The Skeletal Stem Cell

The Skeletal Stem Cell

C H A P T E R 7 The Skeletal Stem Cell Dongsu Park*, Jonathan Hoggatt*, Francesca Ferraro*, David T. Scadden Center for Regenerative Medicine, Massa...

1MB Sizes 4 Downloads 146 Views

C H A P T E R

7

The Skeletal Stem Cell Dongsu Park*, Jonathan Hoggatt*, Francesca Ferraro*, David T. Scadden Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA; Department of Stem Cells and ­ egenerative Biology, Harvard University, Cambridge, MA; Harvard Stem Cell Institute, Cambridge, MA, USA R *Equal contribution

INTRODUCTION The lineage relationships and kinetics of the cellular components of bone are the focus of this chapter. Tissues in mammals are generally maintained by one of two schemas, mature cell replacement by mature cell division or mature cell replenishment from a more immature stem or progenitor population. It is the latter model that applies to bone and we will review here the definition of the skeletal stem cell (SSC), its relationship to descendent semi-committed osteoprogenitors, and the terminally differentiated mesenchymal cells (MC) that make up the mature bone. In addition, we will discuss the migration of mesenchymal osteoprogenitors, their kinetics in regulation of life-long bone production, and the interaction of bone cells with hematopoietic cells in the bone marrow.

DEFINING SKELETAL STEM CELLS The human skeleton is formed during embryogenesis and is estimated to undergo complete turnover four to five times in an average human lifetime [1]. Maintenance of bone tissue is thought to be accomplished by a pool of stem cells. Stem cells are defined by two cardinal features: their ability to self-renew, thereby providing durable replenishment of themselves and their descendants, and their ability to differentiate to achieve specific mature cell fates. Stem cells are therefore functionally defined. Their most rigorous experimental definition requires that they achieve their two cardinal functions in vivo and on a clonal (single cell) level. This has been achieved with relatively few stem cell types to date and is best exemplified by the hematopoietic system. In that

Osteoporosis. http://dx.doi.org/10.1016/B978-0-12-415853-5.00007-8

context, single cells have been shown on transplantation to repopulate all blood lineages in a recipient including the production of new stem cells that can repopulate a second recipient. It is serial transplantation that is used as an indicator of in vivo self-renewal. The accomplishment of the experimental definition of a stem cell is not possible in humans and therefore much of what we understand about stem cells in general and SSCs in particular is derived from animal models. The mouse has been the most common animal model used either by examining mouse cell functions in vivo or studying human cells transplanted into immunodeficient mice. The information discussed in this chapter should, therefore, be viewed with these caveats in mind and recognition that not all conclusions that will be presented from experimental work can be rigidly assumed to hold true in humans. It is also important to realize that the term “stem cells” is often applied to cells that have not met the rigorous definitions indicated above. This is particularly problematic with bone-derived MCs where the term “mesenchymal stem cells” (MSC) has been applied to a wide range of human and mouse cells. Many cells called MSCs have been so labeled because of their functions in vitro not in vivo. Such assessment may or may not reflect the cell's function in the body as MCs are notoriously plastic when cultured. A note of caution to the reader then on equating an MSC with true, functionally defined SSCs: these cells cannot, and should not, be assumed to be the same. Because the MCs that are grown ex vivo have extensive medical testing and interest, but lack rigorous confirmation that they reflect an in vivo stem cell population, they will be termed here as mesenchymal cells (MC).

127

Copyright © 2013 Elsevier Inc. All rights reserved.

128

7.  THE SKELETAL STEM CELL

When considering SSCs, it should also be recognized that there may be different types of SSCs. Multiple cell types comprise the bone and they may be derived from shared or distinct stem cell types. For example, stem cells may produce all the mesenchymal lineages of the bone, as is proposed for putative “MC” or they may produce only a single cell type, such as osteolineage cells, a heterogeneous cell population comprised of bone-lining cells across a spectrum of differentiation and cells incorporated within calcified bone (osteocytes). In this chapter, we will define a “skeletal stem cell” (SSC) as a cell that is harvested from bone and meets the experimental criteria for a stem cell in vivo. Where rigorous experimental definition is lacking, we will point out the limitations. The cells will often also be further delineated as “multipotent-SSCs”, those capable of giving rise to skeletal tissues including osteogenic, adipogenic, and chondrogenic cells, or “osteogenic-SSCs”, those restricted to osteogenic differentiation, but maintaining stem cell features. The distinction between stem cells and progenitor cells also merits comment. Progenitor cells are often highly proliferative components of a tissue that are intermediate in their differentiation state. They are destined to differentiate and do not generally have self-renewal capability. Progenitor cells are, therefore, distinct from stem cells in their self-renewing ability. Because they are intermediate in differentiation state and are often capable of rapid proliferation, progenitors are referred to as a transit, amplifying population. Clear boundaries between the stem and progenitor pools of cells exist conceptually, but are often difficult to define experimentally or by cell markers; therefore, it is common for primitive cells to be clumped together in one heterogeneous description as stem/progenitor cells.

Developmental Origin of Bone and Skeletal Stem Cells The skeletal system derives from the mesodermal layer of the embryonic disc. Intra-embryonic bone is formed either by direct ossification of embryonic connective tissue (intramembranous ossification) or by replacement of hyaline cartilage (endochondral or intracartilaginous ossification) [2]. Intramembranous ossification forms the skeletal infrastructure of the head (roof of the skull and most bone of the face) and begins in the primordial mesenchyme. As the primordial mesenchyme becomes increasingly vascularized, proliferating MCs give rise to osteoprogenitors, which subsequently develop into mature osteoblasts. As osteoblasts collect, they begin to lay down the osteoid, an organic unmineralized portion of the bone matrix. The osteoid is then mineralized with hydroxyapatite crystals in a centrifugal manner beginning at the center of ossification. Concurrently, the

spicules of spongy bone develop and group together to form the trabeculae. As the bone compacts, osteoblasts become entrapped in the newly formed bone and are then referred to as osteocytes. Endochondral ossification takes place at the base of the skull, vertebrae, hips, and limbs through the replacement of a cartilaginous rudiment with bone. The process begins with the formation of the primary ossification center, localized in the diaphysis. Here, the cartilage cells become hypertrophic and the cartilage matrix calcifies. In parallel, the outside portion of the chondrogenic blastema evolves into a perichondrium that subsequently develops into the primitive periosteum. Osteoprogenitors in the periosteum contribute to the formation of a bony collar, and then subsequently certain areas of the calcified matrix disintegrate, opening cavities that then communicate with connective tissue and vessels at the surface that invade the developing marrow cavity. The bone collar holds together the shaft of the bone, which has been weakened by the disintegration of the cartilage. Once bone growth is completed, SSCs capable of forming bone, cartilage, and fat remain in the bone marrow and contribute to renewal of the skeleton throughout life [3].

Isolation and Identification of Skeletal Stem Cells The first experiments demonstrating that postnatal SSCs might exist were performed in the late 1960s, showing that transplantation of bone marrow-derived cells in heterotopic sites was capable of generating a bone template that was then repopulated by marrow cells, thus recapitulating embryonic development [4,5]. In 1970, Friedenstein identified that osteogenic potential was specifically attributable to a small subset of bone marrow cells that were adherent, spindle-shaped, and capable of forming fibroblast colonies (colony-forming unit-fibroblast (CFU-F)) [6]. Much like studies exploring hematopoietic stem and progenitor cells (HSPC) at the time, the identification of a CFU-F allowed for the establishment of clonogenicity, demonstrating that resultant experimental effects were due to a single cell. These cells, after in vitro culture forming CFU-F, were implanted subcutaneously in mice and demonstrated the ability to produce bone (ossicles) [4,6–8]. These cells were also capable of serial transplantation into mice with formation of new ossicles, presumably demonstrating their self-renewal ability [9]. More recently, it has been demonstrated that the cell type in humans that forms colonies, can be transplanted and generates bone and bone marrow and is capable of secondary colony formation, is marked by the expression of the antigen, CD146 [10]. These cells were capable of serial transplantation and ossicle formation at heterotypic sites at a clonal level in immunodeficient mice [10]. This is the most compelling evidence for an

II.  CELLULAR, MOLECULAR, AND DEVELOPMENTAL BIOLOGY OF BONE

Migration of Skeletal Stem Cells

SSC in humans. The only caveat in interpreting these studies is that the cells were assessed after culture rather than as freshly isolated cells. It is, therefore, possible that characteristics of the cells changed during the time of in vitro culture. Also of note, the cells that were generated both in vivo and in vitro were of the osteolineage. Whether the cells also made other MC types in vivo was not assessed and, therefore, it would be proper to consider the CD146+ cell as an osteogenic-SSC. The use of antigenic markers to subselect for cells is exceptionally important for studies of stem cell function and number. The study of CD146+ cells therefore represents an important advance in that it validated the functions of the antigen bearing cells. Other markers have also been studied though with perhaps less rigor and are summarized in Table 7.1. The reader is reminded that the antigens studied have enabled fractionation of bone marrow cells to enrich for cells with SSC features by flow cytometric cell sorting. However, there is not a clear method by which to purify SSC to unity (each cell having SSC features). Some of these markers have also been used to evaluate the location of SSCs in the bone by immunohistochemistry. For example, human cells expressing STRO-1 [11,12], CD105 [13], and CD146 surround blood vessels in vivo (Fig. 7.1). A perivascular location for the SSC is now an accepted concept in the field. It has raised the issue of whether SSCs are equivalent to, or are a subset of, so-called pericytes. It has also raised the issue of whether the cells are capable of trafficking into and out of the vasculature. As might be expected from the interest in SSC and the relative ease of using the model, much work has been done on SSC in the mouse. In one such example, green fluorescent protein (GFP) was driven by the type I collagen (ColI) promoter in a genetically engineered mouse. Whole bone marrow taken from the animal was transplanted into lethally irradiated recipient mice to test whether transplanted SSC could engraft in a manner similar to blood stem cells [93]. GFP-positive cells were tracked within the bone of the transplanted animals and could be seen lining endosteal and trabecular bone, suggesting that SSC transfer had occurred. There was no evidence for the cells progressing to osteocytes, and when the cells were cultured, they did not form mineralized bone nodules, but these studies were indicative of a transplantable osteolineage cell [93]. Others have used flow cytometry to define subpopulations of bone marrow cells and evaluated their ability to form osteoblasts and adipocytes as evidence for SSC. Using the markers, PDGFRα+, Sca-1+, CD45–, and TER119–, it was possible to demonstrate that this population could engraft and form osteogenic and adipogenic tissue, again supporting the notion of a transplantable SSC [94]. Others have tried to use in vivo labeling of defined subsets of MCs to track whether they are capable of

129

functioning as stem cells in the mouse. These so-called lineage tracing studies have indicated that Nestin [95,96] and Mx1 [96] labeled populations of bone marrow perivascular cells with characteristics of SSCs. When the progeny of adult Nestin-expressing bone marrow cells were tracked by using a genetically engineered mouse, the cells were found to contribute to skeletal remodeling through differentiation into cartilage- and bone-forming cells [95]. Interestingly, their contribution to osteoblasts and chondrocytes required 8 months to be visualized, suggesting that these cells may be quiescent and that there may be distinct stem/progenitors that more readily and rapidly contribute to osteogenesis. Perivascular Nestin-positive cells have also been described in histological sections of human bone marrow biopsies [97] (Fig. 7.2). Mx1 is an interferon-inducible promoter that has been used to label cells that contribute to durable osteogenesis over time and can be serially transplantable. This may, therefore, also be a marker for endogenous SSCs [96]. A difference between these two models is that, unlike the delayed appearance of osteoblastic progeny from Nestin-labeled bone marrow cells, Mx1labeled SSCs can replenish the vast majority of osteogenic cells within 20 days and can do so durably over time [96]. Therefore, Mx1-labeled cells may be more rapidly dividing cells. However, Mx1-labeled cells do not appear to contribute to chondrogenesis, suggesting that Mx1 can label active osteogenic-SSCs that supply the majority of new osteoblasts over time, whereas Nestin may mark more quiescent, multipotent-SSCs. Heterogeneity among stem cells within other tissues has also been defined and may reflect a general principle for how the stem cell compartment has distinctive subsets of cells performing related, but different functions [277].

MIGRATION OF SKELETAL STEM CELLS If SSCs are capable of migration then they may participate in bone repair at a distance, opening the possibility of a systemically administered SSC therapy. As early as the beginning of the 20th century, the presence of circulating mesenchymal progenitors was suggested by the presence of CFU-F in the peripheral blood (as reviewed in [98]). Numerous reports confirmed the presence of circulating fibroblastic progenitors in mouse [98–101], adult humans [27,98,99,102–107], and human umbilical cord blood [108–113]. One study reported that some of these circulating cells express osteocalcin (Ocn) and form bone nodules in vitro, although their functional contribution to bone remodeling in vivo is not clear [114]. In a study where skin flaps between animals are joined, so-called parabiotic animals, it is possible to test if cells from one genetically tagged animal can traverse the ­circulatory system and populate the parabiotic partner.

II.  CELLULAR, MOLECULAR, AND DEVELOPMENTAL BIOLOGY OF BONE

130

7.  THE SKELETAL STEM CELL

TABLE 7.1  Phenotypic Markers for Human Skeletal Stem Cell Identification Markers

Molecules

References (+) or (–)

CD4

T4

(–) [14]

CD5

LEU1

(–) [15]

CD9

Non-descript tetraspanin

(+) [14]

CD10

Membrane metallo-endopeptidase (Neprilysin)

(+) [16–18]

CD11a

Integrin α L chain

(–) [14,19]

CD11ba

Integrin α M chain

(–) [20]

CD13

Aminopeptidase N

(+) [15–18,21–23]

CD14a

Monocyte differentiation antigen

(–) [14–16,21–27]

CD15

Fucosyltransferase 4

(–) [14]

CD18

Integrin β 2 chain

(–) [14,19]

CD25

Interleukin-2 receptor α

(–) [14]

CD29

Integrin β 1 chain

(+) [14,15,19,21–25,28–31]

CD31

Platelet/endothelial cell adhesion molecule (PECAM)

(–) [10,14,21,22,32,33]

CD34a

Nondescript sialomucin

(–) [14,26,34–36] (–/+) [37,38]

CD38

Cyclic ADP ribose hydrolase

(–)[21]

CD44

Hyaluronic acid receptor

(+) [14,21,23,25,28–32,39–42] (–) [43]b

CD45

Protein tyrosine phosphatase, receptor type, C (PTPRC)

(–) [14,35,37,44–49] (dim) [50,51]

CD49a

Integrin α 1 chain

(+) [14,48,50,52–54]

CD49b

Integrin α 2 chain

(+) [14] (low) [55]

CD49c

Integrin α 3 chain

(+) [14]

CD49d

Integrin α 4 chain

(–) [14]

CD49e

Integrin α 5 chain

(+) [14] (low) [47]

CD50

Intercellular adhesion molecule 3 (ICAM3)

(–) [14]

CD51

Integrin α V chain

(+) [14,55]

CD54

Intercellular adhesion molecule 1 (ICAM1)

(+) [14,23,24] (–) [19]

CD56

Neural cell adhesion molecule 1 (NCAM1)

(+) [56–58] (–) [15,24,33,40]

CD58

Lymphocyte-function associated antigen 3 (LFA-3)

(+) [14,19]

CD59

Membrane inhibitor of reactive lysis (MIRL); protectin

(+) [59]

CD61

Integrin β 3 chain

(+) [14] (low) [19,24]

CD62E

E-selectin

(–) [14,19]

CD62L

L-selectin

(+) [14]

CD62P

P-selectin

(–) [14,19]

CD63

TSPAN30

(+) [52]

CD71

Transferrin receptor

(+) [14]

II.  CELLULAR, MOLECULAR, AND DEVELOPMENTAL BIOLOGY OF BONE

131

Defining Skeletal Stem Cells

TABLE 7.1  Phenotypic Markers for Human Skeletal Stem Cell Identification—Cont’d Markers

Molecules

References (+) or (–)

CD73a

5'-nucleotidase, ecto

(+) [14,55,60–62]

CD90a

Thy-1 cell surface antigen

(+) [14,50,63,64] (low) [55]

CD102

Intercellular adhesion molecule 2 (ICAM2)

(+) [14] (–) [19]

CD104

Integrin β 4 chain

(+) [14] (low) [19]

CD105a

Endoglin

(+) [14–17,21–25,27,30–32,39–42, 44,49,59,61,62,65–73] (low) [55]

CD106

Vascular cell adhesion molecule 1 (VCAM1)

(+) [14,19,21,24,29,39,70,74–76]

CD109

Non-descript GPI-linked glycoprotein

(+) [18,77]

CD117

C-kit; Stem cell factor receptor

(–) [18] (–/+) [38]

CD119

Interferon gamma receptor 1 (IFNGR1)

(+) [14]

CD120a

Tumor necrosis factor receptor superfamily, member 1A

(+) [14]

CD120b

Tumor necrosis factor receptor superfamily, member 1B

(+) [14]

CD121a

Interleukin-1 receptor, type I (IL-1R)

(+) [14]

CD123

Interleukin-3 receptor, α (IL-3Rα)

(+) [14]

CD124

Interleukin-4 receptor (IL-4R)

(+) [14]

CD126

Interleukin-6 receptor (IL-6R)

(+) [14]

CD127

Interleukin-7 receptor (IL-7R)

(+) [14]

CD130

Interleukin-6 signal transducer (gp130); oncostatin M receptor

(+) [55]

CD133

Prominin 1

(–) [16,18,22,23,32] (+) [54]c

CD140a

Platelet-derived growth factor receptor, α polypeptide (PDGFRα)

(+) [18]

CD140b

Platelet-derived growth factor receptor, β polypeptide (PDGFRβ)

(+) [17,18]

CD144

Cadherin 5, type 2 (vascular endothelium) (VE-cadherin)

(–) [14,66,70]

CD146

Melanoma cell adhesion molecule (MCAM)

(+) [55,74,78]

CD164

Endolyn

(+) [18]

CD166

Activated leukocyte cell adhesion molecule (ALCAM)

(+) [14,21,23,28,31,41,52,79]

CD172a

Signal-regulatory protein α (SIRPA)

(+) [18]

CD178

Fas ligand

(–) [14]

CD200

Orexin (OX-2)

(+) [55]

CD235a

GlycophorinA

(–) [12,35,44,45,48]

CD271

Nerve growth factor receptor (NGFR)

(+) [17,46,56–58,71,80–83]

CD309

Vascular endothelial growth factor-receptor 2 (VEGF-R2)

(+) [26]

CD340

Erythroblastic leukemia viral oncogene homolog 2 (ERBB2)

(+) [17]

α-SMA

α-Smooth muscle actin

(+) [38,74,84]

ALDH

Aldehyde dehydrogenase

(bright) [85]d

CNN1

Calponin

(+) [84] (Continued)

II.  CELLULAR, MOLECULAR, AND DEVELOPMENTAL BIOLOGY OF BONE

132

7.  THE SKELETAL STEM CELL

TABLE 7.1  Phenotypic Markers for Human Skeletal Stem Cell Identification—Cont’d Markers

Molecules

References (+) or (–)

CDCP1

CUB domain-containing protein 1

(+) [86]

D7-FIB

Orphan antigen (fibroblast)

(+) [16,46]

DES

Desmin

(+) [84]

EGFR-3

Epidermal growth factor receptor 3

(–) [14]

FGFR

Fibroblast growth factor receptor

(+) [14]

GD2

Ganglioside

(+) [87]

HSP90β

Heat-shock protein 90 β

(+) [88]

NES

Nestin

(+) [18,89]

SSEA4

Stage-specific embryonic antigen

(+) [90]

Stro-1

Orphan antigen

(+) [12]

TGF-β1R

Transforming growth factor receptor β1 receptor

(+) [14]

TGF-β2R

Transforming growth factor receptor β2 receptor

(+) [14]

TNAP

Tissue nonspecific alkaline phosphatase

(+) [36,56–58] (–) [91]e

VIM

Vimentin

(+) [18,89]

vWF

Von Willebrand factor

(–) [14]

W5C5

Orphan antigen

(+) [17]

a 

Markers as part of the International Society for Cellular Therapy recommendations [92]. CD44 – Qian et al. argue that primary SSCs do not express CD44 and that the CD44-negative fraction contains the clonogenic activity and that in vitro culture results in expression of CD44. c  CD133 – Gindraux et al. suggest that native SSCs express CD133 while cultured SSCs lose expression. d  ALDH – assessed using aldefluor staining for enzyme activity. e  TNAP – Kim et al. suggest that true multipotent-SSC are TNAP negative, and as SSCs progress down osteogenesis they express TNAP. b 

This experimental system was used to test whether osteogenic stem/progenitor cells can traffic from one animal to a bone morphogenic protein-2 (BMP-2)-treated collagen scaffold implanted in the partner [115]. This experiment quite convincingly confirms that osteogenic cells indeed move via the circulation. To determine whether the intravascular migration of osteogenic stem/progenitor cells can be induced, studies have tested the impact of injury or growth factors. A dramatic increase in the number of circulating osteogenic stem/progenitor cells was noted when an animal was primed with vascular injury or high BMP stimulation [116]. These data suggest that inflammatory and chemoattractant signals may enhance the mobility of endogenous SSCs. In addition, the release of transforming growth factor (TGF)-β1 during bone resorption has been shown to induce the migration of skeletal stem/ progenitor cells to bone resorptive sites [117], thereby demonstrating that circulating skeletal stem/progenitor cells even participate in steady-state bone maintenance. Therefore, skeletal stem/progenitor cells do appear to move through the blood and may do so in an inducible manner in response to physiologic challenges.

KINETICS OF SKELETAL CELL TURNOVER Osteoblast Kinetics Early studies of osteoblastic cell kinetics were estimated by the labeling of proliferating cells and the persistence of labeled cells on growing bone surfaces before cells became embedded in matrix. This was done by simultaneous single dose injection of tritiated glycine (3H-glycine) and tritiated thymidine (3H-thymidine) into growing rabbits [118]. Glycine marked the newly produced extracellular matrix protein and therefore, the position of the bone surface and thymidine-labeled cells that made new deoxyribonucleic acid (DNA) as part of the cell replicative process. Autoradiographic analysis of femur bone sections revealed that labeled cells spend about 10 days on the bone surface before becoming embedded in the bone as osteocytes [118]. Later, a study defining the lifetime of osteoblasts in the mouse periodontium demonstrated that periodontal proliferating progenitors go through a single S-phase and become osteoblasts that last for about 20 days [119]. Although these studies did not

II.  CELLULAR, MOLECULAR, AND DEVELOPMENTAL BIOLOGY OF BONE

Kinetics of Skeletal Cell Turnover

133

FIGURE 7.1  Double immunostaining of CD34 (brown, peroxidase) and CD146 (red, alkaline phosphatase) in human bone marrow. While CD34 consistently labels the endothelial lining (A and B), CD146 is present in the subendothelial aspect of sinusoids (A) and arteries (B, C), and in loose microvascular-like structures (A). The presence of CD146 in mural cells composing the arteriolar wall is best appreciated in obliquely oriented vessels (B). C: red and brown rectangles at the extremities of a longitudinally oriented artery inscribe a rare individual CD146-positive (D) and a CD34-positive cell. Scale bars: A–C = 20 μm; D and E = 10 μm. (See color plate.) Source: Ferraro F.

FIGURE 7.2  Detection of nestin in human bone marrow. A: immunoperoxidase staining showing two adjacent cells labeled by nestin (brown) laying within the hematopoietic tissue (scale bar 50 μm). B: the elongated shape of nestin-positive cells is better appreciated at higher magnification of the same microscopic field as in A (scale bar 20 μm). C: red arrow points to a pericyte-like appearance of a nestin-positive cell located between two bone trabeculae (BT) and asterisk indicates a small nestin-positive cell. (See color plate.) Source: Ferraro F.

define with precision the cell types involved in proliferation and had limited ability to measure the turnover rate of nonproliferating cells, they strongly suggested that a fraction of cells in growing bones have a relatively short duration on the bone surface and rapidly become boneembedding osteocytes (10–20 days). Human osteoblast function and average lifespan have been determined independently. After double tetracycline labeling, dividing the total wall width (the amount of bone made by osteoblasts) by the mineral apposition rate was used to examine the osteoblast lifespan in humans. These very indirect studies concluded that the average lifespan of normal human osteoblasts is about 150 days [120,121], though a proportion of osteoblasts become

osteocytes during this process considerably shortening their life span as an osteoblast. The estimates of cell durability in these studies were based on a calculation derived from the measurements of new bone synthesis, which is highly variable in different types of bone and different species, possibly accounting for the wide difference seen in mouse and human studies. In addition, these studies did not precisely define cell types, broadly classifying all cuboidal cells localized along the bone surface as osteoblasts. However, despite the limitations of these studies and the discrepancies between species, the data indicated that osteoblasts have a limited lifespan and implied that they are postmitotic, therefore replaced by unspecified immature cells [65,66,69].

II.  CELLULAR, MOLECULAR, AND DEVELOPMENTAL BIOLOGY OF BONE

134

7.  THE SKELETAL STEM CELL

More modern techniques have taken advantage of the ability to label specific subsets of cells genetically at a particular time and follow them by sequential in vivo imaging [122,123]. This technology is obviously limited to mice at this time, but indicates that mature osteoblast populations have a high rate of basal turnover with a durability of about 60 days. These studies also evaluated the cell populations that replenish the osteoblasts and will be discussed below. Overall, it appears that mature osteoblasts in adult bones turn over rapidly in homeostasis

Osteoblasts are Postmitotic Osteoblasts are a short-lived cell and the basis for their replenishment is a topic of some interest in understanding how the tissue is organized as a system. Multiple studies have suggested the possibility that mature osteoblasts are capable of proliferation in bones exposed to stress conditions or under early developmental processes. For example, when mice were irradiated and proliferation of the cells along the endosteal surface assessed by 5-bromo-2-deoxyuridine (Brd-U) administration (a pyrimidine analog incorporated into newly synthesized DNA), over 10% of the ColI-expressing cells were Brd-U positive compared with about 2% of controls, suggesting a rapid expansion of the resident cell population [124]. Interestingly, most of the increases in ColI and Ocn-expressing cells were observed in the metaphyses of bones. Similar results have been reported in developing bones [125]. Therefore, under certain stress conditions and at particular ages, osteoblasts along endosteal surfaces appear to gain proliferative potential. However, the studies must be interpreted with caution as Brd-U labeling may occur in proliferating osteoprogenitors upstream of the mature osteoblasts, and the observed increase in the Brd-U-positive ColI-expressing osteoblasts could be the outcome of rapid differentiation from a progenitor pool, rather than from mature osteoblast proliferation. This point is raised in particular because in other studies using genetic marking where only mature, Ocn-expressing osteoblasts were labeled, no observable proliferation of mature osteoblasts was noted [96]. This was confirmed in long-term homeostasis in adult animals and in models of injury providing fairly definitive evidence for the osteoblast as a postmitotic cell [96]. However, it cannot be excluded that under particular kinds of stress not tested in the model or at particular ages, a subset of mature osteolineage cells can divide.

Turnover of Other Osteolineage Cell Types Evaluation of what cells do divide to replace osteoblasts in part depends upon the ability to define osteolineage

cells. In bone development, osteoblast differentiation from osteogenic progenitors can be distinguished by changes in stage-specific genes. Runx2 is one of the earliest genetic markers of osteo/chondrogenic progenitors, followed by osterix (Osx) in osteoprogenitors and ColI and Ocn in maturing osteoblastic cells [126]. Unfortunately, in adult animals, these genetic markers do not correlate with reliable surface-expressed phenotypic markers to label skeletal stem and progenitor cells clearly. Furthermore, while the temporal expression of these genetic markers makes it tempting to place these populations of cells in a linear relationship to each other, with Runx2 expression representing an early stem cell to Ocn expression representing a mature cell, with various restricted progenitors inbetween, the relationship amongst these populations may be more complex. Therefore, it is difficult to measure definitively the subsets of cells involved in the replenishment of osteoblasts in adults. However, a number of studies have provided useful information. The anatomic sites where bone-forming cells are thought to reside include the inner layer of the periosteum (inner cambial layer) [127], the endosteum [1], osteonal (Harvarsian) canals, and perivascular locations within bone marrow [95]. Engineered mice where cells can be fluorescently tagged at a particular time and followed kinetically (so called cellular pulse-chase experiments) have provided information as to which cells divide and replace mature cells in adult animals. In the example provided above, the promoter that is only activated at the level of the mature osteoblast (Ocn) was used to drive activation of a cell label and indicated that Ocn-expressing osteoblasts do not divide and do not replace themselves [96]. Rather, they appeared to be replaced by cells expressing Osx as evident by another similarly engineered mouse using the Osx promoter to drive a fluorescent tag. The Osx-expressing cells were also minimally proliferative however, as indicated by Ki-67 staining. Therefore, a more primitive population of cells appeared to be required. These could be marked by induction of Mx1 in mice engineered such that the Mx1 promoter would activate a fluorescent tag [96]. These Mx1-labeled cells generated new osteoblasts under settings of both homeostasis and after microfracture. Mx1-labeled stem/progenitor cells appeared at the site of fracture within 2–5 days and dramatically increased in number at 12 days. They did differentiate into fully mature osteoblasts as evident by Ocn expression and the ability to lay down new bone in vivo. Similar dynamics and contributions of osteogenic progenitors have been observed in injury models using a smooth muscle α-actin promoter (αSMA) transgenic mouse model [128], another promoter also hypothesized to denote SSCs. Therefore, osteoblast replenishment in adults appears to be mediated by the stem/progenitor cell pool without evidence for mature cells replacing other mature cells.

II.  CELLULAR, MOLECULAR, AND DEVELOPMENTAL BIOLOGY OF BONE

Potential Use of Skeletal Stem Cells as Therapy

POTENTIAL USE OF SKELETAL STEM CELLS AS THERAPY If bone cells depend upon a dynamic population of SSCs that can migrate interstitially and by the circulation [96], then it is biologically feasible to consider cellbased bone therapies. This issue has prompted much of the attention on the putative SSC that has been called MSC by in vitro features as discussed above. While the in vitro cultured population that is termed MSC or here, MC, may not have close correspondence with bona fide endogenous SSCs, it may be valuable as a source of cells with osteolineage potential. The latter has been extensively shown through the use of the cells in vitro in bone constructs, in vivo by homotopic transplant into bone, and even by systemic administration of an allogeneic bone marrow graft in children with osteogenesis imperfecta (OI) [129]. Several aspects of the biology of these cells are important considerations for cell therapy. Practical clinical utility of cell therapy will likely require intravenous infusion and SSC homing to the bone marrow microenvironment although several animal models of osteoporosis have shown some success with intrabone injections of MC [130,131]. The homing ability of primary or ex vivo expanded MC has been rigorously studied [94,132,133]. These studies used varying approaches to isolate and expand MCs in vitro and found that the engraftment efficiency toward bone marrow or target tissues of ex vivo expanded MC was variable and dependent on the source of cells, prior culture conditions, passage number, and route of delivery. In general, studies utilizing cultured MC showed poor engraftment in target tissues after systemic administration, confirmed by a comparative study demonstrating that primary murine SSCs have significantly higher efficiency of homing to the bone marrow compared to cultured MC [133]. The downregulation of cell adhesion molecules and/or chemokine receptors (CXCR4, receptor for stromal cell-derived factor (SDF)-1α) in cultureexpanded MC is a possible cause of poor migration and homing. To overcome these limitations of SSCs to home and migrate appropriately in vivo, and thus improve the engraftment of SSCs in a potential clinical setting, multiple approaches have been employed to enhance the homing response of MC. Since the SDF-1α and CXCR4 axis is a key mechanism of SSC homing, retroviral overexpression of CXCR4 in MC resulted in increased MC homing to inflammatory sites or ischemic myocardium [134,135]. Similarly, studies employing either an adenoassociated viral vector (AAV) to transiently cause ectopic expression of the α4 integrin on murine MC [136] or by chemical modification of the cell surface with a peptidomimetic ligand against α4 integrin on the MC cell surface coupled to bisphosphonate [137], leads to substantially increased homing of MC into the bone marrow

135

of recipient mice and a greater than 10-fold increase in new bone formation compared to control. Sackstein et al., using human MC preparations, chemically modified the cell surface of MC via an α-1,3-fucosyltransferase preparation to convert the native CD44 glycoform into hematopoietic cell E-selectin/L-selectin ligand (HCELL) [138]. This allowed for the chemically modified MC to have potent E-selectin binding without effects on cell viability or multipotency. When transplanted into immunocompromised mice, the chemically modified MC were capable of infiltrating the marrow within hours of infusion, with foci of endosteally localized cells and human osteoid generation [138]. These novel approaches may eventually lead to therapeutic strategies allowing for the prospective harvesting of SSCs and directed targeting of those cells to sites of injury or disease to coordinate bone formation and repair. As continued advancements are made in directing SSCs to the bone marrow after systemic delivery, the next phase of clinical therapy will be how best to apply SSCs to treat disease. SSCs from aged patients or donors are undoubtedly reduced in number and have a reduced capacity for osteogenic potential [139–142], and hence therapeutic efficacy, compared to younger SSCs. Therefore, autologous transplantation of SSCs, without modification, may have little long-term value. Peripheral, circulating osteogenic-SSCs are of five times greater abundance in adolescents than the aged, and are also present in higher numbers after injury [114]. In a mouse study, aged mice transplanted with MC from young donors had increased bone density and a longer period of survival [142]. Therefore, allogeneic transplantation from younger, healthier donors might be preferred, but graft tolerance and immunosuppression issues create additional complications. Therefore, if SSCs from patients can be modified or corrected, then autologous transplantation of modified cells, absent of graft rejection issues, may represent the best option. One example where this approach is moving toward clinical testing is in the treatment of OI. Clinical trials using allogeneic bone marrow transplants or SSC-enriched transplants have demonstrated improvement in children with OI [129,143,144]. Given that OI is normally associated with a mutation in a single gene, ColI, it is a highly attractive target for gene therapy. Using an AAV approach, Chamberlain and colleagues have demonstrated the ability to correct gene defects in OI patient SSCs and restore osteogenic potential in vitro [145,146]. In a similar approach, the laboratory-generated induced pluripotent stem cells (iPSCs) from AAVcorrected SSCs and demonstrated that they were able to form bone in immunocompromised mice [147]. The transformation to an iPSC state theoretically allows for unlimited expansion of the cells before differentiation to acquire sufficient numbers for a robust transplant, and

II.  CELLULAR, MOLECULAR, AND DEVELOPMENTAL BIOLOGY OF BONE

136

7.  THE SKELETAL STEM CELL

the iPSC reprogramming transgenes could be removed by activating Cre recombinase [147]. As gene-targeting therapies evolve, and our understanding of the genetic and molecular pathways governing bone degenerative diseases increases, a combinatorial gene therapy/SSC transplant strategy may offer new opportunities for bone therapeutics.

INTERACTIONS WITH THE HEMATOPOIETIC SYSTEM The Endosteal Hematopoietic Niche The bone marrow has long been recognized as the primary source of hematopoiesis in adult mammals. The notion of a bone marrow transplant relies on the fact that bone is a source of HSPCs capable of repopulating myeloablated hosts. Intriguingly, one of the studies describing cells that behaved like osteogenic-SSCs was actually seeking to determine why hematopoiesis was located in the bone [5]. Tavassoli and Crosby placed bone marrow fragments into nonbony tissues to see if the addition on nonhematopoietic bone cells could enable engraftment of hematopoietic cells in those tissues. Hematopoiesis occurred only if a bony ossicle with microcirculation formed. They observed that a “reticular” cell that could differentiate into osteoblasts was required for the bone and bone marrow formation, thus presaging the recognition of a bone-forming SSC and an important relationship between bone and hematopoiesis. Within bone marrow, the endosteal surfaces of bone were recognized as a rich source of primitive hematopoietic cells by studies where tissue at progressive distances from the endosteum was preserved and tested [148,149]. These studies suggested that endosteum was a site of enrichment for hematopoietic stem/progenitor cells and therefore, the location of the HSC niche. This issue is controversial, however. A number of histologic studies have indicated that HSC are perivascular in the unmanipulated mouse bone marrow (see below). This may be different from the bone marrow in the setting of stem cell transplantation. Several different imaging approaches have demonstrated that transplanted HSCs localize to endosteal surfaces, while more mature hematopoietic progenitor cells (HPCs) localize further away [122,150–152]. Hematopoietic stem cells that were closely associated with the endosteum were also shown to have greater transplantation activity than those HSC contained in the central marrow cavity [153]. Given that osteolineage cells line the endosteum, they were theorized as a potential supportive mediator of the HSC niche. When HSCs were acquired from aged donors, it was found that they localize to sites further away from the endosteum than HSCs from younger donors [150], suggesting

that HSC localization within the bone may reflect the difference in HSC function observed with the aging process. Studies by Taichman and colleagues [154,155] demonstrated that osteolineage cells produce many supportive growth factors including granulocyte colony stimulating factor (G-CSF), granulocyte macrophage colony stimulating factor (GM-CSF), interleukin (IL)1, IL-6, and TGF-β. Later, in vivo studies using various genetic approaches demonstrated that osteolineage cells are a significant regulatory component of hematopoiesis [156–159]. In one of these studies where a constitutively active parathyroid hormone (PTH)/parathyroid hormone-related protein (PTHrP) receptor was driven by an osteolineage promoter, the effect of osteolineage cells on HSC expansion was mediated, at least in part, by the Notch ligand Jagged1. Notch signaling regulates cell fate decisions including HSC self-renewal [160–162], thus by changing self-renewal versus differentiation decisions, Notch can increase HSC number without differentiation or increase in HPC or mature cells [160,162]. This regulatory relationship of Notch ligand expression by osteolineage cells may only be important with PTH/PTHrP receptor activation, however, as studies on Notch in homeostasis in adult animals do not support an important role in HSC function [163]. Within the endosteal niche, HSCs are thought to be retained through a variety of adhesion molecule interactions, many of which are likely redundant systems. Early studies exploring the role of osteolineage cells in maintaining HSCs suggested that N-cadherin interactions mediated the positive effects on HSCs [157]; however, more recent studies have contradicted these findings [164,165]. Numerous other adhesion molecules have been implicated as contributing to HSC and HPC tethering, including, but not limited to, the integrins α4β1 – very late antigen (VLA)-4 [166–171], α5β1 –VLA-5 [167,168,170,172], α4β7 – lymphocyte Peyer's patch adhesion molecule-1 (LPAM-1) [173], the alfa 6 integrins (laminins) [174,175], CD44 [167,176], E-selectins [177–179], the angiopoietin receptor tyrosine kinase with immunoglobulin-like and EGF-like domains-2 (Tie-2) [159], endolyn (CD164) [180], the calcium-sensing receptor (CaR) [181], stromal-derived factor-1α (SDF-1α) [182], and osteopontin (OPN) [183,184]. Intriguingly, OPN is also a negative regulator of HSC pool size within the bone marrow niche [184,185], and knockout of OPN in mice results in endogenous hematopoietic mobilization to the periphery and increases the mobilization response to G-CSF [183]. In 2011, osteolineage cells were also found to express agrin, which mediates cell–cell contact with short-term HSCs and initiates proliferation, perhaps functioning as an opposing signal to OPN [186]. These results suggest that OPN (a target for modulation of the endosteal HSC niche) and agrin receptors (targets on HSCs) may serve as future therapeutic strategies

II.  CELLULAR, MOLECULAR, AND DEVELOPMENTAL BIOLOGY OF BONE

Interactions with the Hematopoietic System

to modify and enhance hematopoietic transplant and recovery, in line with previously described strategies targeting other adhesion interactions in the niche [187–192].

Hematopoietic Regulation of the Endosteal Niche Up to this point, the focus has been on the role of osteolineage cells and their associated regulation of hematopoietic cells. However, cells of hematopoietic origin also support and regulate osteolineage cells. Cell–cell contact between osteolineage cells and HPC has been shown to be important for HPC survival [193]. Using an in vitro system, Gillette et al. demonstrated that human CD34+ cells (an HPC/HSC population of cells) co-cultured with osteolineage cells made cell–cell contact and exchanged a portion of their membrane with the osteolineage cells, creating a signaling endosome [194]. This signaling endosome caused osteolineage cells to downregulate Smad signaling and increase production of SDF-1α, suggesting that hematopoietic cells may instruct osteolineage cells to create a more habitable environment, allowing them to affect their own microenvironment directly. These results may suggest that HSCs help to create their own niches, perhaps by instructing neighboring stromal cells to produce supportive factors. Mature hematopoietically derived cells also have the ability to alter the endosteal lining osteolineage cells. One therapeutic manipulation of the hematopoietic niche involves treatment with G-CSF to “mobilize” HSCs and HPCs out of the bone marrow and into the peripheral blood, where they can subsequently be collected by apheresis and used in hematopoietic transplantation. Treatment with G-CSF causes significant suppression of osteolineage cells [195–197], causing apoptosis [196] and a characteristic “flattening” of osteolineage cells [197]. In 2010, Winkler et al. [195] reported that a population of macrophage cells, characterized as F4/80+ Ly-6 G+ CD11b+ and termed “osteomacs” [198], line the endosteal surfaces of bone and that G-CSF treatment resulted in a trafficking, and reduction in number, of osteomacs [195]. Presumably, this reduction in osteomacs was responsible for the attenuation of function of osteolineage cells. To confirm this hypothesis, macrophages were depleted using Mafia transgenic mice or with clodronate-loaded liposomes (Clo-lip) treatment, causing significant mobilization of HSC and HPC. A similar report from Chow et al. also demonstrated that depletion of macrophages with Clo-lip treatment results in hematopoietic mobilization [199], describing a Gr-1negative F4/80+ CD115mid CD169+ macrophage population as a regulator of osteolineage cells. Similar to the osteomac population, the authors suggest that the CD169+ macrophages express a soluble, yet to be identified, factor(s) that positively supports niche cells. In comparable studies by Christopher et al., a soluble factor produced by

137

monocyte/macrophage populations supported osteolineage function [200], suggesting that manipulation of monocyte/macrophage populations or the factor(s) produced by them is a future therapeutic strategy to manipulate the endosteal niche, and possibly suggest a novel strategy to affect bone cells therapeutically. Osteoclasts are also monocyte-derived cells that are present along endosteal surfaces of bone. Osteoblasts and osteoclasts regulate bone formation and bone resorption, respectively, within the bone marrow niche, and pre-osteoclasts cause retraction of osteoblasts [201], theoretically creating transient “holes” in the HSC supporting endosteal niche. Given that treatment with G-CSF causes increases in osteoclast number and activity [202,203], it was hypothesized that osteoclasts may regulate the endosteal niche and alter HSC retention and localization. Kollet et al. treated mice with RANK ligand, which increased osteoclast activity and correlated with a moderate increase in HPC mobilization [204], consistent with an independent report [205]. Correspondingly, stress models such as bleeding or LPS treatment also increased the number of osteoclasts in the endosteal niche and HPC in the peripheral blood, supporting a role for osteoclasts in niche regulation. Kollet et al. suggested that osteoclast-derived proteolytic enzymes, such as cathepsin K, degraded important niche retention molecules, thereby facilitating egress of HSC and HPC [204]. A more recent study by the same laboratory showed decreased osteoclast maturation and activity in CD45 knockout mice, with reduced mobilization to RANK ligand and G-CSF [206], further suggesting osteoclasts are important in regulation of the endosteal niche. In contrast to these studies, Takamatsu et al. earlier reported that while G-CSF treatment increased osteoclast number and bone resorption in both BALB/c mice and humans, the increase in osteoclasts did not occur until 10–15 days or 6–8 days, respectively, after treatment with G-CSF [203], which has also been observed by other groups using similar systems [207,208]. Since niche clearance by G-CSF is typically observed 4 to 5 days after treatment, the importance of osteoclasts remains unclear. Similarly, treatment of mice with osteoclast-inhibiting bisphosphonates does not result in an impaired mobilization response to G-CSF [195,203,209], and in fact increases HSC egress [195,209]. Intriguingly, the endosteal surface of bone, particularly underneath resorbing osteoclasts, is a significant source of soluble extracellular calcium and previous studies by our laboratory demonstrated that HSC expression of calcium-sensing receptors mediates a chemoattraction to soluble Ca2+ [181]. When the calciumsensing receptor was knocked out, mice had reduced HSC content within the bone marrow niche and increased HSC in peripheral blood. These results might suggest that the bone resorbing, and subsequent Ca2+ releasing activity of osteoclasts, may be involved in niche retention.

II.  CELLULAR, MOLECULAR, AND DEVELOPMENTAL BIOLOGY OF BONE

138

7.  THE SKELETAL STEM CELL

Hematopoietic transplantation normally requires conditioning of the bone marrow niche, or a clearance of resident HSC and HPC, to allow for successful engraftment. Conditioning is normally accomplished by treatment with chemotherapeutic agents and/or irradiation to remove HSC and HPC; however, these agents can also affect niche cells themselves. A study from Dominici et al. demonstrated that following a lethal dose of irradiation, megakaryocytes migrated toward the endosteal surfaces of bones and induced a two-fold expansion in osteolineage cells, presumably increasing the available niches for subsequent HSC engraftment [124]. These results are in close alignment with other studies in which mice deficient in GATA-1 and NF-E2 transcription factors had significantly increased megakaryocyte production, leading to a six-fold expansion of osteolineage cells [210], a process requiring direct cell– cell contact between megakaryocytes and osteolineage cells [211,212]. Further support for blood cells affecting bone biology are the observations that excessive blood loss, causing stress on the hematopoietic system, leads to osteoporosis in mice [213] and that splenectomy in mice results in an expansion of megakaryocytes with reduced GATA-1 expression and increases in bone formation [214]. These intriguing findings suggest that there is much yet to be learned about the bidirectional regulatory relationships of blood and bone. Further exploration of these is clearly warranted particularly since there is a substantial prevalence of bone disorders following hematopoietic transplantation and its associated conditioning regimens [215–222].

Endosteal Age and Turnover The endosteal surface of bone, as described earlier, is a dynamic tissue with a high rate of turnover of osteoblasts, which are replenished by earlier osteogenic precursors and SSCs. Several reports, using in vitro co-culture studies have shown that HSCs cultured on more immature osteolineage cells have a higher activity and repopulating ability than HSCs cultured on more mature osteoblasts [223,224], suggesting that the maturational status of osteolineage cells may be a key determinate of HSC niche support. Indeed, several in vivo reports described niche cells responsible for HSC maintenance that resembled SSCs and their immediate downstream precursors [10,225], and in studies in which terminally differentiated osteoblasts were ablated, hematopoiesis was unaffected [226,227]. These findings suggest that the HSC niche is composed of an immature osteolineage cell subset. The aforementioned Nestin+ multipotent-SSCs express high levels of hematopoietic supportive molecules, suggesting that these cells may form a unique bone marrow niche [95] compromised of heterologous stem cell pairs. Intriguingly, these Nestin+

cells were found to wrap around endothelial cells within the bone marrow near the endosteum, but also toward the central marrow, suggesting an alternate HSC niche localization further from the endosteum.

Vascular and Perivascular Cells – the Vascular Niche While it is clear that HSCs reside and thrive within the endosteal bone marrow niche, evidence, including the localization of CXCL12-abundant reticular cells [225,228] and Nestin+ SSCs [95], suggests that the endosteum is not the exclusive and may not even be the predominant niche for hematopoiesis. Stem cell localization studies utilizing in vivo imaging out of our laboratory [122] and others [152] found that while HSCs were near the endosteum, they were not exclusively adjacent to osteolineage cells. Studies assessing the localization of signaling lymphocyte activation molecule (SLAM; CD150+, CD48–, and CD244–) cells, which are highly enriched for HSC, have shown a greater proportion of SLAM cells adjacent to bone marrow sinusoidal blood vessels, suggesting a “vascular niche” for HSC [165,229]. This is supported by the fact that endothelial cells, very similarly to osteolineage cells, can support hematopoiesis both in vitro and in vivo [230–232]. We have also described small endothelial microdomains that express E-selectin and SDF-1α, and act as a point of entry for HSC after transplantation [123]. Regeneration of sinusoidal endothelial cells has been reported to be essential for hematopoietic reconstitution following myelosuppression [233], and bone marrow endothelial cells have been reported to support the growth and expansion of HSC through angiocrine factors [234]. Interestingly, similar to the HSC cell-to-cell contact with osteolineage cells altering the endosteal niche function [194], Slayton et al. reported that donor hematopoietic cells migrated to sites of vascular sinusoidal injury within bones and contributed to repair following transplantation [235], further highlighting the complexity and interconnectedness of the bone marrow microenvironment. While it is not completely clear what factors influence the particular niche location of HSCs, the fact that reticular cells with high production of SDF-1α are present in both the endosteal and vascular niches, may provide a common mechanism of HSC support in both niches [225,228,236]. One report has demonstrated that HSC, shortly after transplantation into nonmyeloablated recipients, preferentially localize to the endosteum in metaphyseal regions of bone in close association with blood vessels, and that this requires hyaluronic acid [237]. Given that the endosteal region is itself highly vascularized may suggest that the HSC niche actually encompasses overlapping regions within the bone.

II.  CELLULAR, MOLECULAR, AND DEVELOPMENTAL BIOLOGY OF BONE

Interactions with the Hematopoietic System

The Bone Marrow Environment is Hypoxic (see Chapter 8 for effects of hypoxia on osteoclasts) Several reports have demonstrated that HSC are present in regions of hypoxia within the bone marrow [238– 240], and those HSC contained in hypoxic areas have greater hematopoietic repopulating ability than those in more perfused areas [241], suggesting that perhaps the overlapping regions of the endosteal and vascular biomes contains the right mix of atmospheric (oxygen and other chemical mediators) conditions to support hematopoiesis. Cellular exposure to hypoxia results in stabilization of hypoxia inducible factor 1-α (HIF-1α), which is a transcriptional regulator of erythropoietin (EPO) production [242]; numerous cell proliferation and survival genes [243–245]; the angiogenic growth factor, vascular endothelial growth factor (VEGF) [246]; and other genes. In mice where the HIF-1α responsive element is mutated on the Vegfa promoter, thereby preventing upregulated expression in hypoxic conditions, HSCs were found to have defects in their function [247], supporting a role of hypoxia-induced gene expression in the niche for maintenance of HSCs. HIF-1α also increases production of SDF-1α [248] and CXCR4 receptor expression [249], and prevents hematopoietic cell damage caused by overproduction of reactive oxygen species (ROS) [250]. Therefore, HSCs residing in areas of low oxygen will have increased activity of HIF-1α, thereby maintaining their fitness and stemness [251], a hypothesis that is supported by the fact that in vitro hypoxic conditions expand human HSC [252] and HPC populations [253–255]. In 2010, using HIF-1α deficient mice, Takubo and colleagues demonstrated that HSC cell cycle quiescence was lost, and HSC numbers diminished after stressful insults [256]. Additional studies exploring the protein stabilization of HIF-1α demonstrated that a precise regulation of HIF-1α activity was necessary for optimal HSC function. Miharada et al. also reported that HIF-1α-deficient mice have reduced expression of Cripto, also known as teratocarcinoma-derived growth factor-1 (TDGF-1), on endosteal osteolineage cells and a reduction of one of Cripto's cognate receptors, GRP78, on HSCs [257]. The authors demonstrated that the Cripto/ GRP78 signaling axis is an important regulator of HSC quiescence downstream of HIF-1α signaling, and postulated that GRP78 can be used as a marker to distinguish between quiescent and active HSCs. Advanced technologies that are able to measure oxygen tension accurately within the bone marrow microenvironment at high resolution overlaid with imaging assessing HSC localization are likely to be highly informative, and perhaps will resolve some discrepancies and unanswered questions as to the specifications of the HSC niche. Oxygen tension is also important in regulating osteolineage cell function. Bone marrow MSCs cultured in hypoxic conditions demonstrated increased self-renewal division

139

and increased integrin expression allowing for more robust migration compared to their normoxic-cultured controls [258], consistent with reports that hypoxic conditioning can increase osteogenic, adipogenic, and chondrogenic differentiation potential [259–263], though other reports propose opposite effects [264] suggesting that regulation of SSCs by hypoxia may be highly dynamic. One report has also demonstrated that VEGF production by osteolineage cells is cooperatively regulated by Osx and Hif-1α [265], suggesting hematopoietic and angiogenic support by osteolineage cells is also regulated by oxygen tension. Further exploration into these mechanisms is likely to lead to advancements in therapies, particularly in terms of bioengineering scaffolds and ex vivo culture conditions for cellular therapeutics of bone degeneration [262,266–268].

Nervous System Mediators While it is clear that cellular and molecular constituents within the microenvironment interact and coregulate each other, the question arises as to whether there is a global regulator of the entire bone system, able to alter numerous components of the niche to respond as needed rapidly. Nestin+ MSCs are highly intriguing, as nestin, an intermediate filament protein, is normally restricted to nerve cells. Intriguingly, work by the same laboratory demonstrated that HSC and HPC mobilization by G-CSF required peripheral β2-adrenergic signals [197] in studies utilizing chemically sympathectomized mice treated with 6-hydroxydopamine; mice treated with the β-blocker propranolol; or mice genetically deficient in the gene for dopamine β-hydroxylase (Dbh), an enzyme that converts dopamine into norepinephrine. They also showed that treatment with the β2-adrenergic agonist clenbuterol reversed the phenotype of Dbh knockout mice [197]. The Frenette laboratory also demonstrated that the nervous system regulates HSC niche retention via circadian rhythms [269,270]. Signaling via β3-adrenergic stimulations causes rhythmic oscillations controlling norepinephrine release, CXCR4 expression, and SDF-1α production, causing regular transient trafficking from the bone marrow niche. Nervous system signaling has also been demonstrated to have direct effects on HSC, as human CD34+ hematopoietic cells express β2-adrenergic and dopamine receptors and neurotransmitters, such as norepinephrine, serve as direct chemoattractants for HSC [271], demonstrating nervous system global control over multiple cellular components of the hematopoietic ecologic system. One report showed that β2-adrenergic signaling causes upregulation of the vitamin D receptor (VDR) on osteolineage cells and that VDR signaling was necessary for osteolineage responses to G-CSF treatment [272]. VDR expression is also regulated by circadian rhythms [273] demonstrating further

II.  CELLULAR, MOLECULAR, AND DEVELOPMENTAL BIOLOGY OF BONE

140

7.  THE SKELETAL STEM CELL

interconnectivity of the nervous system control over the bone microenvironmental system. Yamazaki et al. described a population of nonmyelinating Schwann cells that ensheath sympathetic nerves in the bone marrow [274]. These cells were a primary producer of TGF-β, which they and others have demonstrated is a quiescent signal for HSC [275,276], and when the bone was denervated, TGF-β signaling was reduced leading to a reduction in the HSC pool [274]. Given that TGF-β is also capable of attracting SSCs [117], this may suggest that the nervous system can coordinate SSC trafficking and migration to sites of need, though further exploration is required.

SUMMARY The relationship of bone and bone marrow remains a highly intriguing biologic paradigm for how heterologous cells interrelate to accomplish complex and highly divergent physiologic processes. While the hematopoietic system has been more accessible and more extensively studied in terms of cellular hierarchies and stem cell function, applying approaches and concepts learned from it have again revealed marked connectedness between bone and bone marrow. The bone, like the blood, appears to be organized in a hierarchical manner where stem and progenitor cells are required for replenishment of short-lived mature cells. Indeed, the turnover times for mature bone cells, osteoblasts, and mature red cells are similar in both mouse and man. In addition, both skeletal and hematopoietic stem cell compartments appear to be heterogeneous and both stem cell populations appear capable of migration, particularly through the vascular tree. Further exploration of the similarities and differences between these systems promises to both teach important cell and organismal biology and to offer the potential of defining novel therapeutic strategies for diseases in each system.

References [1]  Bianco P, Robey PG, Simmons PJ. Mesenchymal stem cells: revisiting history, concepts, and assays. Cell Stem Cell 2008;2:313–9. [2]  Larsen WJ. Human Embryology. 2nd ed. Philadelphia: W.B. Saunders Company; 1997. [3]  Steele DG, Bramblett CA. The Anatomy and Biology of the Human Skeleton. 1st ed. College Station, TX: Texas A&M University Press; 1988. [4]  Friedenstein AJ, Petrakova KV, Kurolesova AI, Frolova GP. Heterotopic of bone marrow. Analysis of precursor cells for osteogenic and hematopoietic tissues. Transplantation 1968;6:230–47. [5]  Tavassoli M, Crosby WH. Transplantation of marrow to extramedullary sites. Science 1968;161:54–6. [6]  Friedenstein AJ, Chailakhjan RK, Lalykina KS. The development of fibroblast colonies in monolayer cultures of guinea-pig bone marrow and spleen cells. Cell Tissue Kinet 1970;3:393–403.

[7]  Friedenstein AJ, Latzinik NW, Grosheva AG, Gorskaya UF. Marrow microenvironment transfer by heterotopic transplantation of freshly isolated and cultured cells in porous sponges. Exp Hematol 1982;10:217–27. [8]  Friedenstein AJ, Chailakhyan RK, Gerasimov UV. Bone marrow osteogenic stem cells: in vitro cultivation and transplantation in diffusion chambers. Cell Tissue Kinet 1987;20:263–72. [9]  Friedenstein AJ, Chailakhyan RK, Latsinik NV, Panasyuk AF, Keiliss-Borok IV. Stromal cells responsible for transferring the microenvironment of the hemopoietic tissues. Cloning in vitro and retransplantation in vivo. Transplantation 1974;17:331–40. [10] Sacchetti B, Funari A, Michienzi S, Di CS, Piersanti S, Saggio I, et al. Self-renewing osteoprogenitors in bone marrow sinusoids can organize a hematopoietic microenvironment. Cell 2007;131:324–36. [11]  Gronthos S, Graves SE, Ohta S, Simmons PJ. The STRO-1+ fraction of adult human bone marrow contains the osteogenic precursors. Blood 1994;84:4164–73. [12] Simmons PJ, Torok-Storb B. Identification of stromal cell precursors in human bone marrow by a novel monoclonal antibody, STRO-1. Blood 1991;78:55–62. [13] Robledo MM, Hidalgo A, Lastres P, Arroyo AG, Bernabeu C, Sanchez-Madrid F, et al. Characterization of TGF-beta 1-binding proteins in human bone marrow stromal cells. Br J Haematol 1996;93:507–14. [14] Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, ­Mosca JD, et al. Multilineage potential of adult human mesenchymal stem cells. Science 1999;284:143–7. [15] Schallmoser K, Bartmann C, Rohde E, Reinisch A, Kashofer K, ­Stadelmeyer E, et al. Human platelet lysate can replace fetal ­bovine serum for clinical-scale expansion of functional mesenchymal stromal cells. Transfusion 2007;47:1436–46. [16] Jones EA, Kinsey SE, English A, Jones RA, Straszynski L, ­Meredith DM, et al. Isolation and characterization of bone marrow multipotential mesenchymal progenitor cells. Arthritis Rheum 2002;46:3349–60. [17] Buhring HJ, Battula VL, Treml S, Schewe B, Kanz L, Vogel W. Novel markers for the prospective isolation of human MSC. Ann N Y Acad Sci 2007;1106:262–71. [18] Vogel W, Grunebach F, Messam CA, Kanz L, Brugger W, Buhring HJ. Heterogeneity among human bone marrow-derived mesenchymal stem cells and neural progenitor cells. Haematologica 2003;88:126–33. [19] Majumdar MK, Keane-Moore M, Buyaner D, Hardy WB, ­Moorman MA, McIntosh KR, et al. Characterization and functionality of cell surface molecules on human mesenchymal stem cells. J Biomed Sci 2003;10:228–41. [20] Meirelles LS, Nardi NB. Murine marrow-derived mesenchymal stem cell: isolation, in vitro expansion, and characterization. Br J Haematol 2003;123:702–11. [21] Lu LL, Liu YJ, Yang SG, Zhao QJ, Wang X, Gong W, et al. Isolation and characterization of human umbilical cord mesenchymal stem cells with hematopoiesis-supportive function and other potentials. Haematologica 2006;91:1017–26. [22] Bartmann C, Rohde E, Schallmoser K, Purstner P, Lanzer G, Linkesch W, et al. Two steps to functional mesenchymal stromal cells for clinical application. Transfusion 2007;47:1426–35. [23] Soncini M, Vertua E, Gibelli L, Zorzi F, Denegri M, Albertini A, et al. Isolation and characterization of mesenchymal cells from human fetal membranes. J Tissue Eng Regen Med 2007;1:296–305. [24] Conget PA, Minguell JJ. Phenotypical and functional properties of human bone marrow mesenchymal progenitor cells. J Cell Physiol 1999;181:67–73. [25] Mareschi K, Ferrero I, Rustichelli D, Aschero S, Gammaitoni L, Aglietta M, et al. Expansion of mesenchymal stem cells isolated from pediatric and adult donor bone marrow. J Cell Biochem 2006;97:744–54.

II.  CELLULAR, MOLECULAR, AND DEVELOPMENTAL BIOLOGY OF BONE

Summary

[26] Liu L, Sun Z, Chen B, Han Q, Liao L, Jia M, et al. Ex vivo expansion and in vivo infusion of bone marrow-derived Flk-1+CD31CD34- mesenchymal stem cells: feasibility and safety from monkey to human. Stem Cells Dev 2006;15:349–57. [27] Tondreau T, Meuleman N, Delforge A, Dejeneffe M, Leroy R, Massy M, et al. Mesenchymal stem cells derived from CD133positive cells in mobilized peripheral blood and cord blood: proliferation, Oct4 expression, and plasticity. Stem Cells 2005;23:1105–12. [28] Guo Z, Yang J, Liu X, Li X, Hou C, Tang PH, et al. Biological ­features of mesenchymal stem cells from human bone marrow. Chin Med J (Engl) 2001;114:950–3. [29] Gronthos S, Mankani M, Brahim J, Robey PG, Shi S. Postnatal ­human dental pulp stem cells (DPSCs) in vitro and in vivo. Proc Natl Acad Sci U. S. A. 2000;97:13625–30. [30] Sotiropoulou PA, Perez SA, Salagianni M, Baxevanis CN, ­Papamichail M. Characterization of the optimal culture conditions for clinical scale production of human mesenchymal stem cells. Stem Cells 2006;24:462–71. [31] Mareddy S, Crawford R, Brooke G, Xiao Y. Clonal isolation and characterization of bone marrow stromal cells from patients with osteoarthritis. Tissue Eng 2007;13:819–29. [32] Lodie TA, Blickarz CE, Devarakonda TJ, He C, Dash AB, Clarke J, et al. Systematic analysis of reportedly distinct populations of multipotent bone marrow-derived stem cells reveals a lack of distinction. Tissue Eng 2002;8:739–51. [33] Crisan M, Yap S, Casteilla L, Chen CW, Corselli M, Park TS, et al. A perivascular origin for mesenchymal stem cells in multiple human organs. Cell Stem Cell 2008;3:301–13. [34] Huss R. Perspectives on the morphology and biology of CD34negative stem cells. J Hematother Stem Cell Res 2000;9:783–93. [35] Zannettino AC, Paton S, Kortesidis A, Khor F, Itescu S, Gronthos S. Human mulipotential mesenchymal/stromal stem cells are derived from a discrete subpopulation of STRO-1bright/CD34/ CD45(–)/glycophorin-A-bone marrow cells. Haematologica 2007;92:1707–8. [36] Gronthos S, Fitter S, Diamond P, Simmons PJ, Itescu S, Zannettino AC. A novel monoclonal antibody (STRO-3) identifies an isoform of tissue nonspecific alkaline phosphatase expressed by multipotent bone marrow stromal stem cells. Stem Cells Dev 2007;16:953–63. [37] Kaiser S, Hackanson B, Follo M, Mehlhorn A, Geiger K, Ihorst G, et al. BM cells giving rise to MSC in culture have a heterogeneous CD34 and CD45 phenotype. Cytotherapy 2007;9:439–50. [38] da Silva ML, Chagastelles PC, Nardi NB. Mesenchymal stem cells reside in virtually all post-natal organs and tissues. J Cell Sci 2006;119:2204–13. [39] Schieker M, Pautke C, Haasters F, Schieker J, Docheva D, ­Bocker W, et al. Human mesenchymal stem cells at the single-cell level: simultaneous seven-colour immunofluorescence. J Anat 2007; 210:592–9. [40] Mageed AS, Pietryga DW, DeHeer DH, West RA. Isolation of large numbers of mesenchymal stem cells from the washings of bone marrow collection bags: characterization of fresh mesenchymal stem cells. Transplantation 2007;83:1019–26. [41] Ringe J, Strassburg S, Neumann K, Endres M, Notter M, ­Burmester GR, et al. Towards in situ tissue repair: human mesenchymal stem cells express chemokine receptors CXCR1, CXCR2 and CCR2, and migrate upon stimulation with CXCL8 but not CCL2. J Cell ­Biochem 2007;101:135–46. [42] Martins AA, Paiva A, Morgado JM, Gomes A, Pais ML. Quantification and immunophenotypic characterization of bone marrow and umbilical cord blood mesenchymal stem cells by multicolor flow cytometry. Transplant Proc 2009;41:943–6. [43] Qian H, Le BK, Sigvardsson M. Primary mesenchymal stem and progenitor cells from bone marrow lack expression of CD44. J Biol Chem 2012;287:25795–807.

141

[44] Tondreau T, Lagneaux L, Dejeneffe M, Delforge A, Massy M, Mortier C, et al. Isolation of BM mesenchymal stem cells by ­plastic adhesion or negative selection: phenotype, proliferation kinetics and differentiation potential. Cytotherapy 2004;6:372–9. [45] Peiffer I, Eid P, Barbet R, Li ML, Oostendorp RA, Haydont V, et al. A sub-population of high proliferative potential-quiescent human mesenchymal stem cells is under the reversible control of interferon alpha/beta. Leukemia 2007;21:714–24. [46] Jones EA, English A, Kinsey SE, Straszynski L, Emery P, ­Ponchel F, et al. Optimization of a flow cytometry-based protocol for ­detection and phenotypic characterization of multipotent mesenchymal stromal cells from human bone marrow. Cytometry B Clin Cytom 2006;70:391–9. [47] Baksh D, Zandstra PW, Davies JE. A non-contact suspension ­culture approach to the culture of osteogenic cells derived from a CD49elow subpopulation of human bone marrow-derived cells. Biotechnol Bioeng 2007;98:1195–208. [48] Letchford J, Cardwell AM, Stewart K, Coogans KK, Cox JP, Lee M, et al. Isolation of C15: a novel antibody generated by phage display against mesenchymal stem cell-enriched fractions of adult human marrow. J Immunol Methods 2006;308:124–37. [49] Kastrinaki MC, Andreakou I, Charbord P, Papadaki HA. Isolation of human bone marrow mesenchymal stem cells using different membrane markers: comparison of colony/cloning efficiency, ­differentiation potential, and molecular profile. Tissue Eng Part C Methods 2008;14:333–9. [50] Deschaseaux F, Gindraux F, Saadi R, Obert L, Chalmers D, Herve P. Direct selection of human bone marrow mesenchymal stem cells using an anti-CD49a antibody reveals their CD45med, low phenotype. Br J Haematol 2003;122:506–17. [51] Quirici N, Soligo D, Bossolasco P, Servida F, Lumini C, Deliliers GL. Isolation of bone marrow mesenchymal stem cells by anti-nerve growth factor receptor antibodies. Exp Hematol 2002;30:783–91. [52] Stewart K, Monk P, Walsh S, Jefferiss CM, Letchford J, Beresford JN. STRO-1, HOP-26 (CD63), CD49a and SB-10 (CD166) as markers of primitive human marrow stromal cells and their more ­differentiated progeny: a comparative investigation in vitro. Cell ­Tissue Res 2003;313:281–90. [53] Rider DA, Nalathamby T, Nurcombe V, Cool SM. Selection ­using the alpha-1 integrin (CD49a) enhances the multipotentiality of the mesenchymal stem cell population from heterogeneous bone marrow stromal cells. J Mol Histol 2007;38:449–58. [54] Gindraux F, Selmani Z, Obert L, Davani S, Tiberghien P, Herve P, et al. Human and rodent bone marrow mesenchymal stem cells that express primitive stem cell markers can be directly enriched by using the CD49a molecule. Cell Tissue Res 2007;327:471–83. [55] Delorme B, Ringe J, Gallay N, Le VY, Kerboeuf D, Jorgensen C, et al. Specific plasma membrane protein phenotype of cultureamplified and native human bone marrow mesenchymal stem cells. Blood 2008;111:2631–5. [56] Buhring HJ, Treml S, Cerabona F, de Zwart P, Kanz L, Sobiesiak M. Phenotypic characterization of distinct human bone marrowderived MSC subsets. Ann N Y Acad Sci 2009;1176:124–34. [57] Sobiesiak M, Sivasubramaniyan K, Hermann C, Tan C, Orgel M, Treml S, et al. The mesenchymal stem cell antigen MSCA-1 is identical to tissue non-specific alkaline phosphatase. Stem Cells Dev 2010;19:669–77. [58] Battula VL, Treml S, Bareiss PM, Gieseke F, Roelofs H, de Zwart P, et al. Isolation of functionally distinct mesenchymal stem cell subsets using antibodies against CD56, CD271, and mesenchymal stem cell antigen-1. Haematologica 2009;94:173–84. [59] Lange C, Cakiroglu F, Spiess AN, Cappallo-Obermann H, Dierlamm J, Zander AR. Accelerated and safe expansion of human mesenchymal stromal cells in animal serum-free medium for transplantation and regenerative medicine. J Cell Physiol 2007; 213:18–26.

II.  CELLULAR, MOLECULAR, AND DEVELOPMENTAL BIOLOGY OF BONE

142

7.  THE SKELETAL STEM CELL

[60] Barry F, Boynton R, Murphy M, Haynesworth S, Zaia J. The SH-3 and SH-4 antibodies recognize distinct epitopes on CD73 from human mesenchymal stem cells. Biochem Biophys Res Commun 2001;289:519–24. [61] Liu PG, Zhou DB, Shen T. Identification of human bone marrow mesenchymal stem cells: preparation and utilization of two monoclonal antibodies against SH2, SH3 [in Chinese]. Zhongguo Shi Yan Xue Ye Xue Za Zhi 2005;13:656–9. [62] Odabas S, Sayar F, Guven G, Yanikkaya-Demirel G, Piskin E. Separation of mesenchymal stem cells with magnetic nanosorbents carrying CD105 and CD73 antibodies in flow-through and batch systems. J Chromatogr B Analyt Technol Biomed Life Sci 2008;861:74–80. [63] Ghilzon R, McCulloch CA, Zohar R. Stromal mesenchymal progenitor cells. Leuk Lymphoma 1999;32:211–21. [64] Campioni D, Lanza F, Moretti S, Ferrari L, Cuneo A. Loss of Thy1 (CD90) antigen expression on mesenchymal stromal cells from hematologic malignancies is induced by in vitro angiogenic stimuli and is associated with peculiar functional and phenotypic characteristics. Cytotherapy 2008;10:69–82. [65] Barry FP, Boynton RE, Haynesworth S, Murphy JM, Zaia J. The monoclonal antibody SH-2, raised against human mesenchymal stem cells, recognizes an epitope on endoglin (CD105). Biochem Biophys Res Commun 1999;265:134–9. [66] Tuli R, Tuli S, Nandi S, Wang ML, Alexander PG, Haleem-Smith H, et al. Characterization of multipotential mesenchymal progenitor cells derived from human trabecular bone. Stem Cells 2003;21:681–93. [67] Tondreau T, Lagneaux L, Dejeneffe M, Massy M, Mortier C, ­Delforge A, et al. Bone marrow-derived mesenchymal stem cells already express specific neural proteins before any differentiation. Differentiation 2004;72:319–26. [68] Boiret N, Rapatel C, Veyrat-Masson R, Guillouard L, Guerin JJ, Pigeon P, et al. Characterization of nonexpanded mesenchymal progenitor cells from normal adult human bone marrow. Exp ­Hematol 2005;33:219–25. [69] Stute N, Holtz K, Bubenheim M, Lange C, Blake F, Zander AR. Autologous serum for isolation and expansion of human mesenchymal stem cells for clinical use. Exp Hematol 2004;32:1212–25. [70] Muller I, Kordowich S, Holzwarth C, Spano C, Isensee G, Staiber A, et al. Animal serum-free culture conditions for isolation and expansion of multipotent mesenchymal stromal cells from human BM. Cytotherapy 2006;8:437–44. [71] Jarocha D, Lukasiewicz E, Majka M. Adventage of mesenchymal stem cells (MSC) expansion directly from purified bone marrow CD105+ and CD271+ cells. Folia Histochem Cytobiol 2008;46:307–14. [72] Aslan H, Zilberman Y, Kandel L, Liebergall M, Oskouian RJ, Gazit D, et al. Osteogenic differentiation of noncultured immunoisolated bone marrow-derived CD105+ cells. Stem Cells 2006;24:1728–37. [73] Haynesworth SE, Baber MA, Caplan AI. Cell surface antigens on human marrow-derived mesenchymal cells are detected by monoclonal antibodies. Bone 1992;13:69–80. [74] Shi S, Gronthos S. Perivascular niche of postnatal mesenchymal stem cells in human bone marrow and dental pulp. J Bone Miner Res 2003;18:696–704. [75] Gronthos S, Zannettino AC, Hay SJ, Shi S, Graves SE, Kortesidis A, et al. Molecular and cellular characterisation of highly purified stromal stem cells derived from human bone marrow. J Cell Sci 2003;116:1827–35. [76] Gronthos S, Zannettino AC. A method to isolate and purify human bone marrow stromal stem cells. Methods Mol Biol 2008;449:45–57. [77] Giesert C, Marxer A, Sutherland DR, Schuh AC, Kanz L, Buhring HJ. Antibody W7C5 defines a CD109 epitope expressed on CD34+ and CD34– hematopoietic and mesenchymal stem cell subsets. Ann N Y Acad Sci 2003;996:227–30.

[78] Sorrentino A, Ferracin M, Castelli G, Biffoni M, Tomaselli G, Baiocchi M, et al. Isolation and characterization of CD146+ multipotent mesenchymal stromal cells. Exp Hematol 2008;36:1035–46. [79] Arai F, Ohneda O, Miyamoto T, Zhang XQ, Suda T. Mesenchymal stem cells in perichondrium express activated leukocyte cell adhesion molecule and participate in bone marrow formation. J Exp Med 2002;195:1549–63. [80] Quirici N, Scavullo C, de Girolamo L, Lopa S, Arrigoni E, ­Deliliers GL, et al. Anti-L-NGFR and -CD34 monoclonal antibodies identify multipotent mesenchymal stem cells in human adipose tissue. Stem Cells Dev 2010;19:915–25. [81] Poloni A, Maurizi G, Rosini V, Mondini E, Mancini S, Discepoli G, et al. Selection of CD271(+) cells and human AB serum allows a large expansion of mesenchymal stromal cells from human bone marrow. Cytotherapy 2009;11:153–62. [82] Horn P, Bork S, Diehlmann A, Walenda T, Eckstein V, Ho AD, et al. Isolation of human mesenchymal stromal cells is more efficient by red blood cell lysis. Cytotherapy 2008;10:676–85. [83] Bieback K, Hecker A, Kocaomer A, Lannert H, Schallmoser K, Strunk D, et al. Human alternatives to fetal bovine serum for the expansion of mesenchymal stromal cells from bone marrow. Stem Cells 2009;27:2331–41. [84] Hegner B, Weber M, Dragun D, Schulze-Lohoff E. Differential regulation of smooth muscle markers in human bone marrowderived mesenchymal stem cells. J Hypertens 2005;23:1191–202. [85] Gentry T, Foster S, Winstead L, Deibert E, Fiordalisi M, Balber A. Simultaneous isolation of human BM hematopoietic, e­ ndothelial and mesenchymal progenitor cells by flow sorting based on ­aldehyde dehydrogenase activity: implications for cell therapy. Cytotherapy 2007;9:259–74. [86] Buhring HJ, Kuci S, Conze T, Rathke G, Bartolovic K, Grunebach F, et al. CDCP1 identifies a broad spectrum of normal and malignant stem/progenitor cell subsets of hematopoietic and nonhematopoietic origin. Stem Cells 2004;22:334–43. [87] Martinez C, Hofmann TJ, Marino R, Dominici M, Horwitz EM. Human bone marrow mesenchymal stromal cells express the neural ganglioside GD2: a novel surface marker for the identification of MSCs. Blood 2007;109:4245–8. [88] Gronthos S, McCarty R, Mrozik K, Fitter S, Paton S, Menicanin D, et al. Heat shock protein-90 beta is expressed at the surface of multipotential mesenchymal precursor cells: generation of a novel monoclonal antibody, STRO-4, with specificity for mesenchymal precursor cells from human and ovine tissues. Stem Cells Dev 2009;18:1253–62. [89] Aleksandrova MA, Sukhikh GT, Chailakhyan RK, Podgornyi OV, Marei MV, Poltavtseva RA, et al. Comparative analysis of differentiation and behavior of human neural and mesenchymal stem cells in vitro and in vivo. Bull Exp Biol Med 2006;141:152–60. [90] Gang EJ, Bosnakovski D, Figueiredo CA, Visser JW, Perlingeiro RC. SSEA-4 identifies mesenchymal stem cells from bone marrow. Blood 2007;109:1743–51. [91] Kim YH, Yoon DS, Kim HO, Lee JW. Characterization of different subpopulations from bone marrow-derived mesenchymal stromal cells by alkaline phosphatase expression. Stem Cells Dev 2012;21:2958–68. [92] Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini F, Krause D, et al. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 2006;8:315–7. [93] Wang L, Liu Y, Kalajzic Z, Jiang X, Rowe DW. Heterogeneity of engrafted bone-lining cells after systemic and local transplantation. Blood 2005;106:3650–7. [94] Morikawa S, Mabuchi Y, Kubota Y, Nagai Y, Niibe K, Hiratsu E, et al. Prospective identification, isolation, and systemic transplantation of multipotent mesenchymal stem cells in murine bone marrow. J Exp Med 2009;206:2483–96.

II.  CELLULAR, MOLECULAR, AND DEVELOPMENTAL BIOLOGY OF BONE

Summary

[95] Mendez-Ferrer S, Michurina TV, Ferraro F, Mazloom AR, Macarthur BD, Lira SA, et al. Mesenchymal and haematopoietic stem cells form a unique bone marrow niche. Nature 2010;466:829–34. [96] Park D, Spencer JA, Koh BI, Kobayashi T, Fujisaki J, Clemens TL, et al. Endogenous bone marrow MSCs are dynamic, fate-restricted participants in bone maintenance and regeneration. Cell Stem Cell 2012;10:259–72. [97] Ferraro F, Lymperi S, Mendez-Ferrer S, Saez B, Spencer JA, Yeap BY, et al. Diabetes impairs hematopoietic stem cell mobilization by altering niche function. Sci Transl Med 2011;3;104ra101. [98] He Q, Wan C, Li G. Concise review: multipotent mesenchymal stromal cells in blood. Stem Cells 2007;25:69–77. [99] Kuznetsov SA, Mankani MH, Gronthos S, Satomura K, Bianco P, Robey PG. Circulating skeletal stem cells. J Cell Biol 2001;153:1133–40. [100] Piersma AH, Ploemacher RE, Brockbank KG, Nikkels PG, ­Ottenheim CP. Migration of fibroblastoid stromal cells in murine blood. Cell Tissue Kinet 1985;18:589–95. [101] Metcalf D. Formation in agar of fibroblast-like colonies by cells from the mouse pleural cavity and other sources. J Cell Physiol 1972;80:409–19. [102] Kassis I, Zangi L, Rivkin R, Levdansky L, Samuel S, Marx G, et al. Isolation of mesenchymal stem cells from G-CSF-mobilized human peripheral blood using fibrin microbeads. Bone Marrow Transplant 2006;37:967–76. [103] Wang Y, Johnsen HE, Mortensen S, Bindslev L, Ripa RS, ­Haack-Sorensen M, et al. Changes in circulating mesenchymal stem cells, stem cell homing factor, and vascular growth factors in patients with acute ST elevation myocardial infarction treated with primary percutaneous coronary intervention. Heart 2006;92:768–74. [104] Moosmann S, Hutter J, Moser C, Krombach F, Huss R. ­Milieu-adopted in vitro and in vivo differentiation of mesenchymal tissues derived from different adult human CD34-negative progenitor cell clones. Cells Tissues Organs 2005;179:91–101. [105] Cao C, Dong Y, Dong Y. Study on culture and in vitro osteogenesis of blood-derived human mesenchymal stem cells [in Chinese]. Zhongguo Xiu Fu Chong Jian Wai Ke Za Zhi 2005;19:642–7. [106] Conrad C, Gottgens B, Kinston S, Ellwart J, Huss R. GATA transcription in a small rhodamine 123(low)CD34(+) subpopulation of a peripheral blood-derived CD34(–)CD105(+) mesenchymal cell line. Exp Hematol 2002;30:887–95. [107] Fernandez M, Simon V, Herrera G, Cao C, Del FH, Minguell JJ. Detection of stromal cells in peripheral blood progenitor cell collections from breast cancer patients. Bone Marrow Transplant 1997;20:265–71. [108] Erices A, Conget P, Minguell JJ. Mesenchymal progenitor cells in human umbilical cord blood. Br J Haematol 2000;109:235–42. [109] Goodwin HS, Bicknese AR, Chien SN, Bogucki BD, Quinn CO, Wall DA. Multilineage differentiation activity by cells isolated from umbilical cord blood: expression of bone, fat, and neural markers. Biol Blood Marrow Transplant 2001;7:581–8. [110] Ye ZQ, Burkholder JK, Qiu P, Schultz JC, Shahidi NT, Yang NS. Establishment of an adherent cell feeder layer from human umbilical cord blood for support of long-term hematopoietic progenitor cell growth. Proc Natl Acad Sci U. S. A. 1994;91:12140–4. [111] Hutson EL, Boyer S, Genever PG. Rapid isolation, expansion, and differentiation of osteoprogenitors from full-term umbilical cord blood. Tissue Eng 2005;11:1407–20. [112] Lee MW, Choi J, Yang MS, Moon YJ, Park JS, Kim HC, et al. ­Mesenchymal stem cells from cryopreserved human umbilical cord blood. Biochem Biophys Res Commun 2004;320:273–8. [113] Prindull G, Ben-Ishay Z, Ebell W, Bergholz M, Dirk T, Prindull B. CFU-F circulating in cord blood. Blut 1987;54:351–9.

143

[114] Eghbali-Fatourechi GZ, Lamsam J, Fraser D, Nagel D, Riggs BL, Khosla S. Circulating osteoblast-lineage cells in humans. N Engl J Med 2005;352:1959–66. [115] Otsuru S, Tamai K, Yamazaki T, Yoshikawa H, Kaneda Y. ­Circulating bone marrow-derived osteoblast progenitor cells are recruited to the bone-forming site by the CXCR4/stromal cellderived factor-1 pathway. Stem Cells 2008;26:223–34. [116] Wang CH, Cherng WJ, Yang NI, Kuo LT, Hsu CM, Yeh HI, et al. Late-outgrowth endothelial cells attenuate intimal hyperplasia contributed by mesenchymal stem cells after vascular injury. ­Arterioscler Thromb Vasc Biol 2008;28:54–60. [117] Tang Y, Wu X, Lei W, Pang L, Wan C, Shi Z, et al. TGF-beta1-­ induced migration of bone mesenchymal stem cells couples bone resorption with formation. Nat Med 2009;15:757–65. [118] Owen M, Macpherson S. Cell population kinetics of an osteogenic tissue II. J.Cell Biol 1963;19:33–44. [119] McCulloch CA, Heersche JN. Lifetime of the osteoblast in mouse periodontium. Anat Rec 1988;222:128–35. [120] Jilka RL, Weinstein RS, Parfitt AM, Manolagas SC. Quantifying osteoblast and osteocyte apoptosis: challenges and rewards. J Bone Miner Res 2007;22:1492–501. [121] Parfitt AM, Han ZH, Palnitkar S, Rao DS, Shih MS, Nelson D. ­Effects of ethnicity and age or menopause on osteoblast function, bone mineralization, and osteoid accumulation in iliac bone. J Bone Miner Res 1997;12:1864–73. [122] Lo CC, Fleming HE, Wu JW, Zhao CX, Miake-Lye S, Fujisaki J, et al. Live-animal tracking of individual haematopoietic stem/ progenitor cells in their niche. Nature 2009;457:92–6. [123] Sipkins DA, Wei X, Wu JW, Runnels JM, Cote D, Means TK, et al. In vivo imaging of specialized bone marrow endothelial microdomains for tumour engraftment. Nature 2005;435:969–73. [124] Dominici M, Rasini V, Bussolari R, Chen X, Hofmann TJ, Spano C, et al. Restoration and reversible expansion of the osteoblastic hematopoietic stem cell niche after marrow radioablation. Blood 2009;114:2333–43. [125] Maes C, Kobayashi T, Selig MK, Torrekens S, Roth SI, Mackem S, et al. Osteoblast precursors, but not mature osteoblasts, move into developing and fractured bones along with invading blood vessels. Dev Cell 2010;19:329–44. [126] Maes C, Kobayashi T, Kronenberg HM. A novel transgenic mouse model to study the osteoblast lineage in vivo. Ann N Y Acad Sci 2007;1116:149–64. [127] Malizos KN, Papatheodorou LK. The healing potential of the periosteum molecular aspects. Injury 2005;36(Suppl. 3):S13–9. [128] Grcevic D, Pejda S, Matthews BG, Repic D, Wang L, Li H, et al. In vivo fate mapping identifies mesenchymal progenitor cells. Stem Cells 2012;30:187–96. [129] Horwitz EM, Prockop DJ, Fitzpatrick LA, Koo WW, Gordon PL, Neel M, et al. Transplantability and therapeutic effects of bone marrow-derived mesenchymal cells in children with osteogenesis imperfecta. Nat Med 1999;5:309–13. [130] Ocarino NM, Boeloni JN, Jorgetti V, Gomes DA, Goes AM, ­Serakides R. Intra-bone marrow injection of mesenchymal stem cells improves the femur bone mass of osteoporotic female rats. Connect Tissue Res 2010;51:426–33. [131] Wang Z, Goh J, Das DS, Ge Z, Ouyang H, Chong JS, et al. Efficacy of bone marrow-derived stem cells in strengthening osteoporotic bone in a rabbit model. Tissue Eng 2006;12:1753–61. [132] Anjos-Afonso F, Siapati EK, Bonnet D. In vivo contribution of ­murine mesenchymal stem cells into multiple cell-types under minimal damage conditions. J Cell Sci 2004;117:5655–64. [133] Rombouts WJ, Ploemacher RE. Primary murine MSC show ­highly efficient homing to the bone marrow but lose homing ability following culture. Leukemia 2003;17:160–70. [134] Wu Y, Zhao RC. The role of chemokines in mesenchymal stem cell homing to myocardium. Stem Cell Rev 2012;8:243–50.

II.  CELLULAR, MOLECULAR, AND DEVELOPMENTAL BIOLOGY OF BONE

144

7.  THE SKELETAL STEM CELL

[135] Abbott JD, Huang Y, Liu D, Hickey R, Krause DS, Giordano FJ. Stromal cell-derived factor-1alpha plays a critical role in stem cell recruitment to the heart after myocardial infarction but is not sufficient to induce homing in the absence of injury. Circulation 2004;110:3300–5. [136] Kumar S, Ponnazhagan S. Bone homing of mesenchymal stem cells by ectopic alpha 4 integrin expression. FASEB J 2007;21:3917–27. [137] Guan M, Yao W, Liu R, Lam KS, Nolta J, Jia J, et al. Directing mesenchymal stem cells to bone to augment bone formation and increase bone mass. Nat Med 2012;18:456–62. [138] Sackstein R, Merzaban JS, Cain DW, Dagia NM, Spencer JA, Lin CP, et al. Ex vivo glycan engineering of CD44 programs human multipotent mesenchymal stromal cell trafficking to bone. Nat Med 2008;14:181–7. [139] Veronesi F, Torricelli P, Borsari V, Tschon M, Rimondini L, Fini M. Mesenchymal stem cells in the aging and osteoporotic population. Crit Rev Eukaryot Gene Expr 2011;21:363–77. [140] Meunier P, Aaron J, Edouard C, Vignon G. Osteoporosis and the replacement of cell populations of the marrow by adipose tissue. A quantitative study of 84 iliac bone biopsies. Clin Orthop Relat Res 1971;80:147–54. [141] Manolagas SC. Birth and death of bone cells: basic regulatory mechanisms and implications for the pathogenesis and treatment of osteoporosis. Endocr Rev 2000;21:115–37. [142] Shen J, Tsai YT, Dimarco NM, Long MA, Sun X, Tang L. ­Transplantation of mesenchymal stem cells from young donors delays aging in mice. Sci Rep 2011;1:67. [143] Horwitz EM, Prockop DJ, Gordon PL, Koo WW, Fitzpatrick LA, Neel MD, et al. Clinical responses to bone marrow transplantation in children with severe osteogenesis imperfecta. Blood 2001;97:1227–31. [144] Horwitz EM, Gordon PL, Koo WK, Marx JC, Neel MD, McNall RY, et al. Isolated allogeneic bone marrow-derived mesenchymal cells engraft and stimulate growth in children with osteogenesis imperfecta: implications for cell therapy of bone. Proc Natl Acad Sci U. S. A. 2002;99:8932–7. [145] Chamberlain JR, Deyle DR, Schwarze U, Wang P, Hirata RK, Li Y, et al. Gene targeting of mutant COL1A2 alleles in mesenchymal stem cells from individuals with osteogenesis imperfecta. Mol Ther 2008;16:187–93. [146] Chamberlain JR, Schwarze U, Wang PR, Hirata RK, Hankenson KD, Pace JM, et al. Gene targeting in stem cells from individuals with osteogenesis imperfecta. Science 2004;303:1198–201. [147] Deyle DR, Khan IF, Ren G, Wang PR, Kho J, Schwarze U, et al. Normal collagen and bone production by gene-targeted human osteogenesis imperfecta iPSCs. Mol Ther 2012;20:204–13. [148] Gong JK. Endosteal marrow: a rich source of hematopoietic stem cells. Science 1978;199:1443–5. [149] Lord BI, Testa NG, Hendry JH. The relative spatial distributions of CFUs and CFUc in the normal mouse femur. Blood 1975;46:65–72. [150] Kohler A, Schmithorst V, Filippi MD, Ryan MA, Daria D, ­Gunzer M, et al. Altered cellular dynamics and endosteal location of aged early hematopoietic progenitor cells ­revealed by time-lapse intravital imaging in long bones. Blood 2009;114:290–8. [151] Nilsson SK, Johnston HM, Coverdale JA. Spatial localization of transplanted hemopoietic stem cells: inferences for the localization of stem cell niches. Blood 2001;97:2293–9. [152] Xie Y, Yin T, Wiegraebe W, He XC, Miller D, Stark D, et al. ­Detection of functional haematopoietic stem cell niche using real-time imaging. Nature 2009;457:97–101. [153] Haylock DN, Williams B, Johnston HM, Liu MC, Rutherford KE, Whitty GA, et al. Hemopoietic stem cells with higher hemopoietic potential reside at the bone marrow endosteum. Stem Cells 2007;25:1062–9.

[154] Taichman RS, Emerson SG. Human osteoblasts support hematopoiesis through the production of granulocyte colony-stimulating factor. J Exp Med 1994;179:1677–82. [155] Taichman RS, Reilly MJ, Emerson SG. Human osteoblasts support human hematopoietic progenitor cells in vitro bone marrow cultures. Blood 1996;87:518–24. [156] Calvi LM, Adams GB, Weibrecht KW, Weber JM, Olson DP, Knight MC, et al. Osteoblastic cells regulate the haematopoietic stem cell niche. Nature 2003;425:841–6. [157] Zhang J, Niu C, Ye L, Huang H, He X, Tong WG, et al. ­Identification of the haematopoietic stem cell niche and control of the niche size. Nature 2003;425:836–41. [158] 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;103:3258–64. [159] Arai F, Hirao A, Ohmura M, Sato H, Matsuoka S, Takubo K, et al. Tie2/angiopoietin-1 signaling regulates hematopoietic stem cell quiescence in the bone marrow niche. Cell 2004;118:149–61. [160] Stier S, Cheng T, Dombkowski D, Carlesso N, Scadden DT. Notch1 activation increases hematopoietic stem cell self-­renewal in vivo and favors lymphoid over myeloid lineage outcome. Blood 2002;99:2369–78. [161] Varnum-Finney B, Brashem-Stein C, Bernstein ID. Combined ­effects of Notch signaling and cytokines induce a multiple log ­increase in precursors with lymphoid and myeloid reconstituting ability. Blood 2003;101:1784–9. [162] Karanu FN, Murdoch B, Gallacher L, Wu DM, Koremoto M, Sakano S, et al. The notch ligand jagged-1 represents a novel growth factor of human hematopoietic stem cells. J Exp Med 2000;192:1365–72. [163] Bigas A, Espinosa L. Hematopoietic stem cells: to be or Notch to be. Blood 2012;119:3226–35. [164] 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;4:170–9. [165] 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;1:204–17. [166] Zanjani ED, Flake AW, Almeida-Porada G, Tran N, Papayannopoulou T. Homing of human cells in the fetal sheep model: modulation by antibodies activating or inhibiting very late activation antigen-4-dependent function. Blood 1999;94:2515–22. [167] Vermeulen M, Le Pesteur F, Gagnerault MC, Mary JY, Sainteny F, Lepault F. Role of adhesion molecules in the homing and mobilization of murine hematopoietic stem and progenitor cells. Blood 1998;92:894–900. [168] Peled A, Kollet O, Ponomaryov T, Petit I, Franitza S, Grabovsky V, et al. The chemokine SDF-1 activates the integrins LFA-1, VLA-4, and VLA-5 on immature human CD34(+) cells: role in transendothelial/stromal migration and engraftment of NOD/ SCID mice. Blood 2000;95:3289–96. [169] Papayannopoulou T, Priestley GV, Nakamoto B, Zafiropoulos V, Scott LM. Molecular pathways in bone marrow homing: dominant role of alpha(4)beta(1) over beta(2)-integrins and selectins. Blood 2001;98:2403–11. [170] Levesque JP, Leavesley DI, Niutta S, Vadas M, Simmons PJ. ­Cytokines increase human hemopoietic cell adhesiveness by ­activation of very late antigen (VLA)-4 and VLA-5 integrins. J Exp Med 1995;181:1805–15. [171] Levesque JP, Takamatsu Y, Nilsson SK, Haylock DN, Simmons PJ. Vascular cell adhesion molecule-1 (CD106) is cleaved by neutrophil proteases in the bone marrow following hematopoietic progenitor cell mobilization by granulocyte colony-stimulating factor. Blood 2001;98:1289–97. [172] van der Loo JC, Xiao X, McMillin D, Hashino K, Kato I, Williams DA. VLA-5 is expressed by mouse and human long-term repopulating hematopoietic cells and mediates adhesion to extracellular matrix protein fibronectin. J Clin Invest 1998;102:1051–61.

II.  CELLULAR, MOLECULAR, AND DEVELOPMENTAL BIOLOGY OF BONE

Summary

[173] Katayama Y, Hidalgo A, Peired A, Frenette PS. Integrin ­alpha4beta7 and its counterreceptor MAdCAM-1 contribute to hematopoietic progenitor recruitment into bone marrow following transplantation. Blood 2004;104:2020–6. [174] Qian H, Georges-Labouesse E, Nystrom A, Domogatskaya A, Tryggvason K, Jacobsen SE, et al. Distinct roles of integrins ­alpha6 and alpha4 in homing of fetal liver hematopoietic stem and ­progenitor cells. Blood 2007;110:2399–407. [175] Qian H, Tryggvason K, Jacobsen SE, Ekblom M. Contribution of alpha6 integrins to hematopoietic stem and progenitor cell ­homing to bone marrow and collaboration with alpha4 integrins. Blood 2006;107:3503–10. [176] Schmits R, Filmus J, Gerwin N, Senaldi G, Kiefer F, Kundig T, et al. CD44 regulates hematopoietic progenitor distribution, granuloma formation, and tumorigenicity. Blood 1997;90:2217–33. [177] Sackstein R. The bone marrow is akin to skin: HCELL and the biology of hematopoietic stem cell homing. J Invest Dermatol 2004;122:1061–9. [178] Katayama Y, Hidalgo A, Furie BC, Vestweber D, Furie B, Frenette PS, et al. PSGL-1 participates in E-selectin-mediated progenitor homing to bone marrow: evidence for cooperation between E-selectin ligands and alpha4 integrin. Blood 2003;102:2060–7. [179] Frenette PS, Subbarao S, Mazo IB, von Andrian UH, Wagner DD. Endothelial selectins and vascular cell adhesion molecule-1 promote hematopoietic progenitor homing to bone marrow. Proc Natl Acad Sci U. S. A. 1998;95:14423–8. [180] Forde S, Tye BJ, Newey SE, Roubelakis M, Smythe J, ­McGuckin CP, et al. Endolyn (CD164) modulates the CXCL12-­ mediated migration of umbilical cord blood CD133+ cells. Blood 2007;109:1825–33. [181] Adams GB, Chabner KT, Alley IR, Olson DP, Szczepiorkowski ZM, Poznansky MC, et al. Stem cell engraftment at the endosteal niche is specified by the calcium-sensing receptor. Nature 2006;439:599–603. [182] Ponomaryov T, Peled A, Petit I, Taichman RS, Habler L, ­Sandbank J, et al. Induction of the chemokine stromal-derived factor-1 ­ ­ following DNA damage improves human stem cell function. J Clin Invest 2000;106:1331–9. [183] Grassinger J, Haylock DN, Storan MJ, Haines GO, Williams B, Whitty GA, et al. Thrombin-cleaved osteopontin regulates hemopoietic stem and progenitor cell functions through interactions with alpha9beta1 and alpha4beta1 integrins. Blood 2009;114:49–59. [184] Nilsson SK, Johnston HM, Whitty GA, Williams B, Webb RJ, ­Denhardt DT, et al. Osteopontin, a key component of the hematopoietic stem cell niche and regulator of primitive hematopoietic progenitor cells. Blood 2005;106:1232–9. [185] Stier S, Ko Y, Forkert R, Lutz C, Neuhaus T, Grunewald E, et al. Osteopontin is a hematopoietic stem cell niche component that negatively regulates stem cell pool size. J Exp Med 2005;201:1781–91. [186] Mazzon C, Anselmo A, Cibella J, Soldani C, Destro A, Kim N, et al. The critical role of agrin in the hematopoietic stem cell niche. Blood 2011;118:2733–42. [187] Ramirez P, Rettig MP, Uy GL, Deych E, Holt MS, Ritchey JK, et al. BIO5192, a small molecule inhibitor of VLA-4, mobilizes hematopoietic stem and progenitor cells. Blood 2009;114:1340–3. [188] Papayannopoulou T, Priestley GV, Nakamoto B. Anti-VLA4/ VCAM-1-induced mobilization requires cooperative signaling through the kit/mkit ligand pathway. Blood 1998;91:2231–9. [189] Kikuta T, Shimazaki C, Ashihara E, Sudo Y, Hirai H, Sumikuma T, et al. Mobilization of hematopoietic primitive and committed progenitor cells into blood in mice by anti-vascular adhesion molecule-1 antibody alone or in combination with granulocyte colony-stimulating factor. Exp Hematol 2000;28:311–7.

145

[190] Craddock CF, Nakamoto B, Andrews RG, Priestley GV, ­Papayannopoulou T. Antibodies to VLA4 integrin mobilize long-term repopulating cells and augment cytokine-induced mobilization in primates and mice. Blood 1997;90:4779–88. [191] Broxmeyer HE, Orschell CM, Clapp DW, Hangoc G, Cooper S, Plett PA, et al. Rapid mobilization of murine and human hematopoietic stem and progenitor cells with AMD3100, a CXCR4 ­antagonist. J Exp Med 2005;201:1307–18. [192] Broxmeyer HE, Hangoc G, Cooper S, Campbell T, Ito S, Mantel C, et al. AMD3100 and CD26 modulate mobilization, engraftment, and survival of hematopoietic stem and progenitor cells mediated by the SDF-1/CXCL12-CXCR4 axis. Ann N Y Acad Sci 2007;1106:1–9. [193] Jung Y, Wang J, Havens A, Sun Y, Wang J, Jin T, et al. C ­ ell-to-cell contact is critical for the survival of hematopoietic progenitor cells on osteoblasts. Cytokine 2005;32:155–62. [194] Gillette JM, Larochelle A, Dunbar CE, Lippincott-Schwartz J. Intercellular transfer to signalling endosomes regulates an ex vivo bone marrow niche. Nat Cell Biol 2009;11:303–11. [195] Winkler IG, Sims NA, Pettit AR, Barbier V, Nowlan B, Helwani F, et al. Bone marrow macrophages maintain hematopoietic stem cell (HSC) niches and their depletion mobilizes HSCs. Blood 2010;116:4815–28. [196] Semerad CL, Christopher MJ, Liu F, Short B, Simmons PJ, Winkler I, et al. G-CSF potently inhibits osteoblast activity and CXCL12 mRNA expression in the bone marrow. Blood 2005;106:3020–7. [197] Katayama Y, Battista M, Kao WM, Hidalgo A, Peired AJ, Thomas SA, et al. Signals from the sympathetic nervous system regulate hematopoietic stem cell egress from bone marrow. Cell 2006;124:407–21. [198] Chang MK, Raggatt LJ, Alexander KA, Kuliwaba JS, Fazzalari NL, Schroder K, et al. Osteal tissue macrophages are intercalated throughout human and mouse bone lining tissues and regulate osteoblast function in vitro and in vivo. J Immunol 2008;181:1232–44. [199] Chow A, Lucas D, Hidalgo A, Mendez-Ferrer S, Hashimoto D, Scheiermann C, 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;208:261–71. [200] 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;208:251–60. [201] Perez-Amodio S, Beertsen W, Everts V. (Pre-)osteoclasts induce retraction of osteoblasts before their fusion to osteoclasts. J Bone Miner Res 2004;19:1722–31. [202] Watanabe T, Yatomi Y, Sunaga S, Miki I, Ishii A, Nakao A, et al. Characterization of prostaglandin and thromboxane receptors expressed on a megakaryoblastic leukemia cell line, MEG-01s. Blood 1991;78:2328–36. [203] Takamatsu Y, Simmons PJ, Moore RJ, Morris HA, To LB, Levesque JP, et al. Osteoclast-mediated bone resorption is stimulated during short-term administration of granulocyte colonystimulating factor but is not responsible for hematopoietic progenitor cell ­mobilization. Blood 1998;92:3465–73. [204] Kollet O, Dar A, Shivtiel S, Kalinkovich A, Lapid K, Sztainberg Y, et al. Osteoclasts degrade endosteal components and promote mobilization of hematopoietic progenitor cells. Nat Med 2006;12:657–64. [205] Cho KA, Joo SY, Han HS, Ryu KH, Woo SY. Osteoclast activation by receptor activator of NF-kappaB ligand enhances the mobilization of hematopoietic progenitor cells from the bone marrow in acute injury. Int J Mol Med 2010;26:557–63. [206] Shivtiel S, Kollet O, Lapid K, Schajnovitz A, Goichberg P, ­Kalinkovich A, et al. CD45 regulates retention, motility, and numbers of hematopoietic progenitors, and affects osteoclast remodeling of metaphyseal trabecules. J Exp Med 2008;205:2381–95.

II.  CELLULAR, MOLECULAR, AND DEVELOPMENTAL BIOLOGY OF BONE

146

7.  THE SKELETAL STEM CELL

[207] Christopher MJ, Link DC. Granulocyte colony-stimulating factor induces osteoblast apoptosis and inhibits osteoblast differentiation. J Bone Miner Res 2008;23:1765–74. [208] Hirbe AC, Uluckan O, Morgan EA, Eagleton MC, Prior JL, ­Piwnica-Worms D, et al. Granulocyte colony-stimulating factor enhances bone tumor growth in mice in an osteoclast-dependent manner. Blood 2007;109:3424–31. [209] Miyamoto K, Yoshida S, Kawasumi M, Hashimoto K, ­Kimura T, Sato Y, et al. Osteoclasts are dispensable for hematopoietic stem cell maintenance and mobilization. J Exp Med 2011;208:2175–81. [210] Kacena MA, Shivdasani RA, Wilson K, Xi Y, Troiano N, Nazarian A, et al. Megakaryocyte-osteoblast interaction revealed in mice deficient in transcription factors GATA-1 and NF-E2. J Bone ­Miner Res 2004;19:652–60. [211] Ciovacco WA, Goldberg CG, Taylor AF, Lemieux JM, Horowitz MC, Donahue HJ, et al. The role of gap junctions in megakaryocyte-mediated osteoblast proliferation and differentiation. Bone 2009;44:80–6. [212] Ciovacco WA, Cheng YH, Horowitz MC, Kacena MA. Immature and mature megakaryocytes enhance osteoblast proliferation and inhibit osteoclast formation. J Cell Biochem 2010;109:774–81. [213] Gurevitch O, Khitrin S, Valitov A, Slavin S. Osteoporosis of hematologic etiology. Exp Hematol 2007;35:128–36. [214] Martelli F, Verrucci M, Migliaccio G, Zingariello M, Rana RA, Vannucchi AM, et al. Removal of the spleen in mice alters the cytokine expression profile of the marrow micro-environment and increases bone formation. Ann N Y Acad Sci 2009;1176:77–86. [215] Savani BN, Donohue T, Kozanas E, Shenoy A, Singh AK, Childs RW, et al. Increased risk of bone loss without fracture risk in long-term survivors after allogeneic stem cell transplantation. Biol Blood Marrow Transplant 2007;13:517–20. [216] Yao S, McCarthy PL, Dunford LM, Roy DM, Brown K, Paplham P, et al. High prevalence of early-onset osteopenia/osteoporosis after allogeneic stem cell transplantation and improvement after bisphosphonate therapy. Bone Marrow Transplant 2008;41:393–8. [217] Lee SJ, Seaborn T, Mao FJ, Massey SC, Luu NQ, Schubert MA, et al. Frequency of abnormal findings detected by comprehensive clinical evaluation at 1 year after allogeneic hematopoietic cell transplantation. Biol Blood Marrow Transplant 2009;15:416–20. [218] Petropoulou AD, Porcher R, Herr AL, Devergie A, Brentano TF, Ribaud P, et al. Prospective assessment of bone turnover and clinical bone diseases after allogeneic hematopoietic stem-cell transplantation. Transplantation 2010;89:1354–61. [219] Baker KS, Ness KK, Weisdorf D, Francisco L, Sun CL, Forman S, et al. Late effects in survivors of acute leukemia treated with ­hematopoietic cell transplantation: a report from the Bone Marrow Transplant Survivor Study. Leukemia 2010;24:2039–47. [220] Hautmann AH, Elad S, Lawitschka A, Greinix H, Bertz H, Halter J, et al. Metabolic bone diseases in patients after allogeneic ­hematopoietic stem cell transplantation: report from the Consensus Conference on Clinical Practice in chronic graft-versus-host disease. Transpl Int 2011;24:867–79. [221] McClune B, Majhail NS, Flowers ME. Bone loss and avascular necrosis of bone after hematopoietic cell transplantation. Semin Hematol 2012;49:59–65. [222] Shanis D, Merideth M, Pulanic TK, Savani BN, Battiwalla M, Stratton P, et al. Female long-term survivors after allogeneic ­hematopoietic stem cell transplantation: evaluation and management. Semin Hematol 2012;49:83–93. [223] Cheng YH, Chitteti BR, Streicher DA, Morgan JA, RodriguezRodriguez S, Carlesso N, et al. Impact of maturational status on the ability of osteoblasts to enhance the hematopoietic function of stem and progenitor cells. J Bone Miner Res 2011;26:1111–21.

[224] Chitteti BR, Cheng YH, Streicher DA, Rodriguez-Rodriguez S, Carlesso N, Srour EF, et al. Osteoblast lineage cells expressing high levels of Runx2 enhance hematopoietic progenitor cell proliferation and function. J Cell Biochem 2010;111:284–94. [225] Omatsu Y, Sugiyama T, Kohara H, Kondoh G, Fujii N, Kohno K, et al. The essential functions of adipo-osteogenic progenitors as the hematopoietic stem and progenitor cell niche. Immunity 2010;33:387–99. [226] Corral DA, Amling M, Priemel M, Loyer E, Fuchs S, Ducy P, et al. Dissociation between bone resorption and bone formation in osteopenic transgenic mice. Proc Natl Acad Sci U. S. A. 1998;95:13835–40. [227] Raaijmakers MH, Mukherjee S, Guo S, Zhang S, Kobayashi T, Schoonmaker JA, et al. Bone progenitor dysfunction induces myelodysplasia and secondary leukaemia. Nature 2010;464: 852–7. [228] 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;25:977–88. [229] Kiel MJ, Yilmaz OH, Iwashita T, Yilmaz OH, Terhorst C, Morrison SJ, et al. SLAM family receptors distinguish hematopoietic stem and progenitor cells and reveal endothelial niches for stem cells. Cell 2005;121:1109–21. [230] Yao L, Yokota T, Xia L, Kincade PW, McEver RP. Bone marrow dysfunction in mice lacking the cytokine receptor gp130 in endothelial cells. Blood 2005;106:4093–101. [231] Ohneda O, Fennie C, Zheng Z, Donahue C, La H, Villacorta R, et al. Hematopoietic stem cell maintenance and differentiation are supported by embryonic aorta-gonad-mesonephros regionderived endothelium. Blood 1998;92:908–19. [232] Li W, Johnson SA, Shelley WC, Yoder MC. Hematopoietic stem cell repopulating ability can be maintained in vitro by some primary endothelial cells. Exp Hematol 2004;32:1226–37. [233] Hooper AT, Butler JM, Nolan DJ, Kranz A, Iida K, Kobayashi M, et al. Engraftment and reconstitution of hematopoiesis is dependent on VEGFR2-mediated regeneration of sinusoidal endothelial cells. Cell Stem Cell 2009;4:263–74. [234] Kobayashi H, Butler JM, O'Donnell R, Kobayashi M, Ding BS, Bonner B, Chiu VK, Nolan DJ, Shido K, et al. Angiocrine factors from Akt-activated endothelial cells balance self-renewal and differentiation of haematopoietic stem cells. Nat Cell Biol 2010;12:1046–56. [235] Slayton WB, Li XM, Butler J, Guthrie SM, Jorgensen ML, Wingard JR, et al. The role of the donor in the repair of the marrow vascular niche following hematopoietic stem cell transplant. Stem Cells 2007;25:2945–55. [236] Kiel MJ, Morrison SJ. Uncertainty in the niches that maintain haematopoietic stem cells. Nat Rev Immunol 2008;8:290–301. [237] Ellis SL, Grassinger J, Jones A, Borg J, Camenisch T, Haylock D, et al. The relationship between bone, hemopoietic stem cells, and vasculature. Blood 2011;118:1516–24. [238] Chow DC, Wenning LA, Miller WM, Papoutsakis ET. Modeling pO(2) distributions in the bone marrow hematopoietic compartment. II. Modified Kroghian models. Biophys J 2001;81:685–96. [239] Kubota Y, Takubo K, Suda T. Bone marrow long label-retaining cells reside in the sinusoidal hypoxic niche. Biochem Biophys Res Commun 2008;366:335–9. [240] 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;104:5431–6. [241] Winkler IG, Barbier V, Wadley R, Zannettino AC, Williams S, Levesque JP, et al. Positioning of bone marrow hematopoietic and stromal cells relative to blood flow in vivo: serially reconstituting hematopoietic stem cells reside in distinct nonperfused niches. Blood 2010;116:375–85.

II.  CELLULAR, MOLECULAR, AND DEVELOPMENTAL BIOLOGY OF BONE

Summary

[242] Semenza GL, Nejfelt MK, Chi SM, Antonarakis SE. Hypoxiainducible nuclear factors bind to an enhancer element located 3' to the human erythropoietin gene. Proc Natl Acad Sci U. S. A. 1991; 88:5680–4. [243] Feldser D, Agani F, Iyer NV, Pak B, Ferreira G, Semenza GL, et al. Reciprocal positive regulation of hypoxia-inducible factor 1alpha and insulin-like growth factor 2. Cancer Res 1999;59:3915–8. [244] Cormier-Regard S, Nguyen SV, Claycomb WC. Adrenomedullin gene expression is developmentally regulated and induced by hypoxia in rat ventricular cardiac myocytes. J Biol Chem 1998;273:17787–92. [245] Krishnamachary B, Berg-Dixon S, Kelly B, Agani F, Feldser D, Ferreira G, et al. Regulation of colon carcinoma cell invasion by hypoxia-inducible factor 1. Cancer Res 2003;63:1138–43. [246] Levy AP, Levy NS, Wegner S, Goldberg MA. Transcriptional regulation of the rat vascular endothelial growth factor gene by hypoxia. J Biol Chem 1995;270:13333–40. [247] Rehn M, Olsson A, Reckzeh K, Diffner E, Carmeliet P, Landberg G, et al. Hypoxic induction of vascular endothelial growth factor regulates murine hematopoietic stem cell function in the lowoxygenic niche. Blood 2011;118:1534–43. [248] Ceradini DJ, Kulkarni AR, Callaghan MJ, Tepper OM, Bastidas N, Kleinman ME, et al. Progenitor cell trafficking is regulated by hypoxic gradients through HIF-1 induction of SDF-1. Nat Med 2004;10:858–64. [249] Staller P, Sulitkova J, Lisztwan J, Moch H, Oakeley EJ, Krek W, et al. Chemokine receptor CXCR4 downregulated by von ­Hippel-Lindau tumour suppressor pVHL. Nature 2003;425:307–11. [250] Kirito K, Hu Y, Komatsu N. HIF-1 prevents the overproduction of mitochondrial ROS after cytokine stimulation through induction of PDK-1. Cell Cycle 2009;8:2844–9. [251] Lin Q, Lee YJ, Yun Z. Differentiation arrest by hypoxia. J Biol Chem 2006;281:30678–83. [252] Danet GH, Pan Y, Luongo JL, Bonnet DA, Simon MC. Expansion of human SCID-repopulating cells under hypoxic conditions. J Clin Invest 2003;112:126–35. [253] Smith S, Broxmeyer HE. The influence of oxygen tension on the long-term growth in vitro of haematopoietic progenitor cells from human cord blood. Br J Haematol 1986;63:29–34. [254] Broxmeyer HE, Cooper S, Gabig T. The effects of oxidizing species derived from molecular oxygen on the proliferation in vitro of human granulocyte-macrophage progenitor cells. Ann N Y Acad Sci 1989;554:177–84. [255] Broxmeyer HE, Cooper S, Lu L, Miller ME, Langefeld CD, Ralph P, et al. Enhanced stimulation of human bone marrow macrophage colony formation in vitro by recombinant human macrophage colony-stimulating factor in agarose medium and at low oxygen tension. Blood 1990;76:323–9. [256] Takubo K, Goda N, Yamada W, Iriuchishima H, Ikeda E, Kubota Y, et al. Regulation of the HIF-1alpha level is essential for hematopoietic stem cells. Cell Stem Cell 2010;7:391–402. [257] Miharada K, Karlsson G, Rehn M, Rorby E, Siva K, Cammenga J, et al. Cripto regulates hematopoietic stem cells as a hypoxicniche-related factor through cell surface receptor GRP78. Cell Stem Cell 2011;9:330–44. [258] Saller MM, Prall WC, Docheva D, Schonitzer V, Popov T, Anz D, et al. Increased stemness and migration of human mesenchymal stem cells in hypoxia is associated with altered integrin expression. Biochem Biophys Res Commun 2012;423:379–85. [259] Huang J, Deng F, Wang L, Xiang XR, Zhou WW, Hu N, et al. ­Hypoxia induces osteogenesis-related activities and expression of core binding factor alpha1 in mesenchymal stem cells. Tohoku J Exp Med 2011;224:7–12.

147

[260] Fu WL, Jia ZQ, Wang WP, Zhang JY, Fu X, Duan XN, et al. ­Proliferation and apoptosis property of mesenchymal stem cells derived from peripheral blood under the culture conditions of ­hypoxia and serum deprivation. Chin Med J (Engl) 2011;124:3959–67. [261] Adesida AB, Mulet-Sierra A, Jomha NM. Hypoxia mediated isolation and expansion enhances the chondrogenic capacity of bone marrow mesenchymal stromal cells. Stem Cell Res Ther 2012;3:9. [262] Zou D, Zhang Z, He J, Zhu S, Wang S, Zhang W, et al. Repairing critical-sized calvarial defects with BMSCs modified by a constitutively active form of hypoxia-inducible factor-1alpha and a phosphate cement scaffold. Biomaterials 2011;32:9707–18. [263] Valorani MG, Montelatici E, Germani A, Biddle A, D'Alessandro D, Strollo R, Patrizi MP, Lazzari L, Nye E, Otto WR, Pozzilli P, et al. Pre-culturing human adipose tissue mesenchymal stem cells under hypoxia increases their adipogenic and osteogenic differentiation potentials. Cell Prolif 2012;45:225–38. [264] Wang Y, Li J, Wang Y, Lei L, Jiang C, An S, et al. Effects of hypoxia on osteogenic differentiation of rat bone marrow mesenchymal stem cells. Mol Cell Biochem 2012;362:25–33. [265] Chen D, Tian W, Li Y, Tang W, Zhang C. Osteoblast-specific transcription factor osterix (Osx) and HIF-1alpha cooperatively regulate gene expression of vascular endothelial growth factor (VEGF). Biochem Biophys Res Commun 2012;424:176–81. [266] Duval E, Bauge C, Andriamanalijaona R, Benateau H, Leclercq S, Dutoit S, et al. Molecular mechanism of hypoxia-induced chondrogenesis and its application in in vivo cartilage tissue engineering. Biomaterials 2012;33:6042–51. [267] Wu C, Zhou Y, Fan W, Han P, Chang J, Yuen J, et al. ­Hypoxia-mimicking mesoporous bioactive glass scaffolds with controllable cobalt ion release for bone tissue engineering. ­Biomaterials 2012;33:2076–85. [268] Zou D, Zhang Z, He J, Zhang K, Ye D, Han W, et al. Blood ­vessel formation in the tissue-engineered bone with the constitutively active form of HIF-1alpha mediated BMSCs. Biomaterials 2012;33:2097–108. [269] 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;3:364–6. [270] Mendez-Ferrer S, Lucas D, Battista M, Frenette PS. Haematopoietic stem cell release is regulated by circadian oscillations. ­Nature 2008;452:442–7. [271] Spiegel A, Shivtiel S, Kalinkovich A, Ludin A, Netzer N, ­Goichberg P, et al. Catecholaminergic neurotransmitters regulate migration and repopulation of immature human CD34+ cells through Wnt signaling. Nat Immunol 2007;8:1123–31. [272] Kawamori Y, Katayama Y, Asada N, Minagawa K, Sato M, ­Okamura A, et al. Role for vitamin D receptor in the neuronal control of the hematopoietic stem cell niche. Blood 2010;116:5528–35. [273] Cho Y, Noshiro M, Choi M, Morita K, Kawamoto T, Fujimoto K, et al. The basic helix-loop-helix proteins differentiated embryo chondrocyte (DEC) 1 and DEC2 function as corepressors of retinoid X receptors. Mol Pharmacol 2009;76:1360–9. [274] Yamazaki S, Ema H, Karlsson G, Yamaguchi T, Miyoshi H, ­Shioda S, et al. Nonmyelinating schwann cells maintain hematopoietic stem cell hibernation in the bone marrow niche. Cell 2011;147:1146–58. [275] Larsson J, Karlsson S. The role of Smad signaling in hematopoiesis. Oncogene 2005;24:5676–92. [276] Yamazaki S, Iwama A, Takayanagi S, Eto K, Ema H, Nakauchi H, et al. TGF-beta as a candidate bone marrow niche signal to induce hematopoietic stem cell hibernation. Blood 2009;113:1250–6. [277] Keating A. Mesenchymal stromal cells: new directions. Cell Stem Cell 2012;10(6):709–16.

II.  CELLULAR, MOLECULAR, AND DEVELOPMENTAL BIOLOGY OF BONE