier between the developing hematopoietic cells and the blood. They are therefore the initial site of entrance of all blood cells into the bone marrow from the circulation and also the final place whereby blood cells leave the bone marrow to enter the bloodstream (Sipkins et al., 2005; Winkler and Levesque, 2006). The possibility of a perivascular zone serving as a regulatory niche for stem cells is derived from two studies. In vivo imaging of primitive hematopoietic cells in animals over time revealed that they localized to specific subsections of the marrow microvasculature where cells persisted or increased in number over a 70 day interval (Sipkins et al., 2005). These data suggested the perivascular site as a potential niche, but the hematopoietic populations studied were not highly enriched for HSCs.

It was the discovery of SLAM antigens marking HSCs that enabled histologic assessment of where the HSCs resided in the marrow microenvironment (Kiel et al., 2005). These studies indicated that the majority of HSCs were in the perivascular region with only a minority (∼16%) at the periendosteal region. These data are consistent with the perivascular regions serving as a niche, but several caveats must be kept in mind. The most important is that HSCs traffic into and out of the vasculature, so it remains formally possible that the cells accumulate around vessels because it is an impedance point in their trafficking. As such, this site would not meet the criteria for a niche (a site where self-renewal occurs). Also, in the sponge-like trabecular region where most HSCs reside, it is difficult to discern all anatomic relationships from two-dimensional images. Finally, to date no modification in endothelial function in vivo has been shown to affect HSCs other than what might be expected from peturbed trafficking. Therefore, whether the perivascular zone represents a true niche in the marrow still requires experimental definition

However, developmental changes in hematopoiesis would suggest that perivascular sites are likely to serve as niches and there are in vitro studies indicating that endothelial cells derived from various tissues can support HSCs in culture systems (Chute et al., 2002; Li et al., 2004; Rafii et al., 1995). In embryonic development, HSCs originate in the yolk sac and from hemogenic endothelium in the dorsal aorta in the aorta, gonad, mesonephros (AGM) region. The placenta may also serve as a site of HSC production with the fetal liver becoming the site where HSCs reside, expand and produce blood during the second trimester of gestation. Hematopoiesis establishes in bone marrow only later in the second trimester of embryonic development (Cumano and Godin, 2007; Orkin and Zon, 2008; Orkin and Zon, 2008). With the exception of the bone marrow, all of the sites where self-renewing HSCs can be isolated during embryonic development are devoid of osteoblasts, but contain endothelial cells, which have been closely associated with the generation of the HSCs (Cumano and Godin, 2007; Orkin and Zon, 2008; Orkin and Zon, 2008).

3. Are all HSC niches equal?

The identification of two different cell types as being crucial components of the HSC niche have led to many as yet unanswered questions. Do the osteoblast or the endothelial cell represent true HSC niches, and if so, are these separate niches or do they collectively comprise one HSC niche? Furthermore, are the niches for quiescent and activated HSCs the same or do they differ? And are there other cell types that play equally important roles in the HSC niche?

3.1. One or more HSC niches?

The localization studies of HSCs in their niche in vivo have predominantly relied on visualization of cells in bone marrow sections (Arai et al., 2004; Kiel et al., 2005; Zhang et al., 2003). These two-dimensional sections are not true representations of the three-dimensional structure of the bone marrow microenvironment. Such studies may therefore miss the correct anatomical structure of the HSC niche which may consist of a variety of different cell types rather than being restricted to one cell type. It is highly likely, for example, that the osteoblast-containing HSC niche is in close connection with endothelial cells. Evidence to support this comes from studies demonstrating that the bone marrow (including the endosteal surface) is highly vascularized (de Saint-Georges and Miller, 1992; Narayan et al., 1994). Indeed, vascularization of the cartilage template is a crucial process during the formation of embryonic bone by endochondral ossification, and impaired angiogenesis during this process results in significantly reduced bone formation (Brandi and Collin-Osdoby, 2006; Maes et al., 2002; Maes et al., 2007). Furthermore, the vasculature is highly important in the ongoing process of bone remodeling (Brandi and Collin-Osdoby, 2006). The osteoblast-containing HSC niche is therefore likely also in close proximity to endothelial cells, and these two cell types potentially collectively form the trabecular HSC niche.

There are, however, distinct regions of the bone marrow in which the endothelial cells are not located near osteoblasts, particularly in areas devoid of trabecular bone in the central region of the diaphysis (de Saint-Georges and Miller, 1992; and Dr Natalie Sims, personal communication). It remains to be determined if these endothelial cells also co-localize with HSCs or if they are different to those identified as the perivascular HSC niche. Some recent studies, however, have proposed the existence of two distinct HSC niches that support quiescent or activated HSCs.

3.2. Quiescent versus activated HSC niches

Some studies have noted that the osteoblast-containing niche represents a niche in which HSCs remain quiescent. To date, at least three molecules found on the surface of osteoblasts (N-cadherin, angiopoietin-1 and thrombopoietin) have been shown to regulate HSC quiescence via interaction with their receptors (N-cadherin, Tie-2 or Mpl, respectively), expressed on HSCs (Arai et al., 2004; Haug et al., 2008; Wilson et al., 2004; Wilson et al., 2007; Yoshihara et al., 2007; Zhang et al., 2003). Moreover, osteoblast-specific overexpression of the canonical wingless (Wnt) inhibitor, Dikkopf1 (Dkk1) resulted in loss of HSC quiescence accompanied by reduced serial transplant potential of HSCs (Fleming et al., 2008). These data suggest that Wnt signaling in the niche is important for sustaining HSC quiescence and self-renewal.

In contrast, the perivascular niche has been proposed to represent a niche in which the HSCs are in a more activated state. Such activated HSCs include cycling HSCs or HSCs that have been subjected to stress, such as by treatment with chemotherapeutic drugs (for example, 5-fluorouracil or cyclophosphamide) or cytokines (such as G-CSF; Morrison et al., 1997; Randall and Weissman, 1997; Wilson et al., 2007; Zhang and Li, 2008). It is currently not clear if such a niche may be a transient niche for HSCs, if it represents a stable HSC niche for a particular population of HSCs or if the HSCs at the perivascular niche were merely in transit when they were visualized.

3.3. Different niches for distinct hierarchies of HSCs?

It is now well-recognised that HSCs are not a homogeneous population of stem cells, but are hierarchically organized based on their functional potential, with the most immature HSC capable of sustaining hematopoiesis through serial transplantation (Purton and Scadden, 2007). Some recent studies comparing properties of the HSC populations initially used to identify the prospective HSC niches suggest they may represent different populations of HSCs. Though these studies are controversial, studies identifying the importance of the osteoblast in the HSC niche have commonly utilized the lineage negative, c-Kit-positive, Sca-1-positive (LKS+) population of HSCs (Ikuta and Weissman, 1992; Okada et al., 1992), which is a heterogeneous population of short-term repopulating HSCs, long-term repopulating HSCs and multipotent progenitor cells (Adolfsson et al., 2001; Osawa et al., 1996; Yang et al., 2005). However, further purification of such HSCs was also performed in these studies using the Hoescht 33342 “side population” protocol (Arai et al., 2004; Goodell et al., 1996) or BrdU label retaining population, which has been shown to select for the slowly cycling “quiescent” HSCs (Bradford et al., 1997; Cheshier et al., 1999; Zhang et al., 2003).

One study reported that more than 70% of HSCs identified based on the SLAM markers (CD48-CD150+LKS+) express CD34 (Wilson et al., 2007), a marker that is normally associated with activated (non-quiescent) or short-term repopulating HSCs (Ogawa et al., 2001; Yang et al., 2005). The majority of HSCs identified using SLAM markers are not BrdU label-retaining cells (Kiel and Morrison, 2006) and SLAM-associated HSCs do not express N-cadherin (Haug et al., 2008; Kiel et al., 2007), an adhesion molecule that some have reported maintains quiescent HSCs in the osteoblast niche (Haug et al., 2008; Wilson et al., 2004; Zhang et al., 2003).

Interestingly, during embryonic development HSCs (which are closely associated with vasculature) are actively cycling cells that express CD34 (Ito et al., 2000; Matsuoka et al., 2001; Sanchez et al., 1996). Likewise, mobilized murine HSCs, which are perivascular (Yilmaz et al., 2006), express CD34 (Tajima et al., 2000), whereas in the steady-state condition murine HSCs are CD34 low/negative (Osawa et al., 1996). Hence it is currently unclear if the HSC populations used to initially identify the HSC niches represent two distinct populations of HSCs or if they have the same functional potential. It will be experimentally difficult, but ultimately important to test populations from distinct locations using stringent tests of HSC self-renewal, such as the serial transplant assay (Purton and Scadden, 2007) to determine if there are functional differences in distinctly localized cell populations.

3.4. Do all osteoblasts or endothelial cells support HSCs?

Another current unknown is whether all osteoblasts and/or endothelial cells have the same HSC support potential, or if specialized osteoblasts/endothelial cells or specific locations of osteoblasts/endothelial cells have different roles in regulating HSCs. For example, the trabecular osteoblasts were initially identified as being part of the HSC niche (Calvi et al., 2003; Zhang et al., 2003), however, we commonly refer to the HSC niche as being the endosteal surface (which applies to both cortical and trabecular bone). Of note, cortical and trabecular bone differ in their anatomical location, structure and function. Cortical (compact) bone is located in the diaphyseal region of bone, is thick and dense (80–90% of the bone is calcified) and considered to have predominantly mechanical and protective functions. In contrast, trabecular (cancellous) bone is found in the metaphyseal region of bone, is less dense (15–25% is calcified) and has more of a metabolic function (Baron, 2003). It still remains to be shown whether or not osteoblasts in different anatomical locations have distinct roles with respect to regulating HSCs.

Likewise, endothelial cells are specialized cells that have different functions in distinct tissues and organs (Belloni and Tressler, 1990). There are also differences in properties and functions of endothelial cells derived from different sized blood vessels, eg arteries or capillaries (Belloni and Tressler, 1990). Moreover, HSCs have been shown to home to specific CXCL12-expressing microdomains in the bone marrow vasculature, suggesting specific regions of endothelial cells may function as the perivascular HSC niche (Sipkins et al., 2005).

4. What regulates the HSC niches?

The initial studies describing the osteoblast HSC niche identified two regulators of its size: PPR and BMPR1A (Calvi et al., 2003; Zhang et al., 2003). There are likely many more regulators of the HSC niches to be discovered in future studies. It is also possible that the osteoblast and endothelial HSC niches have roles in regulating each other.

4.1. Do other cell types also play important roles as components or regulators of the HSC niche?

It is highly likely that the HSC niche is more complex than only one cell type. Various studies have revealed that the sympathetic nervous system (SNS) also plays an important role in regulating the HSC niche (Afan et al., 1997; Katayama et al., 2006; Mendez-Ferrer et al., 2008). Anatomical studies have revealed that bone marrow is highly innervated with both myelinated and nonmyelinated nerve fibers (Calvo and Forteza-Vila, 1969; Calvo and Forteza-Vila, 1970; Yamazaki and Allen, 1990), most of which are located near arterioles in the bone marrow (Yamazaki and Allen, 1990). Surgical severance of the femoral nerve resulted in reduced bone marrow cellularity accompanied by significant mobilization of progenitor cells (Afan et al., 1997). In addition, treatment of mice with the neurotoxin 6-hydroxydopamine (which resulted in blockade of neurotransmitter synthesis in adrenergic and dopaminergic nerve fibers) resulted in decreased bone marrow cellularity (Afan et al., 1997).

More recently, the SNS neurotransmittor norepinephrine (NE) was shown to control both the suppression of osteoblasts and downregulation of CXCL12 expression in bone cells that occurs in response to G-CSF treatment (Katayama et al., 2006). When the SNS was disrupted (such as in UDP-galactose ceramide galactosyltransferase-deficient mice, which display defects in nerve conduction due to a lack of myelin), mobilization of HSCs from their niche did not occur. In contrast, when a β2-adrenergic agonist was administered to both control and NE-deficient mice, HSC mobilization was enhanced (Katayama et al., 2006). Additional studies in the mouse model revealed that the cyclical release of HSCs and expression of CXCL12 in the bone marrow microenvironment was regulated by circadian NE secretion by the SNS (Mendez-Ferrer et al., 2008). Neurotransmittors were also recently shown to have roles in human HSC mobilization, proliferation and differentiation (Spiegel et al., 2007).

Other cell types that have potential roles in regulating the HSC niche include chondrocytes (Jacenko et al., 2002), adipocytes (Walkley et al., 2007) and CXCL12-abundant reticular cells (Sugiyama et al., 2006). Further studies will be required to fully elucidate how each of these cell types contributes to the HSC niche.

Hematopoietic cells may also play a role in regulating the HSC niches. For example, monocytes have been shown to express osteopontin, which significantly decreased Notch1 signaling in HSCs that were co-cultured with a supportive bone marrow stromal cell line that expresses Jagged-1 (Iwata et al., 2004; Li et al., 1998). In addition, the monocyte-derived osteoclasts play an important role not only in the regulation of osteoblast activity and hence potentially osteoblast HSC niche size (Martin and Sims, 2005), but elevated Ca2+ concentration released during osteoclast-mediated bone resorption may be important in directing HSCs (which express the calcium-sensing receptor) to the osteoblast-containing HSC niche (Adams et al., 2006). Indeed osteoclasts have been suggested to induce HSC mobilization from the bone marrow microenvironment (Kollet et al., 2006).

4.2. Do the HSC niche components influence each other?

It is currently unknown whether there is cross-talk between the osteoblast and endothelial cells in HSC niches, although there is evidence that endothelial cells and osteoblasts can regulate each other. Angiopoietin-1 (which is expressed by the osteoblast lineage), interacts with Tie-2 expressed by endothelial cells to increase angiogenesis (the formation of new blood vessels) and reduce vascular permeability (Fukuhara et al., 2008; Saharinen et al., 2008). Likewise, osteoblasts secrete vascular endothelial growth factor (VEGF), which also modulates vascularization and permeability of endothelial cells (Ferrara and Davis-Smyth, 1997), and has a central role in bone morphogenesis (Zelzer and Olsen, 2005). Other known interactions between endothelial cells and osteoblasts have been reviewed in detail by Kanczler and Oreffo (Kanczler and Oreffo, 2008).

There have also been a series of reports describing that the perivascular niche is also the niche for mesenchymal stem cells (MSCs), the precursors of the osteoblast (Sacchetti et al., 2007; Shi and Gronthos, 2003). It is currently unclear if the MSC perivascular niche has the same anatomical location and function as the HSC perivascular niche. It may be speculated that, in humans MSCs may be identical to the CXCL12-abundant reticular cells that are also associated with both the perivascular and osteoblast HSC niches (Sacchetti et al., 2007; Sugiyama et al., 2006). Determining whether MSC or the MSC niche participate in altering the HSC niche is an area of great interest that is expected to receive considerable attention in the future.

5. When niches turn bad- evidence for roles of the HSC niches in hematopoietic diseases

Studies have implicated that aberrant hematopoietic microenvironments may play an active role in inducing and/or sustaining hematopoietic disease (Barabe et al., 2007; Ju et al., 2007; Walkley et al., 2007; Walkley et al., 2007). Microenvironment-induced myeloproliferative-like diseases (MPDs) have been shown to occur in mice deficient for retinoic acid receptor gamma (RARγ) or retinoblastoma (Rb). In both mouse models, a marked reduction in trabecular osteoblasts correlated with disease progression, and this was accompanied by loss of HSCs in the bone marrow and increased mobilization of HSCs to extramedullary tissues (Walkley et al., 2007; Walkley et al., 2007). Furthermore, conditional loss of glycoprotein 130 (gp130) in both hematopoietic and endothelial cells resulted in hematopoietic disease, with most mice dying by 12 months of age (Yao et al., 2005). This disease was not intrinsic to the hematopoietic cells, but was attributed to lack of gp130 receptor in the endothelial cells within the BM microenvironment (Yao et al., 2005).

Of clinical relevance, patients with chronic MPDs have been shown to have high levels of VEGF (Panteli et al., 2007), which is not restricted to its effects on endothelial cells as osteoclasts and osteoblasts also express VEGF receptors, and VEGF modulates their activities (Zelzer and Olsen, 2005). Furthermore, some recent reports from the Fred Hutchinson Cancer Research Center, Seattle, USA and the European Group for Blood and Marrow Transplantation have described the occurrence of donor-derived hematopoietic disease in allogeneic bone marrow transplant recipients (Hertenstein et al., 2005; Sala-Torra et al., 2006). Collectively, the studies indicated that the diseases occurred at a frequency of approximately 1 in 800 transplant recipients and included myelodysplastic syndrome, acute myeloid leukemia, chronic myeloid leukemia and acute lymphoblastic leukemia. Interestingly, during the long-term follow-up of the patients and their donors, all of the donors remained healthy, implicating the microenvironment of the patient likely contributed to the disease. Future studies identifying the roles of the cells comprising the HSC niches in both normal and diseased states may provide new insights into the pathophysiology and potentially, treatment opportunities for treating hematologic diseases.


1. Acknowledgments

The authors wish to thank NA Sims, CR Walkley, M Askmyr and TJ Martin for useful discussions and for reading this chapter. This work was funded by the National Health and Medical Research Council (NHMRC) 502612 and National Institutes of Health (NIH) DK71773 to LEP and NIH to DTS. LEP is an NHMRC RD Wright Fellow.

References

Adams, G.B. Chabner, K.T. Alley, I.R. Olson, D.P. Szczepiorkowski, Z.M. Poznansky, M.C. Kos, C.H. Pollak, M.R. Brown, E.M. Scadden, D.T. (2006). Stem cell engraftment at the endosteal niche is specified by the calcium-sensing receptor. Nature 439, 599–603.10.1038/nature04247

Adams, G.B. Martin, R.P. Alley, I.R. Chabner, K.T. Cohen, K.S. Calvi, L.M. Kronenberg, H.M. Scadden, D.T. (2007). Therapeutic targeting of a stem cell niche. Nat Biotechnol 25, 238–243.10.1038/nbt1281

Adams, G.B. Scadden, D.T. (2006). The hematopoietic stem cell in its place. Nat Immunol 7, 333–337.10.1038/ni1331

Adolfsson, J. Borge, O.J. Bryder, D. Theilgaard-Monch, K. Astrand-Grundstrom, I. Sitnicka, E. Sasaki, Y. Jacobsen, S.E. (2001). Upregulation of Flt3 expression within the bone marrow Lin(−)Sca1(+)c-kit(+) stem cell compartment is accompanied by loss of self-renewal capacity. Immunity 15, 659–669.10.1016/S1074-7613(01)00220-5

Afan, A.M. Broome, C.S. Nicholls, S.E. Whetton, A.D. Miyan, J.A. (1997). Bone marrow innervation regulates cellular retention in the murine haemopoietic system. Br J Haematol 98, 569–577.10.1046/j.1365-2141.1997.2733092.x

Arai, F. Hirao, A. Ohmura, M. Sato, H. Matsuoka, S. Takubo, K. Ito, K. Koh, G.Y. Suda, T. (2004). Tie2/angiopoietin-1 signaling regulates hematopoietic stem cell quiescence in the bone marrow niche. Cell 118, 149–161.10.1016/j.cell.2004.07.004

Avecilla, S.T. Hattori, K. Heissig, B. Tejada, R. Liao, F. Shido, K. Jin, D.K. Dias, S. Zhang, F. Hartman, T.E. (2004). Chemokine-mediated interaction of hematopoietic progenitors with the bone marrow vascular niche is required for thrombopoiesis. Nat Med 10, 64–71.10.1038/nm973

Barabe, F. Kennedy, J.A. Hope, K.J. Dick, J.E. (2007). Modeling the initiation and progression of human acute leukemia in mice. Science 316, 600–604.10.1126/science.1139851

Baron, R. (2003). General Principles of Bone Biology. Primer on the Metabolic Bone Diseases and Disorders of Mineral Metabolism Favus, M.J. Washington, DC: American Society for Bone and Mineral Research; , 1–8.

Belloni, P.N. Tressler, R.J. (1990). Microvascular endothelial cell heterogeneity: interactions with leukocytes and tumor cells. Cancer Metastasis Rev 8, 353–389.10.1007/BF00052608

Bradford, G.B. Williams, B. Rossi, R. Bertoncello, I. (1997). Quiescence, cycling, and turnover in the primitive hematopoietic stem cell compartment. Exp Hematol 25, 445–453.

Brandi, M.L. Collin-Osdoby, P. (2006). Vascular biology and the skeleton. J Bone Miner Res 21, 183–192.10.1359/JBMR.050917

Broxmeyer, H.E. Orschell, C.M. Clapp, D.W. Hangoc, G. Cooper, S. Plett, P.A. Liles, W.C. Li, X. Graham-Evans, B. Campbell, T.B. (2005). Rapid mobilization of murine and human hematopoietic stem and progenitor cells with AMD3100, a CXCR4 antagonist. J Exp Med 201, 1307–1318.10.1084/jem.20041385

Calvi, L.M. Adams, G.B. Weibrecht, K.W. Weber, J.M. Olson, D.P. Knight, M.C. Martin, R.P. Schipani, E. Divieti, P. Bringhurst, F.R. (2003). Osteoblastic cells regulate the haematopoietic stem cell niche. Nature 425, 841–846.10.1038/nature02040

Calvo, W. Forteza-Vila, J. (1969). On the development of bone marrow innervation in new-born rats as studied with silver impregnation and electron microscopy. Am J Anat 126, 355–371.10.1002/aja.1001260308

Calvo, W. Forteza-Vila, J. (1970). Schwann cells of the bone marrow. Blood 36, 180–188.

Cheshier, S.H. Morrison, S.J. Liao, X. Weissman, I.L. (1999). In vivo proliferation and cell cycle kinetics of long-term self-renewing hematopoietic stem cells. Proc Natl Acad Sci U S A 96, 3120–3125.10.1073/pnas.96.6.3120

Chute, J.P. Saini, A.A. Chute, D.J. Wells, M.R. Clark, W.B. Harlan, D.M. Park, J. Stull, M.K. Civin, C. Davis, T.A. (2002). Ex vivo culture with human brain endothelial cells increases the SCID-repopulating capacity of adult human bone marrow. Blood 100, 4433–4439.10.1182/blood-2002-04-1238

Cumano, A. Godin, I. (2007). Ontogeny of the hematopoietic system. Annu Rev Immunol 25, 745–785.10.1146/annurev.immunol.25.022106.141538

de Saint-Georges, L. Miller, S.C. (1992). The microcirculation of bone and marrow in the diaphysis of the rat hemopoietic long bones. Anat Rec 233, 169–177.10.1002/ar.1092330202

Dexter, T.M. Allen, T.D. Lajtha, L.G. (1977). Conditions controlling the proliferation of haemopoietic stem cells in vitro. J Cell Physiol 91, 335–344.10.1002/jcp.1040910303

Dexter, T.M. Allen, T.D. Lajtha, L.G. Schofield, R. Lord, B.I. (1973). Stimulation of differentiation and proliferation of haemopoietic cells in vitro. J Cell Physiol 82, 461–473.10.1002/jcp.1040820315

Ferrara, N. Davis-Smyth, T. (1997). The biology of vascular endothelial growth factor. Endocr Rev 18, 4–25.10.1210/er.18.1.4

Fleming, H.E. Janzen, V. Lo Celso, C. Guo, J. Leahy, K.M. Kronenberg, H.M. Scadden, D.T. (2008). Wnt signaling in the niche enforces hematopoietic stem cell quiescence and is necessary to preserve self-renewal in vivo. Cell Stem Cell 2, 274–283.10.1016/j.stem.2008.01.003

Fukuhara, S. Sako, K. Minami, T. Noda, K. Kim, H.Z. Kodama, T. Shibuya, M. Takakura, N. Koh, G.Y. Mochizuki, N. (2008). Differential function of Tie2 at cell-cell contacts and cell-substratum contacts regulated by angiopoietin-1. Nat Cell Biol 10, 513–526.10.1038/ncb1714

Goodell, M.A. Brose, K. Paradis, G. Conner, A.S. Mulligan, R.C. (1996). Isolation and functional properties of murine hematopoietic stem cells that are replicating in vivo. J Exp Med 183, 1797–1806.10.1084/jem.183.4.1797

Haug, J.S. He, X.C. Grindley, J.C. Wunderlich, J.P. Gaudenz, K. Ross, J.T. Paulson, A. Wagner, K.P. Xie, Y. Zhu, R. (2008). N-cadherin expression level distinguishes reserved versus primed states of hematopoietic stem cells. Cell Stem Cell 2, 367–379.10.1016/j.stem.2008.01.017

Hertenstein, B. Hambach, L. Bacigalupo, A. Schmitz, N. McCann, S. Slavin, S. Gratwohl, A. Ferrant, A. Elmaagacli, A. Schwertfeger, R. (2005). Development of leukemia in donor cells after allogeneic stem cell transplantation–a survey of the European Group for Blood and Marrow Transplantation (EBMT). Haematologica 90, 969–975.

Ikuta, K. Weissman, I.L. (1992). 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 89, 1502–1506.10.1073/pnas.89.4.1502

Ito, T. Tajima, F. Ogawa, M. (2000). Developmental changes of CD34 expression by murine hematopoietic stem cells. Exp Hematol 28, 1269–1273.10.1016/S0301-472X(00)00535-X

Iwata, M. Awaya, N. Graf, L. Kahl, C. Torok-Storb, B. (2004). Human marrow stromal cells activate monocytes to secrete osteopontin, which down-regulates Notch1 gene expression in CD34+ cells. Blood 103, 4496–4502.10.1182/blood-2004-01-0256

Jacenko, O. Roberts, D.W. Campbell, M.R. McManus, P.M. Gress, C.J. Tao, Z. (2002). Linking hematopoiesis to endochondral skeletogenesis through analysis of mice transgenic for collagen X. Am J Pathol 160, 2019–2034.

Ju, Z. Jiang, H. Jaworski, M. Rathinam, C. Gompf, A. Klein, C. Trumpp, A. Rudolph, K.L. (2007). Telomere dysfunction induces environmental alterations limiting hematopoietic stem cell function and engraftment. Nat Med 13, 742–747.10.1038/nm1578

Kanczler, J.M. Oreffo, R.O. (2008). Osteogenesis and angiogenesis: the potential for engineering bone. Eur Cell Mater 15, 100–114.

Katayama, Y. Battista, M. Kao, W.M. Hidalgo, A. Peired, A.J. Thomas, S.A. Frenette, P.S. (2006). Signals from the sympathetic nervous system regulate hematopoietic stem cell egress from bone marrow. Cell 124, 407–421.10.1016/j.cell.2005.10.041

Kiel, M.J. Morrison, S.J. (2006). Maintaining hematopoietic stem cells in the vascular niche. Immunity 25, 862–864.10.1016/j.immuni.2006.11.005

Kiel, M.J. Morrison, S.J. (2008). Uncertainty in the niches that maintain haematopoietic stem cells. Nat Rev Immunol 8, 290–301.10.1038/nri2279

Kiel, M.J. Radice, G.L. Morrison, S.J. (2007). Lack of evidence that hematopoietic stem cells depend on N-cadherin-mediated adhesion to osteoblasts for their maintenance. Cell Stem Cell 1, 204–217.10.1016/j.stem.2007.06.001

Kiel, M.J. Yilmaz, O.H. Iwashita, T. Yilmaz, O.H. Terhorst, C. Morrison, S.J. (2005). SLAM family receptors distinguish hematopoietic stem and progenitor cells and reveal endothelial niches for stem cells. Cell 121, 1109–1121.10.1016/j.cell.2005.05.026

Kollet, O. Dar, A. Shivtiel, S. Kalinkovich, A. Lapid, K. Sztainberg, Y. Tesio, M. Samstein, R.M. Goichberg, P. Spiegel, A. (2006). Osteoclasts degrade endosteal components and promote mobilization of hematopoietic progenitor cells. Nat Med 12, 657–664.10.1038/nm1417

Kricun, M.E. (1985). Red-yellow marrow conversion: its effect on the location of some solitary bone lesions. Skeletal Radiol 14, 10–19.10.1007/BF00361188

Li, L. Milner, L.A. Deng, Y. Iwata, M. Banta, A. Graf, L. Marcovina, S. Friedman, C. Trask, B.J. Hood, L. Torok-Storb, B. (1998). The human homolog of rat Jagged1 expressed by marrow stroma inhibits differentiation of 32D cells through interaction with Notch1. Immunity 8, 43–55.10.1016/S1074-7613(00)80457-4

Li, W. Johnson, S.A. Shelley, W.C. Yoder, M.C. (2004). Hematopoietic stem cell repopulating ability can be maintained in vitro by some primary endothelial cells. Exp Hematol 32, 1226–1237.10.1016/j.exphem.2004.09.001

Lian, J.B. Stein, G.S. Aubin, J.E. (2003). Bone formation: maturation and functional activities of osteoblast lineage cells. Primer on the Metabolic Bone Diseases and Disorders of Mineral Metabolism Favus, M.J. Washington, DC: American Society for Bone and Mineral Research; , 13–28.

Lichtman, M.A. (1981). The ultrastructure of the hemopoietic environment of the marrow: a review. Exp Hematol 9, 391–410.

Mackie, E.J. (2003). Osteoblasts: novel roles in orchestration of skeletal architecture. Int J Biochem Cell Biol 35, 1301–1305.10.1016/S1357-2725(03)00107-9

Maes, C. Carmeliet, P. Moermans, K. Stockmans, I. Smets, N. Collen, D. Bouillon, R. Carmeliet, G. (2002). Impaired angiogenesis and endochondral bone formation in mice lacking the vascular endothelial growth factor isoforms VEGF164 and VEGF188. Mech Dev 111, 61–73.10.1016/S0925-4773(01)00601-3

Maes, C. Kobayashi, T. Kronenberg, H.M. (2007). A novel transgenic mouse model to study the osteoblast lineage in vivo. Ann N Y Acad Sci 1116, 149–164.10.1196/annals.1402.060

Mancini, S.J. Mantei, N. Dumortier, A. Suter, U. Macdonald, H.R. Radtke, F. (2005). Jagged1-dependent Notch signaling is dispensable for hematopoietic stem cell self-renewal and differentiation. Blood 105, 2340–2342.10.1182/blood-2004-08-3207

Martin, T.J. Sims, N.A. (2005). Osteoclast-derived activity in the coupling of bone formation to resorption. Trends Mol Med 11, 76–81.10.1016/j.molmed.2004.12.004

Matsuoka, S. Ebihara, Y. Xu, M. Ishii, T. Sugiyama, D. Yoshino, H. Ueda, T. Manabe, A. Tanaka, R. Ikeda, Y. (2001). CD34 expression on long-term repopulating hematopoietic stem cells changes during developmental stages. Blood 97, 419–425.10.1182/blood.V97.2.419

Mendez-Ferrer, S. Lucas, D. Battista, M. Frenette, P.S. (2008). Haematopoietic stem cell release is regulated by circadian oscillations. Nature 452, 442–447.10.1038/nature06685

Morrison, S.J. Wright, D.E. Weissman, I.L. (1997). Cyclophosphamide/granulocyte colony-stimulating factor induces hematopoietic stem cells to proliferate prior to mobilization. Proc Natl Acad Sci U S A 94, 1908–1913.10.1073/pnas.94.5.1908

Narayan, K. Juneja, S. Garcia, C. (1994). Effects of 5-fluorouracil or total-body irradiation on murine bone marrow microvasculature. Exp Hematol 22, 142–148.

Nilsson, S.K. Johnston, H.M. Whitty, G.A. Williams, B. Webb, R.J. Denhardt, D.T. Bertoncello, I. Bendall, L.J. Simmons, P.J. Haylock, D.N. (2005). Osteopontin, a key component of the hematopoietic stem cell niche and regulator of primitive hematopoietic progenitor cells. Blood 106, 1232–1239.10.1182/blood-2004-11-4422

O’Malley, D.P. Kim, Y.S. Perkins, S.L. Baldridge, L. Juliar, B.E. Orazi, A. (2005). Morphologic and immunohistochemical evaluation of splenic hematopoietic proliferations in neoplastic and benign disorders. Mod Pathol 18, 1550–1561.

Ogawa, M. Tajima, F. Ito, T. Sato, T. Laver, J.H. Deguchi, T. (2001). CD34 expression by murine hematopoietic stem cells. Developmental changes and kinetic alterations. Ann N Y Acad Sci 938, 139–145.

Okada, S. Nakauchi, N. Nagayoshi, K. Nishikawa, S. Miura, Y. Suda, T. (1992). In Vivo and In Vitro Cell Function of c-kit- and Sca-1-Positive Murine Hematopoietic Cells. Blood 80, 3044–3050.

Orkin, S.H. Zon, L.I. (2008). Hematopoiesis: an evolving paradigm for stem cell biology. Cell 132, 631–644.10.1016/j.cell.2008.01.025

Orkin, S.H. Zon, L.I. (2008). SnapShot: hematopoiesis. Cell 132, 712.

Osawa, M. Hanada, K. Hamada, H. Nakauchi, H. (1996). Long-term lymphohematopoietic reconstitution by a single CD34-low/negative hematopoietic stem cell. Science 273, 242–245.10.1126/science.273.5272.242

Panteli, K. Bai, M. Hatzimichael, E. Zagorianakou, N. Agnantis, N.J. Bourantas, K. (2007). Serum levels, and bone marrow immunohistochemical expression of, vascular endothelial growth factor in patients with chronic myeloproliferative diseases. Hematology 12, 481–486.10.1080/10245330701554664

Papayannopoulou, T. Scadden, D.T. (2008). Stem-cell ecology and stem cells in motion. Blood 111, 3923–3930.10.1182/blood-2007-08-078147

Purton, L.E. Scadden, D.T. (2007). Limiting factors in murine hematopoietic stem cell assays. Cell Stem Cell 1, 263–270.10.1016/j.stem.2007.08.016

Rafii, S. Shapiro, F. Pettengell, R. Ferris, B. Nachman, R.L. Moore, M.A. Asch, A.S. (1995). Human bone marrow microvascular endothelial cells support long-term proliferation and differentiation of myeloid and megakaryocytic progenitors. Blood 86, 3353–3363.

Randall, T.D. Weissman, I.L. (1997). Phenotypic and functional changes induced at the clonal level in hematopoietic stem cells after 5-fluorouracil treatment. Blood 89, 3596–3606.

Sacchetti, B. Funari, A. Michienzi, S. Di Cesare, S. Piersanti, S. Saggio, I. Tagliafico, E. Ferrari, S. Robey, P.G. Riminucci, M. Bianco, P. (2007). Self-renewing osteoprogenitors in bone marrow sinusoids can organize a hematopoietic microenvironment. Cell 131, 324–336.10.1016/j.cell.2007.08.025

Saharinen, P. Eklund, L. Miettinen, J. Wirkkala, R. Anisimov, A. Winderlich, M. Nottebaum, A. Vestweber, D. Deutsch, U. Koh, G.Y. (2008). Angiopoietins assemble distinct Tie2 signalling complexes in endothelial cell-cell and cell-matrix contacts. Nat Cell Biol 10, 527–537.10.1038/ncb1715

Sala-Torra, O. Hanna, C. Loken, M.R. Flowers, M.E. Maris, M. Ladne, P.A. Mason, J.R. Senitzer, D. Rodriguez, R. Forman, S.J. (2006). Evidence of donor-derived hematologic malignancies after hematopoietic stem cell transplantation. Biol Blood Marrow Transplant 12, 511–517.10.1016/j.bbmt.2006.01.006

Sanchez, M.J. Holmes, A. Miles, C. Dzierzak, E. (1996). Characterization of the first definitive hematopoietic stem cells in the AGM and liver of the mouse embryo. Immunity 5, 513–525.10.1016/S1074-7613(00)80267-8

Schofield, R. (1978). The relationship between the spleen colony-forming cell and the haemopoietic stem cell. Blood Cells 4, 7–25.

Shi, S. Gronthos, S. (2003). Perivascular niche of postnatal mesenchymal stem cells in human bone marrow and dental pulp. J Bone Miner Res 18, 696–704.10.1359/jbmr.2003.18.4.696

Sipkins, D.A. Wei, X. Wu, J.W. Runnels, J.M. Cote, D. Means, T.K. Luster, A.D. Scadden, D.T. Lin, C.P. (2005). In vivo imaging of specialized bone marrow endothelial microdomains for tumour engraftment. Nature 435, 969–973.10.1038/nature03703

Spiegel, A. Shivtiel, S. Kalinkovich, A. Ludin, A. Netzer, N. Goichberg, P. Azaria, Y. Resnick, I. Hardan, I. Ben-Hur, H. (2007). Catecholaminergic neurotransmitters regulate migration and repopulation of immature human CD34+ cells through Wnt signaling. Nat Immunol 8, 1123–1131.10.1038/ni1509

Stier, S. Ko, Y. Forkert, R. Lutz, C. Neuhaus, T. Grunewald, E. Cheng, T. Dombkowski, D. Calvi, L.M. Rittling, S.R. Scadden, D.T. (2005). Osteopontin is a hematopoietic stem cell niche component that negatively regulates stem cell pool size. J Exp Med 201, 1781–1791.10.1084/jem.20041992

Sugiyama, T. Kohara, H. Noda, M. Nagasawa, T. (2006). Maintenance of the hematopoietic stem cell pool by CXCL12-CXCR4 chemokine signaling in bone marrow stromal cell niches. Immunity 25, 977–988.10.1016/j.immuni.2006.10.016

Taichman, R.S. Emerson, S.G. (1994). Human osteoblasts support hematopoiesis through the production of granulocyte colony-stimulating factor. J Exp Med 179, 1677–1682.10.1084/jem.179.5.1677

Taichman, R.S. Emerson, S.G. (1998). The role of osteoblasts in the hematopoietic microenvironment. Stem Cells 16, 7–15.

Taichman, R.S. Reilly, M.J. Emerson, S.G. (1996). Human osteoblasts support human hematopoietic progenitor cells in vitro bone marrow cultures. Blood 87, 518–524.

Taichman, R.S. Reilly, M.J. Emerson, S.G. (2000). The Hematopoietic Microenvironment: Osteoblasts and The Hematopoietic Microenvironment. Hematology 4, 421–426.

Tajima, F. Sato, T. Laver, J.H. Ogawa, M. (2000). CD34 expression by murine hematopoietic stem cells mobilized by granulocyte colony-stimulating factor. Blood 96, 1989–1993.

Tokoyoda, K. Egawa, T. Sugiyama, T. Choi, B.I. Nagasawa, T. (2004). Cellular niches controlling B lymphocyte behavior within bone marrow during development. Immunity 20, 707–718.10.1016/j.immuni.2004.05.001

Trentin, J.J. (1971). Determination of bone marrow stem cell differentiation by stromal hemopoietic inductive microenvironments (HIM). Am J Pathol 65, 621–628.

Visnjic, D. Kalajzic, Z. Rowe, D.W. Katavic, V. Lorenzo, J. Aguila, H.L. (2004). Hematopoiesis is severely altered in mice with an induced osteoblast deficiency. Blood 103, 3258–3264.10.1182/blood-2003-11-4011

Walkley, C.R. Olsen, G.H. Dworkin, S. Fabb, S.A. Swann, J. McArthur, G.A. Westmoreland, S.V. Chambon, P. Scadden, D.T. Purton, L.E. (2007). A Microenvironment-Induced Myeloproliferative Syndrome Caused by Retinoic Acid Receptor gamma Deficiency. Cell 129, 1097–1110.10.1016/j.cell.2007.05.014

Walkley, C.R. Shea, J.M. Sims, N.A. Purton, L.E. Orkin, S.H. (2007). Rb Regulates Interactions between Hematopoietic Stem Cells and Their Bone Marrow Microenvironment. Cell 129, 1081–1095.10.1016/j.cell.2007.03.055

Weiss, L. (1974). A scanning electron microscopic study of the spleen. Blood 43, 665–691.

Williams, W. Nelson, DA. (1995). Examination of the marrow. Hematology Williams Beulter, E. Lichtman, M.A. Coller, B.S. Kipps, T.J. New York: McGraw-Hill; , 15–22.

Wilson, A. Murphy, M.J. Oskarsson, T. Kaloulis, K. Bettess, M.D. Oser, G.M. Pasche, A.C. Knabenhans, C. Macdonald, H.R. Trumpp, A. (2004). c-Myc controls the balance between hematopoietic stem cell self-renewal and differentiation. Genes Dev 18, 2747–2763.10.1101/gad.313104

Wilson, A. Oser, G.M. Jaworski, M. Blanco-Bose, W.E. Laurenti, E. Adolphe, C. Essers, M.A. Macdonald, H.R. Trumpp, A. (2007). Dormant and self-renewing hematopoietic stem cells and their niches. Ann N Y Acad Sci 1106, 64–75.10.1196/annals.1392.021

Winkler, I.G. Levesque, J.P. (2006). Mechanisms of hematopoietic stem cell mobilization: when innate immunity assails the cells that make blood and bone. Exp Hematol 34, 996–1009.10.1016/j.exphem.2006.04.005

Wolf, N.S. Trentin, J.J. (1968). Hemopoietic colony studies. V. Effect of hemopoietic organ stroma on differentiation of pluripotent stem cells. J Exp Med 127, 205–214.10.1084/jem.127.1.205

Yamazaki, K. Allen, T.D. (1990). Ultrastructural morphometric study of efferent nerve terminals on murine bone marrow stromal cells, and the recognition of a novel anatomical unit: the “neuro-reticular complex”. Am J Anat 187, 261–276.10.1002/aja.1001870306

Yang, L. Bryder, D. Adolfsson, J. Nygren, J. Mansson, R. Sigvardsson, M. Jacobsen, S.E. (2005). Identification of Lin(−)Sca1(+)kit(+)CD34(+)Flt3- short-term hematopoietic stem cells capable of rapidly reconstituting and rescuing myeloablated transplant recipients. Blood 105, 2717–2723.10.1182/blood-2004-06-2159

Yao, L. Yokota, T. Xia, L. Kincade, P.W. McEver, R.P. (2005). Bone marrow dysfunction in mice lacking the cytokine receptor gp130 in endothelial cells. Blood 106, 4093–4101.10.1182/blood-2005-02-0671

Yilmaz, O.H. Kiel, M.J. Morrison, S.J. (2006). SLAM family markers are conserved among hematopoietic stem cells from old and reconstituted mice and markedly increase their purity. Blood 107, 924–930.10.1182/blood-2005-05-2140

Yin, T. Li, L. (2006). The stem cell niches in bone. J Clin Invest 116, 1195–1201.10.1172/JCI28568

Yoshihara, H. Arai, F. Hosokawa, K. Hagiwara, T. Takubo, K. Nakamura, Y. Gomei, Y. Iwasaki, H. Matsuoka, S. Miyamoto, K. (2007). Thrombopoietin/MPL signaling regulates hematopoietic stem cell quiescence and interaction with the osteoblastic niche. Cell Stem Cell 1, 685–697.10.1016/j.stem.2007.10.020

Zelzer, E. Olsen, B.R. (2005). Multiple roles of vascular endothelial growth factor (VEGF) in skeletal development, growth, and repair. Curr Top Dev Biol 65, 169–187.10.1016/S0070-2153(04)65006-X

Zhang, J. Li, L. (2008). Stem cell niche: microenvironment and beyond. J Biol Chem 283, 9499–9503.10.1074/jbc.R700043200

Zhang, J. Niu, C. Ye, L. Huang, H. He, X. Tong, W.G. Ross, J. Haug, J. Johnson, T. Feng, J.Q. (2003). Identification of the haematopoietic stem cell niche and control of the niche size. Nature 425, 836–841.10.1038/nature02041

Zhu, J. Garrett, R. Jung, Y. Zhang, Y. Kim, N. Wang, J. Joe, G.J. Hexner, E. Choi, Y. Taichman, R.S. Emerson, S.G. (2007). Osteoblasts support B-lymphocyte commitment and differentiation from hematopoietic stem cells. Blood 109, 3706–3712.10.1182/blood-2006-08-041384

This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

To whom correspondence should be addressed. E-mail: lpurton@svi.edu.au and scadden.david@mgh.harvard.edu
*Edited by Leslie Silberstein. Last revised October 21, 2008. Published November 15, 2008. This chapter should be cited as: Purton, L.E. and Scadden, D.T., The hematopoietic stem cell niche (November 15, 2008), StemBook, ed. The Stem Cell Research Community, StemBook, doi/10.3824/stembook.1.28.1, https://www.stembook.org.