0163-769X/00/$03.00/0 Endocrine Reviews 21(2): 115–137 Copyright © 2000 by The Endocrine Society Printed in U.S.A.

Birth and Death of Bone Cells: Basic Regulatory Mechanisms and Implications for the Pathogenesis and Treatment of

STAVROS C. MANOLAGAS Division of Endocrinology & Metabolism, Center for Osteoporosis & Metabolic Bone Diseases, University of Arkansas for Medical Sciences, and the Central Arkansas Veterans Healthcare System, Little Rock, Arkansas 72205, USA

ABSTRACT these cells. The purpose of this article is 3-fold: 1) to review the role The adult skeleton regenerates by temporary cellular structures and the molecular mechanism of action of regulatory molecules, such that comprise teams of juxtaposed and osteoblasts and as cytokines and hormones, in and osteoblast birth and replace periodically old bone with new. A considerable body of evi- apoptosis; 2) to review the evidence for the contribution of changes in dence accumulated during the last decade has shown that the rate of bone cell birth or death to the pathogenesis of the most common forms genesis of these two highly specialized cell types, as well as the of osteoporosis; and 3) to highlight the implications of bone cell birth prevalence of their apoptosis, is essential for the maintenance of bone and death for a better understanding of the mechanism of action and homeostasis; and that common metabolic bone disorders such as os- efficacy of present and future pharmacotherapeutic agents for osteo- teoporosis result largely from a derangement in the birth or death of porosis. (Endocrine Reviews 21: 115–137, 2000)

I. Introduction B. Bisphosphonates and calcitonin II. Physiological Bone Regeneration C. Novel pharmacotherapeutic strategies A. Remodeling by the basic multicellular unit (BMU) XI. Summary and Conclusions III. Osteoblastogenesis and Osteoclastogenesis A. Growth factors and their antagonists B. Cytokines I. Introduction C. Systemic hormones And Athena lavished a marvelous splendor on the prince so that D. Adhesion molecules all the people gazed in wonder as he came forward. The elders IV. Reciprocal Relationship Between Osteoblastogenesis making way as he took his father’s seat. The first to speak was an and Adipogenesis old lord, Aegyptius, stooped with age, who knew the world by V. Serial and Parallel Models of Osteoblast and Osteoclast heart. Development VI. Function of the Mature Cells Homer, the Odyssey: translation by Robert Fagles A. Osteoblasts B. Osteocytes OSS OF height (stooping), Dowager’s hump, and ky- C. Lining cells L phosis are some of the most visible signs of old age in D. Osteoclasts humans. The primary reason for these involutional changes VII. Death of Bone Cells by Apoptosis is a progressive loss of bone mass that affects the axial (pri- VIII. Regulation of Bone Cell Proliferation and Activity marily trabecular) as well as the appendicular (primarily IX. Pathogenesis of Osteoporosis cortical) skeleton. Loss of bone mass, along with microar- A. Sex steroid deficiency chitectural deterioration of the skeleton, leads to enhanced B. Senescence bone fragility and increased fractures—the C. Glucocorticoid excess known as osteoporosis (1). Both men and women start losing X. Pharmacotherapeutic Implications of Osteoblast and bone in their 40s. However, women experience a rapid phase Osteocyte Apoptosis of loss during the first 5–10 yr after menopause, due to the A. Intermittent PTH administration loss of estrogen (2). In men this phase is obscure, since there is only a slow and progressive decline in sex steroid pro- duction; hence, the loss of bone in men is linear and slower Address reprint requests to: Stavros C. Manolagas, M.D., Ph.D., Center for Osteoporosis & Metabolic Bone Diseases, Division of (3). In addition to losing bone faster at the early postmeno- Endocrinology & Metabolism, University of Arkansas Medical School, pausal years, women also accumulate less skeletal mass than 4301 West Markam Street, Little Rock, Arkansas 72205 USA. E-mail: men during growth, particularly in puberty, resulting in [email protected] smaller bones with thinner cortices and smaller diameter. *Research from the author’s laboratory described in this review was supported by the NIH (P01-AG13918, R01-AR43003), the Department of Consequently, the incidence of bone fractures is 2- to 3-fold Veterans Affairs (merit grant and a research enhancement award pro- higher in women as compared with men (4). gram, REAP), and the 1999 Allied Signal Award for Research on Aging. In addition to sex steroid deficiency and the aging process

115 116 MANOLAGAS Vol. 21, No. 2 itself, loss of bone mass is accentuated when several other prevent accumulation of old bone. Remodeling, with positive conditions are present. The most common are chronic glucocor- balance, does occur in the growing skeleton as well. Its pur- ticoid excess (5), particularly its iatrogenic form, hyperthyroid- pose, quite different from those proposed for the adult skel- ism as well as inappropriately high T4 replacement, alcoholism, eton, is to expand the marrow cavity while increasing tra- prolonged immobilization, gastrointestinal disorders, hyper- becular thickness (12). calciuria, some types of malignancy, and cigarette smoking (6). Bone loss and eventually fractures are the hallmarks of A. Remodeling by the basic multicellular unit (BMU) osteoporosis, regardless of the underlying cause or causes. The bone loss associated with normal aging in women has Removal of bone (resorption) is the task of osteoclasts. been divided into two phases: one that is due to menopause Formation of new bone is the task of osteoblasts. Bone re- and one that is due to aging and affects men as well (7, 8). sorption and bone formation, however, are not separate, In elderly women these two phases eventually overlap, mak- independently regulated processes. In the uninjured adult ing it difficult to distinguish the effect of sex steroid defi- skeleton, all osteoclasts and osteoblasts belong to a unique ciency from the effect of the aging process itself. The effect temporary structure, known as a basic multicellular unit or of the aging process itself is also frequently obscured because BMU (13). Although during modeling one cannot distinguish of secondary hyperparathyroidism (9), resulting from im- anatomical units analogous to BMU per se, sculpting of the paired calcium absorption from the intestine with advancing growing skeleton requires spatial and temporal orchestra- age (Ͼ75 yr old). The bone loss that is due to glucocorticoid tion of the destination of osteoblasts and osteoclasts, albeit excess shares several features with the bone loss due to with different rules and coordinates to those operating in the senescence, but also has unique features of its own. None- BMU of the remodeling skeleton. The BMU, approximately theless, as is the case with the other types of bone loss, the 1–2 mm long and 0.2–0.4 mm wide, comprises a team of heterogeneity of the underlying conditions, some of which osteoclasts in the front, a team of osteoblasts in the rear, a (e.g., postmenopausal state, rheumatoid arthritis, etc.) inde- central vascular capillary, a nerve supply, and associated pendently contribute to skeletal deterioration, can distort the connective tissue (13). In healthy human adults, 3–4 million clinical and histological picture (10). Irrespective of the over- BMUs are initiated per year and about 1 million are operating lap, it is important to recognize that the pathogenetic mech- at any moment (Table 1). Each BMU begins at a particular anisms are quite distinct in the various forms of osteoporosis place and time (origination) and advances toward a target, and that sex hormone deficiency and aging have indepen- which is a region of bone in need of replacement, and for a dent effects. variable distance beyond its target (progression) and even- During the last few years, there have been significant ad- tually comes to rest (termination) (10). In cortical bone, the vances in our understanding of the pathogenetic mecha- BMU travels through the bone, excavating and replacing a nisms responsible for the bone loss associated with sex ste- tunnel. In cancellous bone, the BMU moves across the tra- roid deficiency, old age, and glucocorticoid excess. All these becular surface, excavating and replacing a trench. In both conditions do not cause loss of bone mass by turning on a situations, the cellular components of the BMUs maintain a completely new process. Instead, they cause a derangement well orchestrated spatial and temporal relationship with in the normal process of bone regeneration. Therefore, to each other. Osteoclasts adhere to bone and subsequently understand the pathogenesis of osteoporosis and rationalize remove it by acidification and proteolytic digestion. As the its treatment, one must first appreciate the basic principles of BMU advances, osteoclasts leave the resorption site and os- physiological bone regeneration. teoblasts move in to cover the excavated area and begin the process of new bone formation by secreting osteoid, which is eventually mineralized into new bone. The lifespan of the BMU is 6–9 months; much longer than II. Physiological Bone Regeneration the lifespan of its executive cells (Table 1). Therefore, con- The skeleton is a highly specialized and dynamic organ tinuous supply of new osteoclasts and osteoblasts from their that undergoes continuous regeneration. It consists of highly respective progenitors in the bone marrow is essential for the specialized cells, mineralized and unmineralized connective origination of BMUs and their progression on the bone sur- tissue matrix, and spaces that include the bone marrow cav- TABLE 1. Vital statistics of adult bone remodelinga ity, vascular canals, canaliculi, and lacunae. During devel- opment and growth, the skeleton is sculpted to achieve its ● Lifespan of BMU ϳ6–9 months shape and size by the removal of bone from one site and ● Speed ϳ25 ␮m/day ● Bone volume replaced by a single BMU ϳ0.025 mm3 deposition at a different one; this process is called modeling. ● Lifespan of osteoclasts ϳ2 weeks Once the skeleton has reached maturity, regeneration con- ● Lifespan of osteoblasts (active) ϳ3 months tinues in the form of a periodic replacement of old bone with ● Interval between successive remodeling events at the same new at the same location (11). This process is called remod- location ϳ2–5 years. ● ϳ b eling and is responsible for the complete regeneration of the Rate of turnover of whole skeleton 10% per year adult skeleton every 10 yr. The purpose of remodeling in the a From A. Michael Parfitt (13) b adult skeleton is not entirely clear, although in bones that are The 10% per year approximation for the entire skeleton is based on an average 4% turnover per year in cortical bone, which represents load bearing, remodeling most likely serves to repair fatigue roughly 75% of the entire skeleton; and an average 28% turnover per damage and to prevent excessive aging and its consequences. year in trabecular bone, which represents roughly 25% of the skeleton Hence, the most likely purpose of bone remodeling is to (0.75 ϫ 4 ϭ 3 and 0.25 ϫ 28 ϭ 7; 3 ϩ 7 ϭ 10). April, 2000 BIRTH AND DEATH OF BONE CELLS 117 face. Consequently, the balance between the supply of new the gene encoding an osteoblast-specific transcription factor, cells and their lifespan are key determinants of the number known as osteoblast specific factor 2 (Osf2) or core binding of either cell type in the BMU and the work performed by factor a1 (Cbfa1), hereafter referred to as Cbfa1 (24). In turn, each type of cells and are critical for the maintenance of bone Cbfa1 activates osteoblast-specific genes such as osteopontin, homeostasis. bone sialoprotein, type I collagen, and osteocalcin. The im- portance of Cbfa1 for osteoblasts has been highlighted by the evidence that knockout of the Cbfa1 gene in mice prevents III. Osteoblastogenesis and Osteoclastogenesis osteoblast development (25, 26). In addition to Cbfa1, BMP-4 Both osteoblasts and osteoclasts are derived from precur- induces a homeobox-containing gene, distal-less 5(Dlx5), sors originating in the bone marrow. The precursors of os- which also seems to act as a transcription factor, probably as teoblasts are multipotent mesenchymal stem cells, which a heterodimer with another homeobox-containing protein also give rise to bone marrow stromal cells, chondrocytes, (Msx2). Like Cbfa1, Dlx5 regulates the expression of osteo- muscle cells, and adipocytes (14–16), whereas the precursors blast-specific genes such as osteocalcin and alkaline phos- of osteoclasts are hematopoietic cells of the monocyte/mac- phatase, as well as mineralization (27–29). Other factors such ␤ ␤ rophage lineage (17, 18). Long before these cells could be as transforming growth factor (TGF ), platelet-derived cultured, the existence of multipotent mesenchymal stem growth factor (PDGF), insulin-like growth factors (IGFs), and cells was suspected (19), based on the evidence that fibro- members of the fibroblast growth factor (FGF) family can all blastic colonies formed in cultures of adherent bone marrow stimulate osteoblast differentiation (30, 31). However, ␤ cells can differentiate, under the appropriate stimuli, into whereas TGF , PDGF, FGF, and IGFs are able to influence the each of the above mentioned cells; these progenitors were replication and differentiation of committed osteoblast pro- named colony forming unit fibroblasts (CFU-F). When genitors toward the osteoblastic lineage, they cannot induce CFU-F are cultured in the presence of ␤-glycerophosphate osteoblast differentiation from uncommitted progenitor and ascorbic acid, the majority of the colonies form a min- cells. eralized bone nodule; these bone-forming colonies are In addition to growth factors, bone cells produce proteins known as CFU-osteoblast (CFU-OB) (16). Osteoblast progen- that modulate the activity of growth factors either by binding itors may originate not only from stromal mesenchymal pro- to them and thereby preventing interaction with their re- genitors of the marrow, but also pericytes — mesenchymal ceptors, by competing for the same receptors, or by promot- cells adherent to the endothelial layer of vessels (20). ing the activity of a particular factor. For example, osteoblasts Whereas osteoclast precursors reach bone from the circula- produce several IGF-binding proteins (IGFBPs). Of these, tion, osteoblast precursors most likely reach bone by migra- IGFBP-4 binds to IGF and blocks its action, whereas IGFBP-5 tion of progenitors from neighboring connective tissues. promotes the stimulatory effects of IGF on osteoblasts (30). The development and differentiation of osteoblasts and During the last few years, several proteins able to antagonize osteoclasts are controlled by growth factors and cytokines BMP action have also been discovered. Of them, noggin, produced in the bone marrow microenvironment as well as chordin, and cerberus were initially found in the Spemann adhesion molecules that mediate cell-cell and cell-matrix organizer of the Xenopus embryo and shown to be essential interactions. Several systemic hormones as well as mechan- for neuronal or head development (32–35). Noggin and chor- ical signals also exert potent effects on osteoclast and osteo- din inhibit the action of BMPs by binding directly and with blast development and differentiation. Although many de- high affinity with the latter proteins (36, 37). Such binding is tails remain to be established concerning the operation of this highly specific for BMP-2 and 4, as noggin binds BMP-7 with ␤ network, a few themes have emerged (21). First, several of the very low affinity and does not bind TGF or IGF-I. Addition growth factors and cytokines control each other’s production of human recombinant noggin to bone marrow cell cultures in a cascade fashion and, in some instances, form positive and from normal adult mice inhibits not only osteoblast, but also negative feedback loops. Second, there is extensive func- osteoclast, formation, and these effects can be reversed by tional redundancy among them. Third, some of the same exogenous BMP-2 (23). Consistent with this evidence, BMP-2 factors are capable of influencing the differentiation of both and -4 and BMP-2/4 receptor transcripts and proteins are osteoblasts and osteoclasts. Fourth, systemic hormones in- found in bone marrow cultures and in bone marrow-derived fluence the process of osteoblast and osteoclast formation via stromal/osteoblastic cell lines, as well as in murine adult their ability to control the production and/or action of local whole bone. Noggin expression has also been documented in mediators. all these cell preparations. These findings indicate that BMP-2 and -4 are expressed in the bone marrow in postnatal A. Growth factors and their antagonists life and serve to maintain the continuous supply of osteo- blasts. The only factors capable of initiating osteoblastogenesis from uncommitted progenitors are bone morphogenetic pro- B. Cytokines teins (BMPs) (22). BMPs have been long implicated in skeletal development during embryonic life and fracture healing. Since the early stages of hematopoiesis and osteoclasto- More recently, it has become apparent that BMPs, and in genesis proceed along identical pathways, it is not surprising particular BMP-2 and –4, also initiate the commitment of that a large group of cytokines and colony-stimulating fac- mesenchymal precursors of the adult bone marrow to the tors that are involved in hematopoiesis also affect osteoclast osteoblastic lineage (23). BMPs stimulate the transcription of development (38). This group includes the interleukins IL-1, 118 MANOLAGAS Vol. 21, No. 2

IL-3, IL-6, IL-11, leukemia inhibitory factor (LIF), oncostatin mature osteoblast phenotype, characterized by increased al- M (OSM), ciliary neurotropic factor (CNTF), tumor necrosis kaline phosphatase and osteocalcin expression, and a con- factor (TNF), granulocyte macrophage-colony stimulating comitant decrease in proliferation (53, 54). Moreover, IL-6 factor (GM-CSF), M-CSF, and c-kit ligand. As opposed to the type cytokines stimulate the development of osteoblasts above mentioned cytokines that stimulate osteoclast devel- from noncommitted embryonic fibroblasts obtained from opment, IL-4, IL-10, IL-18, and interferon-␥ inhibit osteoclast 12-day-old murine fetuses (55). Consistent with the in vitro development. In the case of IL-18 the effect is mediated evidence, several in vivo studies have demonstrated in- through GM-CSF (39). creased bone formation in transgenic mice overexpressing IL-6 has attracted particular attention because of evidence OSM or LIF (56, 57). that it plays a pathogenetic role in several disease states TGF␤ is another example of a factor affecting both bone characterized by accelerated bone remodeling and excessive formation and (58). Thus, in addition to its focal or systemic bone resorption (40). IL-6 is produced at ability to stimulate osteoblast differentiation, TGF␤ increases high levels by cells of the stromal/osteoblastic lineage in bone resorption by stimulating osteoclast formation. Injec- response to stimulation by a variety of other cytokines and tion of TGF␤ into the subcutaneous tissue that overlies the growth factors such as IL-1, TNF, TGF␤, PDGF, and IGF-II calvaria of adult mice causes increased bone resorption ac- (41–43). Binding of IL-6 or other members of the same cy- companied by the development of unusually large oste- tokine family (IL-11, LIF, OSM) to cytokine-specific cell sur- oclasts, as well as increased bone formation. The effects of face receptors (in the case of IL-6, the IL-6R␣) causes recruit- TGF␤ might be mediated by other cytokines involved in ment and homo- or heterodimerization of the signal osteoclastogenesis as TGF␤ can stimulate their production. transducing protein gp130, which is then tyrosine phosphor- Mice lacking the TGF␤1 gene due to targeted disruption ylated by members of the Janus family of tyrosine kinases exhibit excessive production of inflammatory cells, suggest- (JAKs) (44). This event results in tyrosine phosphorylation of ing that this growth factor normally operates to suppress several downstream signaling molecules, including mem- hematopoiesis (59). bers of the signal transducers and activators of transcription (STAT) family of transcription factors (45, 46). Phosphory- C. Systemic hormones lated STATs, in turn, undergo homo- and heterodimerization and translocate to the nucleus where they activate cytokine- The two principal hormones of the calcium homeostatic ␣ responsive gene transcription (47). The -subunit of the IL-6 system, namely PTH and l,25-dihydroxyvitamin D3 [1,25- receptor also exists in a soluble form (sIL-6R), but unlike most (OH)2D3], are potent stimulators of osteoclast formation (17, soluble cytokine receptors, it functions as an agonist by bind- 60). The ability of these hormones to stimulate osteoclast ing to IL-6 and then interacting with membrane-associated development and to regulate calcium absorption and excre- gp130 to stimulate JAK/STAT signaling (44). On the other tion from the intestine and kidney, respectively, are the key hand, the soluble form of gp130 blocks IL-6 action (48). elements of extracellular calcium homeostasis. Calcitonin, Alone or in concert with other agents, IL-6 stimulates the third of the classical bone-regulating hormones, inhibits osteoclastogenesis and promotes bone resorption. The cells osteoclast development and activity and promotes osteoclast that mediate the actions of the IL-6 type cytokines on oste- apoptosis. Although the antiresorptive properties of calcito- oclast formation appear to be the stromal/osteoblastic cells, nin have been exploited in the management of bone diseases as stimulation of IL-6R␣ expression on these cells allows with increased resorption, the role of this hormone in bone them to support osteoclast formation in response to IL-6 (49). physiology in humans, if any, remains questionable (61–63). These findings indicate that the osteoclastogenic property of PTH, PTH-related peptide, and 1,25-(OH)2D3 stimulate the IL-6 depends not only on its ability to act directly on hema- production of IL-6 and IL-11 by stromal/osteoblastic cells topoietic osteoclast progenitors, but also on the activation of (49, 64–66). Several other hormones, including estrogen, an- gp130 signaling in the stromal/osteoblastic cells that provide drogen, glucocorticoids, and T4, exert potent regulatory in- essential support for osteoclast formation. STAT3 activation fluences on the development of osteoclasts and osteoblasts in stromal/osteoblastic cells is essential for gp130-mediated by regulating the production and/or action of several cyto- osteoclast formation (50). Despite the effects of IL-6 on os- kines (21, 64, 67–69). teoclastogenesis in experimental in vitro systems, IL-6 is not required for osteoclastogenesis in vivo under normal phys- D. Adhesion molecules iological conditions. In fact, osteoclast formation is unaf- fected in sex steroid-replete mice treated with a neutralizing In addition to autocrine, paracrine, and endocrine signals, anti-IL-6 antibody, or in IL-6-deficient mice (51, 52). The most cell-cell and cell-matrix interactions are also required for the likely explanation for this is that the ␣-subunit of the IL-6 development of osteoclasts and osteoblasts (70–72). Such receptor in bone is a limiting factor, and that both a change interactions are mediated by proteins expressed on the sur- in the receptor and the cytokine are required for the IL-6- face of these cells and are responsible for contact between mediated increased osteoclastogenesis, seen in pathological osteoclast precursors with stromal/osteoblastic cells and fa- states. cilitation of the action of paracrine factors anchored to the IL-6 type cytokines are capable of influencing the differ- surface of cells that are required for bone cell development. entiation of osteoblasts as well. Thus, receptors for these Adhesion molecules are also involved in the migration of cytokines are expressed on a variety of stromal/osteoblastic osteoblast and osteoclast progenitors from the bone marrow cells, and ligand binding induces progression toward a more to sites of bone remodeling as well as the cellular polarization April, 2000 BIRTH AND DEATH OF BONE CELLS 119 of osteoclasts and the initiation and cessation of osteoclastic suppresses the muscle cell phenotype when transfected into bone resorption. More important, for the purpose of this G8 myoblastic cells (95). Taken together, these findings review, adhesion molecules play a role in the control of strongly suggest that PPAR␥2 plays a hierarchically domi- osteoblast and osteoclast development and apoptosis (73–77). nant role in the determination of the fate of mesenchymal The list of adhesion molecules that influence bone cell progenitors, due to its ability to inhibit the expression of development and function includes the integrins, particu- other lineage-specific transcription factors. Studies with a ␣ ␤ ␣ ␤ larly v 3 and 2 1, selectins, and cadherins, as well as a clonal cell line (2T3) suggest that BMP-2 induces osteoblast family of transmembrane proteins containing a disintegrin or adipocyte differentiation in mesenchymal precursors, de- and metalloprotease domain (ADAMS). Each of these pro- pending on whether the BMP receptor type IA or IB is ac- teins recognizes distinct ligands. For example, some integrins tivated. Therefore, BMP receptors may also play a critical role recognize a specific amino acid sequence (RGD) present in in both the specification and reciprocal differentiation of collagen, fibronectin, osteopontin, thrombospondin, bone osteoblast and adipocyte progenitors (96). sialoprotein, and vitronectin (78).

V. Serial and Parallel Models of Osteoblast and IV. Reciprocal Relationship Between Osteoclast Development Osteoblastogenesis and Adipogenesis Even though millions of small packets of bone are con- The cells that comprise the bone marrow stroma can serve stantly remodeled, bone mass is preserved thanks to a re- several diverse functions including support of hematopoiesis markably tight balance between the amount of bone resorbed and osteoclastogenesis, fat accumulation, and bone forma- and formed during each cycle of remodeling. In any estab- tion (79). This functional adaptation is apparently accom- lished BMU, bone resorption and formation are happening plished by the plasticity of some of the stem cell progeny as at the same time; new osteoblasts assemble only at sites exemplified by the ability of stromal cells to convert between where osteoclasts have recently completed resorption, a phe- the osteoblast and adipocyte phenotype. Thus, a stromal cell nomenon referred to as coupling, and formation begins to type known as the Westen-Bainton cell exhibits PTH recep- occur while resorption advances. The end result is a new tors and high alkaline phosphatase activity and gives rise to packet of bone, either a cylindrical osteon or Haversian sys- osteoblasts during fetal development and in hyperparathy- tem, or a plate-like hemiosteon, that has replaced the older roidism. On the other hand, when marrow hematopoietic bone that was removed (97). activity is reduced using chemotherapeutic agents, these As the BMU advances, cells are successively recruited at cells convert into adipocytes and can support myeloid cell each new cross-sectional location. Osteoblasts do not arrive production (80–84). Further, adipocytes isolated by limiting until the osteoclasts have moved on. However, during the dilution from cultures of rabbit bone marrow can form bone longitudinal progression of the BMU as a whole, new oste- in diffusion chamber implants (85). Conversely, addition of oclasts and new osteoblasts are needed simultaneously, al- certain fatty acids to cultures of osteoblastic cells causes them though not at the same location. Two models of osteoblast to differentiate into adipocyte-like cells (86). recruitment, a serial and a parallel (Fig. 1), can explain the It is likely that interconversion of stromal cells among distinction between the cross-sectional and longitudinal phenotypes, as well as commitment to a particular lineage events during BMU progression (38). According to the serial with suppression of alternative phenotypes, is governed by model, factors released from resorbed bone or the local in- specific transcription factors. Indeed, Cbfa1 is required for crease in mechanical strain resulting from bone resorption, commitment of mesenchymal progenitors to the osteoblast stimulate osteoblast precursor cell proliferation and differ- lineage. Mice that are deficient in this factor lack osteoblasts entiation (98–100). According to the parallel model, osteo- and mineralized bone matrix (26); and expression of Cbfa1 blast and osteoclast precursor proliferation and differentia- in fibroblastic cells induces transcription of osteoblast- tion occur concurrently in response to whatever signal specific genes (24). On the other hand, CCAAT/enhancer conveys the need for initiation of new BMUs, and whatever binding protein ␣ (C/EBP␣), C/EBP␤, and C/EBP␦, as well hormone prolongs their progression (10, 38). With either as peroxisome proliferator activated receptor ␥1 (PPAR␥1) model, new osteoblasts must be directed to the right location. and PPAR␥2 orchestrate adipocyte differentiation (87–90). Concurrent osteoblast and osteoclast production makes Introduction of C/EBP␣ in fibroblastic cells induces adipo- teleological sense as at least one of the means of maintaining cyte differentiation (91, 92), and transfection of fibroblastic a balance between bone formation and resorption under cells with PPAR␥2 and subsequent activation with an ap- normal conditions. In support of the existence of a parallel propriate ligand causes the development of adipocytes (93). pathway of osteoblast and osteoclast formation, it is well Using clonal cell lines isolated from the murine bone mar- established that osteoclasts cannot be formed in vitro unless row, it has been demonstrated that PPAR␥2 can convert appropriate stromal cells, analogous to the bone marrow stromal cells from a plastic osteoblastic phenotype that re- stromal cells that support hematopoiesis, are present to pro- versibly expresses adipocyte characteristics to terminally dif- vide essential support. The precise phenotype of the cells that ferentiated adipocytes. Moreover, PPAR␥2 suppresses the support osteoclast development remains unknown, but they expression of Cbfa1 and thereby osteoblast-specific genes are clearly related to both the osteoblast and the bone mar- (94). Similar to the inhibitory effect of PPAR␥2 on the os- row stromal/adipocytic lineages (101–104). Interestingly, teoblast phenotype, the combination of PPAR␥ and C/EBP␣ bone marrow-derived cells with both osteoblastic and adi- 120 MANOLAGAS Vol. 21, No. 2

FIG. 1. Serial and parallel models of os- teoblast and osteoclast development. For explanation, please see text. PreOC, Preosteoclast; GF, growth fac- tors released from the matrix of resorbed bone; preOB, osteoblast pro- genitors. The expressions of RANK li- gand and RANK on preosteoblasts and preosteoclasts, respectively, are de- picted to indicate their critical contri- bution in osteoclastogenesis and thereby the dependency of osteoclastogenesis on preosteoblastic cells.

pogenic characteristics support the formation of osteoclasts, osteoclast progenitors, while RANK-ligand is expressed in but marrow-derived cells that exhibit a purely osteoblastic or committed preosteoblastic cells and T lymphocytes (108– adipocytic phenotype do not (94). More to the point, noggin, 110). RANK-ligand binds to RANK with high affinity. This a BMP antagonist, blocks not only osteoblastogenesis but interaction is essential and, together with M-CSF, sufficient also osteoclastogenesis in murine bone marrow cultures, in- for osteoclastogenesis. 1,25-(OH)2D3, PTH, PTHrP, gp130 ac- dicating that commitment of mesenchymal progenitors to the tivating cytokines (e.g., IL-6, IL-11), and IL-1 induce the ex- osteoblastic lineage is prerequisite for osteoclastogenesis pression of the RANK-ligand in stromal/osteoblastic cells (23). This evidence suggests that the early less differentiated (50, 107). Osteoprotegerin (OPG), the third of the three pro- progeny of common mesenchymal progenitors of the osteo- teins, unlike the other two, is a secreted disulfide-linked blastic and adipocytic lineage can support osteoclast devel- dimeric glycoprotein. A hydrophobic leader peptide and opment, but more differentiated cells that have committed to three and one-half TNF receptor-like cysteine-rich pseu- either the osteoblast or the adipocyte pathway lose this prop- dorepeats characterize the amino terminus of this protein. erty. It is possible, but as yet untested, that the cells that Unlike other members of the TNF receptor family, OPG does provide support for osteoclast development are a distinct not posses a transmembrane domain. OPG has very potent progeny of mesenchymal progenitors, which displays per- inhibitory effects on osteoclastogenesis and bone resorption manently a phenotype with mixed adipocytic/osteoblastic in vitro and in vivo (111). Consistent with an important role characteristics, but never progresses to a terminally differ- of OPG in the regulation of osteoclast formation, OPG trans- entiated osteoblast or adipocyte. For convenience and lack of genic mice develop , whereas OPG knockout a better term, the cells that support osteoclast development mice exhibit severe osteoporosis (112). The antiosteoclasto- are frequently referred to as stromal/osteoblastic to indicate genic property of OPG is due to its ability to act as a decoy their similarities to both bone marrow stromal cells and os- by binding to RANK-ligand and blocking the RANK-ligand/ teoblasts. RANK interaction. In addition to skeletal metabolism, the In full agreement with the in vitro evidence for the de- RANK/RANK-ligand/OPG circuit may regulate several pendency of osteoclast development on support by cells re- other biological systems. Indeed, OPG is produced by many lated to osteoblasts, mice lacking osteoblasts due to Cbfa1 tissues other than bone, including skin, liver, stomach, in- deficiency also lack osteoclasts (26). In addition, marrow cells testine, lung, heart, kidney, and placenta as well as hema- from SAMP6 mice, a strain with defective osteoblastogenesis, topoietic and immune organs. Consistent with this, mice exhibit decreased osteoclastogenesis (105) and do not exhibit deficient in RANK-ligand completely lacked lymph nodes as the expected increase in osteoclastogenesis nor do they lose well as osteoclasts (113). Moreover, OPG is also a receptor for bone after loss of sex steroids (106). the cytotoxic ligand TRAIL (TNF-related apoptosis-inducing The molecular mechanism of the dependency of osteoclas- ligand) to which it binds with high affinity and inhibits togenesis on cells of the mesenchymal lineage has been elu- TRAIL-mediated apoptosis in lymphocytes (114) and also cidated during the last 2 yr with the discovery of three regulates antigen presentation and T cell activation (115). proteins involved in the TNF signaling pathway (reviewed Osteoblastic cells and T lymphocytes, the two cell types in Ref. 107). Two of these proteins are membrane-bound that express high levels of RANK-ligand, are also the two cell cytokine-like molecules: the receptor activator of nuclear fac- types that express high levels of the osteoblast-specific tran- tor-␬B (NF-␬B) (RANK) and the RANK-ligand. Other names scription factor Cbfa1 (24). More intriguingly, both the mu- used in the literature for RANK are osteoprotegerin ligand rine and human RANK-ligand genes contain two functional (OPG-L) and TRANCE. RANK is expressed in hematopoietic Cbfa1 sites, and mutation of these sites abrogates the tran- April, 2000 BIRTH AND DEATH OF BONE CELLS 121 scriptional activity of the RANK-ligand gene promoter (116). matrix, mature osteoblasts are essential for its mineraliza- Therefore, the cell-specific expression of RANK-ligand in tion, the process of deposition of hydroxyapatite (123, 124). cells of the stromal/osteoblastic lineage and concurrent dif- Osteoblasts are thought to regulate the local concentrations ferentiation of osteoblasts and osteoclasts might be dictated, of calcium and phosphate in such a way as to promote the at least in part, by interaction between an osteoblast-specific formation of hydroxyapatite. In view of the highly ordered, transcription factor and RANK-ligand. BMP 2 and -4 stim- well aligned, collagen fibrils complexed with the noncollag- ulate Cbfa1 expression. Based on these lines of evidence, it enous proteins formed by the osteoblast in lamellar bone, it has been postulated that the molecular underpinnings of the is likely that mineralization proceeds in association with, and control of the rate of bone regeneration and the concurrent perhaps governed by, the heteropolymeric matrix fibrils production of osteoclasts and osteoblasts could well be a themselves. Osteoblasts express relatively high amounts of BMP3Cbfa13RANK-ligand gene expression cascade in alkaline phosphatase, which is anchored to the external sur- cells of the bone marrow stromal/osteoblastic lineage (117, face of the plasma membrane. Alkaline phosphatase has been 118). According to this hypothesis, BMPs may provide the long thought to play a role in bone mineralization. Consistent tonic baseline control of both processes, and thereby the rate with this, deficiency of alkaline phosphatase due to genetic of bone remodeling, upon which other inputs (e.g., biome- defects leads to , a condition characterized chanical, hormonal, etc.) operate. by defective bone mineralization (125). However, the precise Studies with transgenic and knockout animal models as mechanism of mineralization and the exact role of alkaline well as models with spontaneous genetic mutations have phosphatase in this process remain unclear. Bone mineral- identified at least three transcription factors that are required ization lags behind matrix production and, in remodeling for osteoclast differentiation: PU-1, fos, and NF-␬b. A review sites in the adult bone, occurs at a distance of 8–10 ␮m from of the precise role of these factors is beyond the scope of this the osteoblast. Matrix synthesis determines the volume of article, but the reader is referred to a recent excellent review bone but not its density. Mineralization of the matrix in- of the topic (119). creases the density of bone by displacing water, but does not alter its volume.

VI. Function of the Mature Cells B. Osteocytes A. Osteoblasts Some osteoblasts are eventually buried within lacunae The fully differentiated osteoblasts produce and secrete of mineralized matrix. These cells are termed osteocytes proteins that constitute the bone matrix (120). The matrix is and are characterized by a striking stellate morphology, subsequently mineralized under the control of the same cells. reminiscent of the dendritic network of the nervous sys- A major product of the bone-forming osteoblast is type I tem (126, 127). Osteocytes are the most abundant cell type collagen. This polymeric protein is initially secreted in the in bone: there are 10 times as many osteocytes as osteo- form of a precursor, which contains peptide extensions at blasts. Osteocytes are regularly spaced throughout the both the amino-terminal and carboxyl ends of the molecule. mineralized matrix and communicate with each other and The propeptides are proteolytically removed. Further extra- with cells on the bone surface via multiple extensions of cellular processing results in mature three-chained type I their plasma membrane that run along the canaliculi; os- collagen molecules, which then assemble themselves into a teoblasts, in turn, communicate with cells of the bone collagen fibril. Individual collagen molecules become inter- marrow stroma which extend cellular projections onto connected by the formation of pyridinoline cross-links, endothelial cells inside the sinusoids. Thus, a syncytium which are unique to bone. Bone-forming osteoblasts synthe- extends from the entombed osteocytes all the way to the size a number of other proteins that are incorporated into the vessel wall (128) (Fig. 2). As a consequence, the strategic bone matrix, including osteocalcin and osteonectin, which location of osteocytes makes them excellent candidates for constitute 40% to 50% of the noncollagenous proteins of mechanosensory cells able to detect the need for bone bone. Mice deficient in osteocalcin develop a phenotype augmentation or reduction during functional adaptation marked by higher bone mass and improved bone quality, of the skeleton, and the need for repair of microdamage, suggesting that osteocalcin functions normally to limit bone and in both cases to transmit signals leading to the ap- formation without compromising mineralization (121). Con- propriate response; albeit this remains hypothetical (129). versely, mice deficient in osteonectin exhibit decreased os- Osteocytes evidently sense changes in interstitial fluid teoclast and osteoblast numbers and bone remodeling and flow through canaliculi (produced by mechanical forces) profound , suggesting that, under normal condi- and detect changes in the levels of hormones, such as tions, this protein may play a role in the birth or survival of estrogen and glucocorticoids, that influence their survival these cells (122). Other osteoblast-derived proteins include and that circulate in the same fluid (130–132). Therefore, glycosaminoglycans, which are attached to one of two small disruption of the osteocyte network is likely to increase core proteins: PGI (or biglycan) and decorin; the latter has bone fragility. been implicated in the regulation of collagen fibrillogenesis. A number of other minor proteins such as osteopontin, bone C. Lining cells sialoprotein, fibronectin, vitronectin, and thrombospondin serve as attachment factors that interact with integrins. The surface of normal quiescent bone (i.e., bone that is In addition to being the cells that produce the osteoid not undergoing remodeling) is covered by a 1–2-␮m thick 122 MANOLAGAS Vol. 21, No. 2

FIG. 2. Functional syncytium compris- ing osteocytes, osteoblasts, bone mar- row stromal cells, and endothelial cells. [Adapted from G. Marotti and repro- duced with the permission of the Editor of Journal of Clinical Investigation 104: 1363–1374, 1999 (219).

layer of unmineralized collagen matrix on top of which vacuolar Hϩ-ATPase) located in the ruffled border mem- there is a layer of flat and elongated cells. These cells are brane. The protein components of the matrix, mainly colla- called lining cells and are descendents of osteoblasts (13). gen, are degraded by matrix metalloproteinases, and ca- Conversion of osteoblasts to lining cells represents one of thepsins K, B, and L are secreted by the osteoclast into the the fates of osteoblasts that have completed their bone area of bone resorption (134). The degraded bone matrix forming function; another being entombment into the ma- components are endocytosed along the ruffled border within trix as osteocytes. Osteoclasts cannot attach to the unmin- the resorption lacunae and then transcytosed to the mem- eralized collagenous layer that covers the surface of nor- brane area opposite the bone, where they are released (135, mal bone. Therefore, other cells, perhaps the lining cells, 136). Another feature of osteoclasts is the presence of high secrete collagenase, which removes this matrix before os- amounts of the phosphohydrolase enzyme, tartrate-resistant teoclasts can attach to bone. It has been proposed that acid phosphatase (TRAPase). This feature is commonly used targeting of osteoclast precursors to a specific location on for the detection of osteoclasts in bone specimens (137). Mice bone depends on a “homing” signal given by lining cells; deficient in TRAPase exhibit a mild osteopetrotic phenotype and that lining cells are instructed to do so by osteocytes, (due to an intrinsic defect of osteoclastic resorptive activity) the only bone cells that can sense the need for remodeling and defective mineralization of the cartilage in developing at a specific time and place (133). bones (138).

D. Osteoclasts VII. Death of Bone Cells by Apoptosis Mature osteoclasts are usually large (50 to 100 ␮m diam- eter) multinucleated cells with abundant mitochondria, nu- The average lifespan of human osteoclasts is about 2 merous lysosomes, and free ribosomes. Their most remark- weeks, while the average lifespan of osteoblasts is 3 able morphological feature is the ruffled border, a complex months (Table 1). After osteoclasts have eroded to a par- system of finger-shaped projections of the membrane, the ticular distance, either from the central axis in cortical bone function of which is to mediate the resorption of the calcified or to a particular depth from the surface in cancellous bone matrix (17, 123). This structure is completely sur- bone, they die and are quickly removed by phagocytes rounded by another specialized area, called the clear zone. (139). The majority (65%) of the osteoblasts that originally The cytoplasm in the clear zone area has a uniform appear- assembled at the remodeling site also die (140). The re- ance and contains bundles of actin-like filaments. The clear maining are converted to lining cells that cover quiescent zone delineates the area of attachment of the osteoclast to the bone surfaces or are entombed within the mineralized bone surface and seals off a distinct area of the bone surface matrix as osteocytes (Fig. 3A). Both osteoclasts and osteo- that lies immediately underneath the osteoclast and which blasts die by apoptosis, or programmed cell death, a pro- eventually will be excavated. The ability of the clear zone to cess common to several regenerating tissues (141). As in seal off this area of bone surface allows the formation of a other tissues, bone cells undergoing apoptosis are recog- microenvironment suitable for the operation of the resorp- nized by condensation of chromatin, the degradation of tive apparatus. DNA into oligonucleosome-sized fragments, and the for- The mineral component of the matrix is dissolved in the mation of plasma and nuclear membrane blebs (Fig. 4). acidic environment of the resorption site, which is created by Eventually the cell breaks apart to form so-called apoptotic the action of an ATP-driven proton pump (the so-called bodies. Osteoblast apoptosis explains the fact that 50–70% April, 2000 BIRTH AND DEATH OF BONE CELLS 123

FIG. 3. Osteoblast apoptosis and its implications. A, The average life span of a matrix forming osteoblast (ϳ200 h in the mouse) is indicated by the continuous line. The process of apoptosis represents only a small fraction of this time period. The alternative two fates of osteoblasts are to become lining cells or osteocytes. The fraction of osteoblasts that undergo apoptosis in vivo (fApoptosis) can be estimated from a bone biopsy specimen. The duration of the apoptosis phase that can be observed in the specimen (tApop) depends on the sensitivity of the detection method. For example, in the case of the TUNEL technique (without CuSO4 enhancement), the TUNEL-labeled phase of apoptosis is estimated to be approximately 2 h. In a steady state, the fraction of cells at a particular stage is the same as the corresponding fraction of time spent in that stage. Assuming an apoptosis detection time of 2 h and a 200-h life span, a prevalence of TUNEL positive osteoblasts in the biopsy of 0.005 indicates that half of the osteoblasts die by apoptosis. B, A change in the timing and extent of osteoblast apoptosis (fApoptosis) from 50% to zero should increase the number of osteoblasts present at the site of bone formation and thereby the work output, i.e., the amount of bone formed by a given team of matrix-forming osteoblasts. It will also lead to an increase in the density of osteocyte apoptosis, as illustrated by the example shown. of the osteoblasts initially present at the remodeling site of The same growth factors and cytokines that stimulate os- human bone cannot be accounted for after enumeration of teoclast and osteoblast development can also influence their lining cells and osteocytes (142). Moreover, the frequency apoptosis. For example, TGF␤ promotes osteoclast apoptosis of osteoblast apoptosis in vivo is such that changes in its while it inhibits osteoblast apoptosis. IL-6 type cytokines timing and extent could have a significant impact in the have antiapoptotic effects on animal and human osteoblastic number of osteoblasts present at the site of bone formation cells (and at least in vitro they antagonize proapoptotic effects (130). Osteocytes are long-lived but not immortal cells; of glucocorticoids) as well as on osteoclasts and their pro- some die by apoptosis (132, 143, 144). Osteocyte apoptosis genitors (54, 140, 145, 146). could be of importance to the origination and/or progres- sion of the BMU. Indeed, osteocytes are the only cells in bone that can sense the need for remodeling at a specific VIII. Regulation of Bone Cell Proliferation and time and place. Moreover, osteocytes are in direct physical Activity contact with lining cells on the bone surface, and targeting of osteoclast precursors to a specific location on bone A large body of literature suggests that growth factors, depends on a “homing” signal given by lining cells (133). cytokines, hormones, and drugs regulate the proliferation of 124 MANOLAGAS Vol. 21, No. 2

FIG. 4. Two osteoblasts undergoing ap- optosis in a section of murine cancellous bone (TUNEL staining with toluidine blue counterstain, ϫ630). Apoptotic os- teoblasts (shown in brown) are adjacent to an intact osteoblast (shown in blue), on the surface of a trabecula occupying the right lower portion of the picture in which two intact (blue stained) osteo- cytes are also seen. Apoptotic osteo- blasts display nuclear condensation and fragmentation. [Photomicrograph provided by Robert S. Weinstein, M.D., University of Arkansas for Medical Sci- ences.]

committed cells or the biosynthetic and functional activity of esis, or osteoclastogenesis, as used in this article, implicitly the differentiated osteoblasts and osteoclasts. Hence, in ad- combine commitment and amplification. dition to cell number, alterations in the functional activity of The in vivo relevance of numerous reports of in vitro ob- individual cells, i.e., cell vigor, may contribute to changes in servations of changes in the biosynthetic activity of osteo- the rate of bone resorption and formation. However, because blastic cells, e.g., changes in the level of expression of osteo- of the inherent difficulty in demonstrating changes in indi- calcin or alkaline phosphatase in response to a given agent, vidual cell vigor in vivo, the vast majority of such literature is also a matter of conjecture. Most likely, given the nature and its conclusions rely heavily, if not exclusively, on in vitro of commonly used in vitro cell models which, by and large, experimentation. A detailed discussion of this work is be- represent preterminally differentiated cells, such observa- yond the scope of this review, and the reader is referred to tions might be more relevant to postcommitment differen- other articles (30, 147–151). Nonetheless, some general as- tiation events, than to altered activity of the fully differen- pects merit discussion here, as they are important for putting tiated cell. Moreover, even if some agents can alter the the significance of birth rate and apoptosis into a broader function of terminally differentiated cells in short-term cul- perspective. tures in vitro, heretofore there is no evidence that short-term In regenerating tissues, the initial commitment of a stem change in the rate of collagen production or bone matrix cell progeny is followed by amplification with several or digestion, for example, are ultimately translated into differ- many rounds of cell division. In most tissues, division of stem ences in the amounts of bone matrix formed or resorbed. cells is infrequent, and almost all of the divisions that pro- In the bone literature there is considerable ambiguity duce the final population of differentiated cells occur in the when using the term “activation.” It is important that one so-called transit compartment (152). This notion is obscured distinguishes between activation as a switch from an off state by the frequent practice of showing linear diagrams repre- to an on state, and activation as modulation of activity of an senting the transition from one cell type to another and already active cell. Morphological evidence summarized ignoring completely the amplification during the transition. elsewhere does not support the notion that completely in- In general, terminally differentiated cells do not divide, and active osteoclasts are waiting for a stimulus to make them an osteoblast, defined as a cell making bone matrix, and active (117). However, this evidence does not address the osteoclast, as a cell resorbing bone, are in this category. issue of whether there are variations in the rate of bone Therefore, the in vivo relevance of much of the in vitro evi- matrix dissolution by individual osteoclasts after they started dence on the regulation of osteoblastic or osteoclastic cell work. proliferation, using established cell lines or primary cultures Studies with the widely used bone slice pit bioassay have of isolated cells, must be largely confined to changes in this shown that several regulatory factors can cause a decrease or transit compartment. Irrespective of whether a given regu- increase in the resorptive ability of individual osteoclasts latory factor, be that a cytokine or a hormone, influences the (153–155). However, it is not clear to what extent these ob- initial commitment of a stem cell, or the subsequent ampli- servations reflect a change in cell vigor as opposed to a fication of its progeny, or both, the end result is a change in change in the precariously short lifespan of osteoclasts in the rate of production and therefore the number of cells vitro. In estrogen deficiency, individual osteoclasts are seem- available for the execution of the biological task. For the sake ingly more “active” as they dig deeper resorption cavities of simplicity, the terms birth, rate of birth, osteoblastogen- often leading to trabecular perforation, but it has been con- April, 2000 BIRTH AND DEATH OF BONE CELLS 125 vincingly argued that this is due to delayed apoptosis (133). IX. Pathogenesis Of Osteoporosis In Paget’s disease osteoclasts are certainly far more aggres- From the brief discussion of the principles of physiological sive than normal, perhaps as a result of their uniquely large bone regeneration and the role of osteoblasts and osteoclasts size and number of nuclei (156). Today, the most compelling in the process, it is obvious that the rate of supply of new evidence in support of the notion that the vigor of individual osteoblasts and osteoclasts and the timing of the death of osteoclasts may not always be maximal comes from in vitro these cells by apoptosis are critical determinants of the ini- as well as in vivo experiments with soluble RANK-ligand tiation of new BMUs and/or extension or shortening of the (157). Specifically, it has been found that RANK-ligand acts lifetime of existing ones. Recent advances in our understand- on mature rat osteoclasts in vitro to stimulate more frequent ing of the pathogenesis of the various forms of osteoporosis cycles of resorption and induce rearrangements of the cy- have confirmed this truism by revealing that over- or un- toskeleton. Moreover, intravenous administration of RANK- dersupply of these cells relative to the need for remodeling ligand to mice elevates the circulating concentration of are the fundamental problems in all these conditions (160) ionized calcium within 1 h. RANK-ligand has potent anti- (Table 2). apoptotic effects on cultured osteoclasts (Ref. 158 and Wil- liam Boyle, personal communication). Therefore, definitive conclusions regarding RANK-ligand’s ability to modulate A. Sex steroid deficiency osteoclast vigor will have to await dissection of the contri- bution of its effects on osteoclast survival in vivo. Similar to The mechanism of action of sex steroids on the skeleton is osteoclasts, the bone-forming ability of osteoblasts may not not fully understood. At menopause (or after castration in be always maximal, as suggested by the evidence that PTH men), the rate of bone remodeling increases precipitously. can rapidly enhance bone formation when administered by This fact may be explained by evidence, derived primarily subcutaneous injections to rats (159). from studies in mice, that loss of sex steroids up-regulates the To conclude this section, it is intuitive that the amount of formation of osteoclasts and osteoblasts in the marrow by bone resorbed or formed by a team of osteoclasts and os- up-regulating the production and action of cytokines that are teoblasts should be a function of the total cell number as well responsible for osteoclastogenesis and osteoblastogenesis as the vigor of individual cells. However, whereas cell num- (21, 161, 162). Indeed, both estrogen and androgen suppress ber can be quantified on bone sections from animals and the production of IL-6, as well as the expression of the two humans with conventional histomorphometric techniques, subunits of the IL-6 receptor, IL-6R␣ and gp130, in cells of the quantification of individual cell vigor cannot. This situation bone marrow stromal/osteoblastic lineage (40, 163). Sup- makes it difficult to judge at present whether cell vigor is or pression of IL-6 production by estrogen or selective estrogen is not a critical component in the pathogenesis of abnormal receptor modulators (SERMs), such as raloxifene, does not skeletal regeneration in common acquired metabolic bone require direct binding of the estrogen receptor to DNA. In- diseases such as postmenopausal, senile, or steroid-induced stead, it is due to protein-protein interaction between the osteoporosis. For this reason and space limitations, changes estrogen receptor and transcription factors such as NF-K␤ in bone cell vigor or other potential mechanisms of osteo- and C/EBP. This mechanism provides a model that best fits porosis resulting from changes in extraskeletal tissues, for current understanding of the molecular pharmacology of example altered calcium absorption or excretion, will not be estrogen and SERMs (164). Consistent with the suppressive discussed in the following section. This also reflects the au- effect of sex steroids on IL-6 and its receptor, several, albeit thor’s intention to focus on the dynamics of bone cell num- not all, studies have shown that the level of expression of ber, rather than a dismissal of other mechanisms. IL-6, as well as IL-6R␣ and gp130, is elevated in estrogen-

TABLE 2. Cellular changes and their culprits in the three most common causes of bone loss

Cellular changes Probable culprits Sex steroid deficiency 1Osteoblastogenesis Increased IL-6; TNF; IL-1RI/IL-RII 1Osteoclastogenesisa MCSF; decreased TGF␤; OPG 1Lifespan of osteoclasts Loss of pro- and antiapoptotic effects of sex steroids, respectively 2Lifespan of osteoblasts 2Lifespan of osteocytes Senescence 2Osteoblastogenesisb Increased PPAR␥2, PGJ2, noggin; Deceased IL-11, IGFs 2Osteoclastogenesis 1Adipogenesis 2Lifespan of osteocytes Glucocorticoid excess 2Osteoblastogenesis Decreased Cbfal and TGF␤ R1; and BMP-2 and IGF1 action 2Osteoclastogenesis 1Adipogenesis Increased PPAR␥2 1Lifespan of osteoclastsc 2Lifespan of osteoblasts Decreased Bcl-2/BAX ratio 2Lifespan of osteocytes a Oversupply of osteoclasts relative to the need for remodeling. b Undersupply of osteoblasts relative to the need for cavity repair. c Osteoclast numbers may transiently increase in the earlier stages of steroid therapy, without an increase in osteoclastogenesis, indicating increased lifespan. 126 MANOLAGAS Vol. 21, No. 2 deficient mice and rats as well as in humans, in the bone are more widely separated and less well connected. An marrow and in the peripheral blood (165–168). Furthermore, equivalent amount of cancellous bone distributed as widely neutralization of IL-6 with antibodies or knockout of the IL-6 separated, disconnected, thick trabeculae is biomechanically gene in mice prevents the expected cellular changes in the less competent than when arranged as more numerous, con- marrow and in trabecular bone sections and protects the mice nected, thin trabeculae. Concurrent loss of cortical bone oc- from bone loss after loss of sex steroids (51, 67). Consistent curs by enlargement and coalescence of subendocortical with its pathogenetic role in the bone loss caused by loss of spaces, a process due to deeper penetration of endocortical sex steroids, IL-6 seems to play a similar role in several other osteoclasts. conditions associated with increased bone resorption as ev- This deeper erosion can be now explained by evidence that idenced by increased local or systemic production of IL-6 and estrogen acts on mature osteoclasts to promote their apo- the IL-6 receptor in patients with multiple myeloma, Paget’s ptosis; consequently, loss of estrogen leads to prolongation disease, rheumatoid arthritis, Gorham-Stout or disappearing of the lifespan of osteoclasts (133). Specifically, estrogen pro- bone disease, hyperthyroidism, primary and secondary hy- motes osteoclast apoptosis in vitro and in vivo by 2- to 3-fold, perparathyroidism, as well as McCune Albright Syndrome an effect seemingly mediated by TGF␤ (139). In direct con- (66, 68, 169–174). trast to their proapoptotic effects on osteoclasts, estrogen (as In line with the fact that loss of sex steroids increases the well as androgen) exerts antiapoptotic effects on osteoblasts rate of bone remodeling, in addition to up-regulating oste- and osteocytes; consequently, loss of estrogen or androgen oclastogenesis, loss of sex steroids increases the number of leads to shorter lifespan of osteoblasts and osteocytes (184). osteoblast progenitors in the murine bone marrow. These Extension of the working life of the bone-resorbing cells and changes are temporally associated with increased bone for- simultaneous shortening of the working life of the bone- mation and parallel the increased osteoclastogenesis and forming cells, can explain the imbalance between bone re- bone resorption (175). As IL-6 type cytokines can stimulate sorption and formation that ensues after loss of sex steroids. osteoblast development and differentiation (54, 55, 146), in- Furthermore, the increase in osteocyte apoptosis could fur- creased sensitivity to IL-6 and other members of this cyto- ther weaken the skeleton by impairment of the osteocyte- kine’s family may account also for the increased osteoblast canalicular mechanosensory network. The increase in bone formation that follows the loss of gonadal function. In view remodeling that occurs with estrogen deficiency would of the fact that mesenchymal cell differentiation and oste- partly replace some of the nonviable osteocytes in cancellous oclastogenesis are tightly linked, stimulation of mesenchy- bone, but cortical apoptotic osteocytes might accumulate mal cell differentiation toward the osteoblastic lineage after because of their anatomic isolation from scavenger cells and sex steroid loss may be the first event that ensues after the the need for extensive degradation to small molecules to hormonal change, and increased osteoclastogenesis and dispose of the osteocytes through the narrow canaliculi. bone loss might be downstream consequences of this change Hence, the accumulation of apoptotic osteocytes caused by (106). loss of estrogen, or glucocorticoid excess (130), could increase In addition to IL-6, estrogen also suppress TNF and M-CSF bone fragility even before significant loss of bone mass, be- (176, 177), and estrogen loss may increase the sensitivity of cause of the impaired detection of microdamage and repair osteoclasts to IL-1 by increasing the ratio of the IL-1RI over of substandard bone. the IL-1 receptor antagonist (IL-RII) (178). As in the case of In conclusion, the increased rate of bone remodeling in IL-6, the effects of estrogen on TNF and M-CSF are mediated estrogen deficiency may be due to increased production of via protein-protein interactions between the estrogen recep- both osteoclasts and osteoblasts, and the imbalance between tor and other transcription factors. In agreement with the bone resorption and formation is due to an extension of the evidence that IL-1 and TNF play a role in the bone loss caused working lifespan of the osteoclast and shortening of the by loss of estrogen, administration of IL-1RA and/or a TNF working lifespan of the osteoblast. Moreover, a delay of soluble receptor ameliorates the bone loss caused by ovari- osteoclast apoptosis seems responsible for the deeper re- ectomy in rats and mice (179–181). Because of the interde- sorption cavities and thereby the trabecular perforation as- pendent nature of the production of IL-1, IL-6, and TNF, a sociated with estrogen deficiency. significant increase in one of them may amplify, in a cascade Clinical observations of decreased bone mass in a male fashion, the effect of the others (161). Interestingly, recent in with mutant estrogen receptor (185), and increased bone vitro studies with human osteoblastic cells indicate that OPG mass after treatment with estrogen in two males with P-450 production is stimulated by estrogen, suggesting that this aromatase deficiency (186, 187), have raised the possibility cytokine may also play an important role in the antioste- that estrogen derived by peripheral aromatization of andro- oclastogenic (antiresorptive) action of estrogen on bone (182). gens is critical for the maintenance of bone mass in men as Increased remodeling, resulting from up-regulation of os- well as in women (188). However, in all three cases, the teoblastogenesis and osteoclastogenesis, alone can cause a decreased bone mass in young males with estrogen defi- transient acceleration of bone mineral loss because bone re- ciency in the face of androgen sufficiency could be due to sorption is faster than bone formation, and new bone is less failure in achieving peak bone mass from defects occurring dense than older bone. However, in addition to increased during development or growth, not to loss of bone mass, as bone remodeling, loss of sex steroids leads to a qualitative it is the case with common forms of osteoporosis. In addition, abnormality: osteoclasts erode deeper than normal cavities individuals with complete androgen insensitivity, due to (133, 183). In this manner, sex steroid deficiency leads to the mutations in the androgen receptor gene on the X chromo- removal of some cancellous elements entirely; the remainder some and increased testosterone and estrogen production, April, 2000 BIRTH AND DEATH OF BONE CELLS 127 have decreased bone mass, in spite of the elevated estrogen HODE, are endogenous ligands and activators of PPAR␥ levels (189). Moreover, androgens, including nonaromatiz- (201), and PPAR␥ promotes monocyte/macrophage differ- able ones, have identical effects to those of estrogen on the entiation and uptake of oxidized LDL (202). Taken together biosynthetic activity and the birth as well as the death of bone with these findings, the evidence that activated PPAR␥2 cells in vitro and in vivo, at least in rodents (67, 106, 190). It promotes adipocyte differentiation at the expense of osteo- is therefore more likely that both estrogen and androgen are blastogenesis in the bone marrow by suppressing Cbfa1 (94) important for the maintenance of bone mass in the adult male suggests a mechanistic link among dietary fat/lipoproteins, skeleton. bone marrow stromal cell differentiation, osteoporosis, and atherogenesis. In support of the existence of such a link, ␥ B. Senescence activation of PPAR by thiazolidinediones or oxidatively modified LDL inhibits osteoblastogenesis of bone marrow- The amount of bone formed during each remodeling cycle derived stromal cells in vitro (94, 203, 204). Moreover, high decreases with age in both sexes. This is indicated by a fatty acid content in rabbit serum or high fat diet of mice for consistent histological feature of the osteopenia that occurs 4 months decreases osteogenic cell differentiation in ex vivo during aging, namely a decrease in wall thickness, especially bone marrow cell cultures (86, 204). These new advances may in trabecular bone (191–193). Wall thickness is a measure of explain clinical observations that atherosclerosis and osteo- the amount of bone formed in a remodeling packet of cells porosis coexist (205, 206). and is determined by the number or activity of osteoblasts at Quantitative trait loci (QTLs) analysis of osteopenia-asso- the remodeling site. ciated loci using closely related mouse strains have mapped Studies measuring bone turnover by histomorphometry five loci to regions of chromosomes 2, 7, 11, and 16 (207). (194), or indirectly by circulating markers (195–197), have Association of these same loci with bone mineral density has suggested that in aging women, even in extreme old age, been reproduced in crosses of different recombinant-inbred bone turnover is most likely increased by secondary hyper- mouse strains (208, 209). Such recurrent appearance of QTL, parathyroidism or by the continuing effect of estrogen de- especially in crosses involving distantly related strains, im- ficiency. Increased turnover and reduced wall thickness are plies that polymorphism at these loci may be favored by not inconsistent, as the former is the result of increased evolution and might underlie variation in peak activation frequency, and the decreased wall thickness—an among humans. Intriguingly, of the more than 12 genes index of decreased bone formation by osteoblasts—in senes- affecting bone homeostasis that were localized near these cence is local and relative to the demand created by resorp- QTLs, 2 are prostaglandin synthases, a third is the BMP-2/4 tion. antagonist noggin, a fourth is the proapoptotic protein bax, Changes in the birth of bone cells in the bone marrow and the fifth is IL-11. Hence, the transcription factor PPAR␥2 provide a mechanistic explanation for the contribution of and its ligand, PGJ2, noggin, and IL-11 are potentially re- senescence to bone loss, independently from sex steroid de- sponsible for the decreased osteoblastogenesis with advanc- ficiency. Specifically, using SAMP6 mice, a murine model of ing age. This contention is supported by the evidence that the age-related osteopenia (but sufficient in sex steroids and with reciprocal relationship between decreased osteoblastogen- intact reproductive function), a tight association among re- esis and increased adipogenesis in the SAMP6 mouse may be duced number of osteoblast progenitors, decreased bone for- explained by a change in the expression of PPAR␥ or its mation, and decreased bone mass has been established (105). ligands in early mesenchymal progenitors; that BMP-2/4, in Decreased osteoblastogenesis with advancing age has been balance with noggin, may determine the tonic baseline con- confirmed in the human bone marrow (198, 199). Impor- trol of the rate of osteoblastogenesis; and that IL-11 is a potent tantly, the decrease in osteoblastogenesis is accompanied by inhibitor of adipogenesis, which stimulates osteoblast dif- increased adipogenesis and myelopoiesis, as well as de- ferentiation and the expression of which is reduced in creased osteoclastogenesis, the latter most likely caused by a SAMP6 mice. In addition to these factors, growth factors such reduction in the stromal/osteoblastic cells that support os- as IGFs have also been implicated in the bone loss associated teoclast formation (105, 200). This suggests that in aging there with senescence (210, 211). Irrespective of the identity of the must be changes in the expression of genes that favor the precise mediator, the reciprocal change between adipogen- differentiation of multipotent mesenchymal stem cells to- esis and osteoblastogenesis can explain the association of ward adipocytes at the expense of osteoblasts. The evidence decreased relative bone formation and the resulting osteope- that PPAR␥2 induces the terminal differentiation of marrow nia with the increased adiposity of the marrow seen with cells with both osteoblastic and adipocytic characteristics to advancing age in animals and humans (105, 142, 212–215). adipocytes, and simultaneously suppresses Cbfa1 expres- sion and terminal differentiation to osteoblasts (94), raises the C. Glucocorticoid excess possibility that increased expression of PPAR␥2 or its li- gands, e.g., PGJ2, may be some of the culprits responsible for The cardinal histological features of glucocorticoid-induced the reciprocal change between adipogenesis and osteoblas- osteoporosis are decreased bone formation rate, decreased wall togenesis with advancing age (Table 2). thickness of trabeculae (a strong indication of decreased work Uptake of oxidized low-density lipoproteins (LDL) play an output by osteoblasts), and in situ death of portions of bone. important role in foam cell formation and the pathogenesis Increased bone resorption, decreased osteoblast proliferation of atherosclerosis. Two of the major components of oxidized and biosynthetic activity, and sex-steroid deficiency, as well as LDL, 9-hydroxy-9,11-octadecadienoic acid (HODE) and 13- hyperparathyroidism resulting from decreased intestinal cal- 128 MANOLAGAS Vol. 21, No. 2 cium absorption and hypercalciuria due to defective vitamin D from patients with glucocorticoid-induced osteoporosis ex- metabolism, have all been proposed as mechanisms for the loss hibit abundant apoptotic osteocytes adjacent to the subchon- of bone that ensues with glucocorticoid excess (216). dral fracture crescent (221). Glucocorticoid-induced osteo- The decreased bone formation and osteonecrosis can now cyte apoptosis, a cumulative and unrepairable defect, could be explained by evidence that glucocorticoid excess has a uniquely disrupt the proposed mechanosensory role of the suppressive effect on osteoblastogenesis in the bone marrow osteocyte network and thus promote collapse of the femoral and also promotes the apoptosis of osteoblasts and osteo- head. cytes (118). Indeed, mice receiving glucocorticoids for 4 At this time, the mediators of the cellular changes caused weeks, a period equivalent to ϳ3–4 yr in humans, exhibit by glucocorticoid excess are only a matter of conjecture. decreased bone mineral density associated with a decrease in Nonetheless, glucocorticoids directly suppress BMP-2 and the number of osteoblast, as well as osteoclast, progenitors in Cbfa1-2—two critical factors for osteoblastogenesis—and the bone marrow and a dramatic reduction in cancellous may also decrease the production of IGFs while they stim- bone area and in trabecular width compared with placebo ulate the transcriptional activity of PPAR␥2 (222–225) (Table controls. These changes are associated with a significant re- 2). duction in osteoid area and a decrease in the rates of mineral apposition and bone formation. More strikingly, glucocor- ticoid administration to mice causes a 3-fold increase in the X. Pharmacotherapeutic Implications of Osteoblast prevalence of osteoblast apoptosis in vertebrae and induced and Osteocyte Apoptosis apoptosis in 28% of the osteocytes in metaphyseal cortical Estrogen replacement therapy (ERT), various bisphospho- bone. Nevertheless, even though there is a significant cor- nates (e.g., alendronate), the SERM raloxifene, calcitonin, and relation between the severity of the bone loss and the extent sodium fluoride, as well as calcium and vitamin D, are ap- of reduction in bone formation, some of the bone loss may proved modalities for the prevention and treatment of bone be due to an early increase in bone resorption as evidenced loss, irrespective of its cause. Decreased osteoclast progenitor by an early increase in osteoclast perimeter of vertebral can- development and/or decreased osteoclast recruitment and cellous bone after 7 days of steroid treatment. In vivo studies promotion of apoptosis of mature osteoclasts leading to a with mice from this author’s group show that at this early slowing of the rate of bone remodeling are thought to be the time point (7 days after glucocorticoid administration) os- main mechanisms of the so-called “antiresorptive” agents teoclastogenesis in ex vivo bone marrow cultures is decreased estrogen, bisphosphonates, SERMs, and calcitonin. Sodium by half, while the number of osteoclasts in bone sections fluoride has anabolic properties, but its therapeutic range is doubles (Robert Weinstein, personal communication), sug- very narrow. Calcium and vitamin D are rarely sufficient on gesting that an early effect of glucocorticoid excess might be their own, but they are considered a very useful supplemen- increased osteoclast survival. In vitro studies by others, on the tation in any regimen for osteoporosis. other hand, have shown that glucocorticoids inhibit OPG and concurrently stimulate the expression of RANK-ligand in A. Intermittent PTH administration human osteoblastic, primary, and immortalized bone mar- row stromal cells (217). Taken together, these lines of evi- The ideal therapy for osteoporosis, especially in elderly dence suggest that the initial rapid phase of bone loss with patients who already have advanced bone loss, would be an glucocorticoid treatment could be due to an extension of the anabolic agent that will increase bone mass by rebuilding lifespan of preexisting osteoclasts, mediated by RANK- bone. It is well established that daily injections of low doses ligand (117). of PTH—an agent better known for its role in calcium ho- The same histomorphometric changes that have been meostasis—increases bone mass in animals and humans found in mice after a 4-week treatment with steroids have (226–231) as does the PTH-related protein (PTHrP), the only been confirmed in biopsies from patients receiving long-term other known ligand of the PTH receptor (232). Indeed, al- glucocorticoid therapy. Moreover, as in mice, an increase in though constant, high levels of PTH cause increased bone osteoblast and osteocyte apoptosis is found in human biop- resorption and fibrosa cystica, low and intermittent sies. Compared with osteoblast apoptosis, apoptotic osteo- doses of PTH, too small to affect serum calcium concentra- cytes are far more prevalent, at least in metaphyseal cortices, tions, promote bone formation and increase bone mineral probably because of the anatomical isolation of osteocytes density at the lumbar spine and hip. This so-called anabolic from scavenger cells. Consistent with these findings, glu- effect can be now explained by evidence that PTH increases cocorticoids promote osteoblast and osteocyte apoptosis in the life span of mature osteoblasts in vivo by reducing the vitro (218, 219). Decreased production of osteoclasts can ex- prevalence of their apoptosis from 1.7–2.2% to as little as plain the reduction in bone turnover with chronic glucocor- 0.1–0.4% rather than by affecting the generation of new os- ticoid excess, whereas decreased production and apoptosis teoblasts (218). The antiapoptotic effect of PTH in mice was of osteoblasts can explain the decline in bone formation and sufficient to account for the increase in bone mass and was trabecular width. Accumulation of apoptotic osteocytes may confirmed in vitro using rodent and human osteoblasts and also explain the so-called “osteonecrosis,” also known as osteocytes. Like PTH, PGE inhibits periosteal cell apoptosis aseptic or , another manifestation of ste- via cAMP-dependent stimulation of sphingosine kinase roid-induced osteoporosis that causes collapse of the femoral (233). Interestingly, whereas PTH inhibits apoptosis in cells head in as many as 25% of patients (220). This contention is overexpressing Gs, an activator of adenylate cyclase, PTH supported by evidence that whole femoral heads obtained stimulates apoptosis via G protein-coupled receptors in cells April, 2000 BIRTH AND DEATH OF BONE CELLS 129 overexpressing Gq (an activator of JNK and calcium signal- also have a positive effect on bone formation. An explanation ing), suggesting that the antiapoptotic effects of PTH are for this evidence is now provided by studies demonstrating mediated by signals transduced through the Gs pathway that bisphosphonates such as etidronate, alendronate, pam- (234). idronate, olpadronate, or amino-olpadronate (IG9402, a Osteocytes in the newly made lamellar cancellous bone bisphosphonate that lacks antiresorptive activity), as well as in the mice receiving daily PTH injections were closer calcitonin have antiapoptotic effects on osteoblasts and os- together and more numerous than those found in the teocytes (219). These effects are associated with a rapid in- animals receiving vehicle alone (218). The closely spaced, crease in the phosphorylated fraction of extracellular regu- more numerous osteocytes are the predictable conse- lated signal kinases (ERKs) and are blocked by specific quence of protecting osteoblasts from apoptosis (Fig. 3B). inhibitors of ERK activation. In agreement with the in vitro The antiapoptotic effect of PTH on osteoblasts as well as results, alendronate abolishes the increase in the prevalence osteocytes has been confirmed in vitro using primary bone of vertebral, cancellous bone osteocyte and osteoblasts ap- cell cultures and established cell lines. Elucidation of this optosis induced by administration of prednisolone in mice. mechanism provides for the first time proof that inhibition These findings raise for the first time the possibility that of osteoblast apoptosis may represent a novel therapeutic increased survival of osteoblasts and osteocytes may both strategy for augmenting bone mass. Be that as it may, contribute to the efficacy of bisphosphonates and calcitonin several alternative mechanisms, including activation of in the management of disease states due to loss of bone. lining cells, have been proposed, and they may also con- If both “antiresorptive” and “anabolic” agents [e.g., in- tribute to the anabolic effect of PTH (235–237). Nonethe- termittent PTH] prevent osteoblast and osteocyte apopto- less, lining cells cover at least 3 times more surface than sis, why is increased formation so much more apparent in osteoblasts. Therefore, conversion of lining cells to bone- the case of the “anabolic” agents? The discussion of phys- forming osteoblasts alone would be insufficient to cover iological bone regeneration in the beginning of this review the increased cancellous bone area observed in rats and to article provides an answer. Bone formation occurs only on account for the expanded bone perimeter and the in- sites of previous osteoclastic bone resorption, i.e., on sites creased osteocyte number and density observed with PTH undergoing remodeling. Each remodeling cycle is a trans- treatment in mice (218). action that, once consummated, is irrevocable. Therefore, Daily subcutaneous injections of PTH are safe and effec- agents with antiapoptotic properties that do not have an- tive in the treatment of patients with corticosteroid-induced tiresorptive/antiremodeling properties, i.e., they do not osteoporosis (230). The elucidation of the importance of os- decrease the number of remodeling units, are expected to teoblast and osteocyte apoptosis in the mechanism of glu- rebuild more bone and therefore increase the overall bone cocorticoid-induced osteoporosis, and the elucidation of the mass, because of the greater number of profitable trans- importance of preventing apoptosis in the anabolic effects of actions. Hence, by decreasing the prevalence of osteoblast PTH on bone, readily explain how PTH can be such an apoptosis, agents with pure antiapoptotic properties, such effective therapy in this condition. Hence, PTH and perhaps as intermittent PTH, can expand the pool of mature os- future PTH mimetics represent, for the first time, pathophys- teoblasts at sites of new bone formation and allow these iology-based, i.e., rational as opposed to empirical, pharma- cells more time to make bone, to a much greater degree cotherapies for osteopenias, in particular, those in which than the antiresorptive agents that also slow remodeling. osteoblast progenitor formation is suppressed. In any case, However, in the case of either class of agents, upholding future studies to assess the antifracture efficacy of these the osteocyte-canalicular network by preventing osteocyte compounds will be needed before their effectiveness for the apoptosis, should contribute to antifracture efficacy, over management of osteoporosis can be established. and above that resulting from their effects on bone mass (Fig. 5). Therefore, the distinction between antiresorptive B. Bisphosphonates and calcitonin and anabolic agents may be more apparent than real when it comes to antifracture efficacy. Bisphosphonates, stable analogs of pyrophosphate, and calcitonin are potent inhibitors of bone resorption and ef- C. Novel pharmacotherapeutic strategies fective therapies for the management of osteoporosis and other diseases characterized by bone loss (238, 239). The main Based on the understanding of the role of growth factors mechanism of the antiresorptive actions of these agents is on osteoblast development, proliferation, and differentia- decreased development of osteoclast progenitors, decreased tion, several of them (e.g., GH, the insulin-like growth factors osteoclast recruitment, and promotion of apoptosis of mature I and II, TGF␤, BMPs, and FGF) have been advocated as osteoclasts leading to a slowing rate of bone remodeling (133, potential future therapeutic agents for the management of 240–242). Nonetheless, the antifracture efficacy of these bone loss (247, 248). However, with the exception of BMPs, agents is disproportional to their effect on bone mass (243), which may be of value in local augmentation of bone mass suggesting an additional effect on bone strength unrelated to and acceleration of fracture healing, none of them has been effects on bone resorption. Moreover, long-term treatment of shown to be efficacious (let alone safe or convenient and human and nonhuman primates with bisphosphonates has practical) for the management of common metabolic bone been shown to increase wall thickness, an index of increased disorders such as osteoporosis. The recent elucidation of the osteoblast numbers or activity (244–246), raising the possi- mechanism of the anabolic effects of PTH, and specifically bility that they may not only inhibit bone resorption, but may the demonstration of increased work output of a cell pop- 130 MANOLAGAS Vol. 21, No. 2

FIG. 5. Hypothetical model of the im- plications of the effects of antiresorp- tive (i.e., antiremodeling) vs. non-anti- resorptive agents on prolonging the life span of mature osteoblasts and osteo- cytes. For explanation, please see text.

ulation by suppressing apoptosis to augment tissue mass, a potent neuroprotective compound (264–266), and does ex- points to an entirely new avenue for future drug discovery. hibit potent antiapoptotic effects on osteoblasts and osteo- Indeed, in addition to PTH and PTH mimetics, which by cytes in vitro. In support of the hypothesis that ANGELS can virtue of their peptidic nature carry the inconvenience of be used as a novel, advantageous mode of therapy for the daily injections, one can for the first time envisage nonpep- augmentation of bone mass and/or fracture prevention in tide inhibitors of proapoptotic pathways in osteoblasts and diseases characterized by low bone mass and increased fra- osteocytes as therapeutic agents for osteopenias and espe- gility, preliminary evidence indicates that estratriene-3-ol cially those in which osteoblast progenitors are low, such as increases BMD and bone strength in both estrogen-replete age-related and glucocorticoid-induced osteoporosis. and estrogen-deficient mice (261). In view of this preclinical As discussed in Section VIII in this review, estrogen and finding and the evidence that androgen (190), as well as androgen deficiency increase osteoblast and osteocyte apo- estrogen, have antiapoptotic effects on osteoblasts and os- ptosis in humans, rats, and mice; and these changes have teocytes, one is encouraged to think that estrogenic, andro- been shown to be reversed by replacement therapy, at least genic, or even nonsteroidal compounds that can activate in mice (132, 144, 184, 190). In full agreement with these in antiapoptotic, but not antiremodeling, signals on osteoblasts vivo observations, 17␤-estradiol inhibits osteoblast and os- and osteocytes, are candidates for future osteoporosis treat- teocyte apoptosis in vitro. The antiapoptotic effect of 17␤- ments that, unlike existing ones that prevent or retard bone estradiol on osteoblasts and osteocytes require the presence loss, may augment bone mass. of the estrogen receptor-␣ or -␤ (249). Nonetheless, unlike the classical mechanism of estrogen receptor action that involves direct or indirect interaction with the transcriptional appa- XI. Summary and Conclusions ratus, the estrogen receptor-dependent antiapoptotic effect of 17␤-estradiol is due to rapid (within 5 min) phosphory- In 1995, it was proposed that “changes in the numbers of lation of ERKs (250). Moreover, the antiapoptotic effect of bone cells, rather than changes in the activity of individual 17␤-estradiol can be reproduced by 17␣-estradiol, a com- cells, form the pathogenetic basis of osteoporosis”; and that pound thought of as an inactive analog of 17␤-estradiol, as “excessive osteoclastogenesis and inadequate osteoblasto- well as a membrane-impermeable conjugate of 17␤-estradiol genesis are responsible for the mismatch between the for- ␤ with BSA (17 E2-BSA). Numerous effects of estrogen have mation and resorption of bone in postmenopausal and age- been observed over the last few years in a variety of cell related osteopenia” (21). Since then, this paradigm shift of types, including osteoblasts, the rapidity of which makes a thinking has led to important new discoveries that, along genomic mechanism of action unlikely (251–256). Many of with several other independent breakthroughs, refine the these rapid actions have been attributed to the ability of concept of “cell number” and broaden its relevance to the estrogen to act at the cell membrane on a membrane-asso- physiology and pathophysiology of bone at large. Moreover, ciated estrogen receptor (257–260). The antiapoptotic effects these discoveries provide a new landscape for critical re- of estrogen on osteoblasts and osteocytes fall into this cate- evaluation of our current thinking about therapeutic strate- gory of “nongenomic” actions. Based on this, the term “ac- gies for bone diseases. Indeed, it is now clear that bone cells tivators of non-genomic estrogen-like signaling” (ANGELS), must be continually replaced, and the number present de- has been coined for compounds that mimic the nongenomic pends not only on their birth rate, which reflects the fre- effects of estrogen, but have reduced classical estrogenic quency of cell division of the appropriate precursor cell, but actions (261). A paradigm of such agents is the synthetic also on the life span, which most likely reflects the timing of compound estratriene-3-ol, which has decreased transcrip- death by apoptosis. The process of replacement of osteoblasts tional activity as compared with 17␤-estradiol (262, 263), is and osteoclasts is tightly coordinated and orchestrated at the April, 2000 BIRTH AND DEATH OF BONE CELLS 131 early progenitor level. Changes in the birth rate and/or ap- 15. Owen M 1985 Lineage of osteogenic cells and their relationship to optosis of bone cells may account for previously unexplained the stromal system. In: Peck WA (ed) Bone and Mineral Research. bone diseases, such as the osteoporosis caused by sex steroid Elsevier, Amsterdam, vol 3:1–25 16. Triffitt JT 1996 The stem cell of the osteoblast. In: Bilezikian JP, deficiency, old age, and glucocorticoid excess. Moreover, Raisz LG, Rodan GA (eds) Principles of Bone Biology. Academic attenuation of the rate of apoptosis of osteoblastic cells may Press, San Diego, CA, pp 39–50 be a key mechanism for the effects of anabolic agents, such 17. Roodman GD 1996 Advances in bone biology: the osteoclast. En- as PTH. Proof of the principle that the work performed by a docr Rev 17:308–332 18. Suda T, Takahashi N, Martin TJ 1992 Modulation of osteoclast cell population can be increased by suppression of apoptosis differentiation. Endocr Rev 13:66–80 provides clues for the development of novel pharmacother- 19. Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, apeutic strategies for pathological conditions such as osteo- Mosca JD, Moorman MA, Simonetti DW, Craig S, Marshak DR porosis in which tissue mass diminution has compromised 1999 Multilineage potential of adult human mesenchymal stem functional integrity. Nevertheless, changes in cell birth and cells. Science 284:143–147 20. Schor AM, Canfield AE, Sutton AB, Arciniegas E, Allen TD 1995 death, as well as other mechanisms including changes in Pericyte differentiation. Clin Orthop 313:81–91 bone cell activity, need to be investigated in humans more 21. Manolagas SC, Jilka RL 1995 Bone marrow, cytokines, and bone extensively before definitive conclusions on the pathogenesis remodeling—emerging insights into the pathophysiology of os- of the various causes of bone loss and the development of teoporosis. N Engl J Med 332:305–311 22. Rosen V, Cox K, Hattersley G 1996 Bone morphogenetic proteins. osteoporosis can be reached. In: Bilezikian JP, Raisz LG, Rodan GA (eds) Principles of Bone Biology. Academic Press, San Diego, CA, pp 661–671 23. Abe E, Yamamoto M, Taguchi Y, Lecka-Czernik B, Economides Acknowledgments AN, Stahl N, Jilka RL, Manolagas SC 2000 Essential requirement of BMPs 2/4 for both osteoblast and osteoclast formation in bone The author is grateful to A. Michael Parfitt, Robert L. Jilka, Robert S. marrow cultures from adult mice: antagonism by noggin. J Bone Weinstein, Teresita Bellido, Charles A. O’Brien, Robert S. Reis, Paula Miner Res 15:663–673 Roberson, Beata Lecka-Czernik, Etsuko Abe, Donald L. Bodenner, and 24. Ducy P, Zhang R, Geoffroy V, Ridall AL, Karsenty G 1997 Osf2/ Stavroula Kousteni for many insightful discussions, sharing of ideas, Cbfa1: a transcriptional activator of osteoblast differentiation. Cell and contribution of research findings during the preparation of the 89:747–754 manuscript; and to Tonya Smith for secretarial assistance. 25. Gao YH, Shinki T, Yuasa T, Kataoka-Enomoto H, Komori T, Suda T, Yamaguchi A 1998 Potential role of Cbfa1, an essential tran- scriptional factor for osteoblast differentiation, in osteoclastogen- References esis: regulation of mRNA expression of osteoclast differentiation fac- tor (ODF). Biochem Biophys Res Commun 252:697–702 1. Riggs BL, Melton III LJ 1986 Involutional osteoporosis. N Engl 26. Komori T, Yagi H, Nomura S, Yamaguchi A, Sasaki K, Deguchi J Med 314:1676–1686 K, Shimizu Y, Bronson RT, Gao Y-H, Inada M, Sato M, Okamoto 2. Lindsay R 1988 Sex steroids in the pathogenesis and prevention of R, Kitamura Y, Yoshiki S, Kishimoto T 1997 Targeted disruption osteoporosis. In: Riggs BL (ed) Osteoporosis: Etiology, Diagnosis of Cbfa1 results in a complete lack of bone formation owing to and Management. Raven Press, New York, pp 333–358 maturational arrest of osteoblasts. Cell 89:755–764 3. Melton III LJ, Atkinson EJ, O’Connor MK, O’Fallon WM, Riggs 27. Ryoo HM, Hoffmann HM, Beumer T, Frenkel B, Towler DA, BL 1998 Bone density and fracture risk in men. J Bone Miner Res Stein GS, Stein JL, Van Wijnen AJ, Lian JB 1997 Stage-specific 13:1915–1923 expression of Dlx-5 during osteoblast differentiation: involvement 4. Orwoll ES, Klein RF 1995 Osteoporosis in men. Endocr Rev 16: in regulation of osteocalcin gene expression. Mol Endocrinol 11: 87–116 1681–1694 5. Reid IR 1998 Glucocorticoid effects on bone. J Clin Endocrinol 28. Newberry EP, Latifi T, Towler DA 1998 Reciprocal regulation of Metab 83:1860–1862 osteocalcin transcription by the homeodomain proteins Msx2 and 6. Cummings SR, Nevitt MC, Browner WS, Stone K, Fox KM, Dlx5. Biochemistry 37:16360–16368 Ensrud KE, Cauley J, Black D, Vogt TM 1995 Risk factors for hip 29. Miyama K, Yamada G, Yamamoto TS, Takagi C, Miyado K, Sakai fracture in white women. Study of Osteoporotic Fractures Research M, Ueno N, Shibuya H 1999 A BMP-inducible gene, dlx5, regulates Group. N Engl J Med 332:767–773 osteoblast differentiation and mesoderm induction. Dev Biol 208: 7. Riggs BL, Melton III LJ 1983 Evidence for two distinct syndromes 123–133 of involutional osteoporosis. Am J Med 75:899–901 30. Canalis E 1996 Skeletal Growth Factors. In: Marcus R, Feldman D, 8. Parfitt AM 1992 The two-stage concept of bone loss revisited. Kelsey J (eds) Osteoporosis. Academic Press, San Diego, CA, pp Triangle 31:99–110 261–279 9. Silverberg SJ, Fitzpatrick LA, Bilezikian JP 1995 Hyperparathy- 31. Bonewald LF, Dallas SL 1994 Role of active and latent transform- roidism. In: Becker KL (ed) Principles and Practice of Endocrinol- ing growth factor ␤ in bone formation. J Cell Biochem 55:350–357 ogy and Metabolism. JB Lippincott, Philadelphia, pp 512–519 32. Piccolo S, Sasai Y, Lu B, De Robertis EM 1996 Dorsoventral pat- 10. Parfitt AM 1996 Skeletal heterogeneity and the purposes of bone terning in Xenopus: inhibition of ventral signals by direct binding remodeling: Implications for the understanding of osteoporosis. In: of chordin to BMP-4. Cell 86:589–598 Marcus R, Feldman D, Kelsey J (eds) Osteoporosis. Academic Press, 33. Holley SA, Neul JL, Attisano L, Wrana JL, Sasai Y, O’Connor MB, San Diego, CA, pp 315–329 De Robertis EM, Ferguson EL 1996 The Xenopus dorsalizing factor 11. Frost HM 1973 Bone Remodeling and Its Relationship to Metabolic noggin ventralizes Drosophila embryos by preventing DPP from Bone Disease. Charles C. Thomas, Springfield, MA activating its receptor. Cell 86:607–617 12. Parfitt AM, Rauch F, Travers R, Glorieux F 1999 A new model of 34. Valenzuela DM, Economides AN, Rojas E, Lamb TM, Nunez L, cancellous bone growth. J Bone Miner Res S206 (Abstract) Jones P, Ip NY, Espinosa III R, Brannan CI, Gilbert DJ, Copeland 13. Parfitt AM 1994 Osteonal and hemi-osteonal remodeling: the spa- NG, Jenkins NA, Beau MML, Harland RM, Yancopoulos GD 1995 tial and temporal framework for signal traffic in adult human bone. Identification of mammalian noggin and its expression in the adult J Cell Biochem 55:273–286 nervous system. J Neurosci 15:6077–6084 14. Friedenstein AJ, Chailakhjan RK, Latsinik NV, Panasyuk AF, 35. Bouwmeester T, Kim S, Sasai Y, Lu B, De Robertis EM 1996 Keiliss-Borok IV 1974 Stromal cells responsible for transferring the Cerberus is a head-inducing secreted factor expressed in the an- microenvironment of the hemopoietic tissues. Cloning in vitro and terior endoderm of Spemann’s organizer. Nature 382:595–601 retransplantation in vivo. Transplantation 17:331–340 36. Zimmerman LB, De Jesu´ s-Escobar JM, Harland RM 1996 The 132 MANOLAGAS Vol. 21, No. 2

Spemann organizer signal noggin binds and inactivates bone mor- 54. Bellido T, Borba VZ, Roberson P, Manolagas SC 1997 Activation phogenetic protein 4. Cell 86:599–606 of the Janus kinase/STAT (signal transducer and activator of tran- 37. Re’em-Kalma Y, Lamb T, Frank D 1995 Competition between scription) signal transduction pathway by interleukin-6-type cy- noggin and bone morphogenic protein 4 activities may regulate tokines promotes osteoblast differentiation. Endocrinology dorsalization during Xenopus development. Proc Natl Acad Sci 138:3666–3676 USA 92:12141–12145 55. Taguchi Y, Yamamoto M, Yamate T, Lin SC, Mocharla H, 38. Manolagas SC, Jilka RL, Bellido T, O’Brien CA, Parfitt AM 1996 DeTogni P, Nakayama N, Boyce BF, Abe E, Manolagas SC 1998 Interleukin-6-type cytokines and their receptors. In: Bilezikian JP, Interleukin-6-type cytokines stimulate mesenchymal progenitor Raisz LG, Rodan GA (eds) Principles of Bone Biology. Academic differentiation toward the osteoblastic lineage. Proc Assoc Am Press, San Diego, CA, pp 701–713 Physicians 110:559–574 39. Udagawa N, Horwood NJ, Elliott J, Mackay A, Owens J, Okamura 56. Malik N, Haugen HS, Modrell B, Shoyab M, Clegg CH 1995 H, Kurimoto M, Chambers TJ, Martin TJ, Gillespie MT 1997 Developmental abnormalities in mice transgenic for bovine on- Interleukin-18 (interferon-␥-inducing factor) is produced by osteo- costatin M. Mol Cell Biol 15:2349–2358 blasts and acts via granulocyte/macrophage colony-stimulating 57. Ware CB, Horowitz MC, Renshaw BR, Hunt JS, Liggitt D, Koblar factor and not via interferon-␥ to inhibit osteoclast formation. J Exp SA, Gliniak BC, McKenna HJ, Papayannopoulou T, Thoma B Med 185:1005–1012 1995 Targeted disruption of the low-affinity leukemia inhibitory 40. Manolagas SC 1998 The role of IL-6 type cytokines and their factor receptor gene causes placental, skeletal, neural and metabolic receptors in bone. Ann NY Acad Sci 840:194–204 defects and results in perinatal death. Development 121:1283–1299 41. Girasole G, Jilka RL, Passeri G, Boswell S, Boder G, Williams DC, 58. Bonewald L 1996 Transforming growth factor-␤. In: Bilezikian JP, Manolagas SC 1992 17␤-Estradiol inhibits interleukin-6 produc- Raisz L, Rodan GA (eds) Principles of Bone Biology. Academic tion by bone marrow-derived stromal cells and osteoblasts in vitro: Press, San Diego, CA, pp 647–659 ␤ a potential mechanism for the antiosteoporotic effect of estrogens. 59. Kulkarni AB, Karlsson S 1993 Transforming growth factor- 1 J Clin Invest 89:883–891 knockout mice: a mutation in one cytokine gene causes a dramatic 42. Franchimont N, Canalis E 1995 Platelet-derived growth factor inflammatory disease. Am J Pathol 143:3–9 stimulates the synthesis of interleukin-6 in cells of the osteoblast 60. Suda T, Udagawa N, Takahashi N 1996 Cells of bone: osteoclast lineage. Endocrinology 136:5469–5475 generation. In: Bilezikian JP, Raisz L, Rodan GA (eds) Principals of 43. Franchimont N, Durant D, Rydziel S, Canalis E 1999 Platelet- Bone Biology. Academic Press, San Diego, CA, pp 87–102 derived growth factor induces interleukin-6 transcription in osteo- 61. Rico H, Hernandez ER, Revilla M, Go´mez-Castresana F 1992 blasts through the activator protein-1 complex and activating tran- Salmon calcitonin reduces vertebral fracture rate in postmeno- scription factor-2. J Biol Chem 274:6783–6789 pausal crush fracture syndrome. Bone Miner 16:131–138 44. Stahl N, Yancopoulos GD 1993 The alphas, betas, and kinases of 62. Ikegame M, Rakopoulos M, Zhou H, Houssami S, Martin TJ, cytokine receptor complexes. Cell 74:587–590 Moseley JM, Findlay DM 1995 Calcitonin receptor isoforms in 45. Stahl N, Boulton TG, Farruggella T, Ip NY, Davis S, Witthuhn mouse and rat osteoclasts. J Bone Miner Res 10:59–65 BA, Quelle FW, Silvennoinen O, Barbieri G, Pellegrini S, Ihle JN, 63. Martin TJ, Udagawa N 1998 Hormonal regulation of osteoclast Yancopoulos GD 1994 Association and activation of Jak-Tyk ki- function. Trends Endocrinol Metab 9:6–12 nases by CNTF-LIF- OSM-IL-6 ␤ receptor components. Science 64. Girasole G, Passeri G, Jilka RL, Manolagas SC 1994 Interleukin- 263:92–95 11: a new cytokine critical for osteoclast development. J Clin Invest 46. Boulton TG, Stahl N, Yancopoulos D 1994 Ciliary neurotrophic 93:1516–1524 factor/leukemia inhibitory factor/interleukin 6/oncostatin M fam- 65. Greenfield EM, Shaw SM, Gornik SA, Banks MA 1995 Adenyl ily of cytokines induces tyrosine phosphorylation of a common set cyclase and interleukin 6 are downstream effectors of parathyroid of proteins overlapping those induced by other cytokines and hormone resulting in stimulation of bone resorption. J Clin Invest growth factors. J Biol Chem 269:11648–11655 96:1238–1244 47. Darnell Jr JE 1997 STATs and gene regulation. Science 277:1630– 66. Grey A, Mitnick MA, Masiukiewicz U, Sun BH, Rudikoff S, Jilka 1635 RL, Manolagas SC, Insogna K 1999 A role of interleukin-6 in 48. Narazaki M, Yasukawa K, Saito T, Ohsugi Y, Fukui H, Koishihara parathyroid hormone-induced bone resorption in vivo. Endocri- Y, Yancopoulos GD, Taga T, Kishimoto T 1993 Soluble forms of nology 140:4683–4690 the interleukin-6 signal-transducing receptor component gp130 in 67. Bellido T, Jilka RL, Boyce BF, Girasole G, Broxmeyer H, Dal- human serum possessing a potential to inhibit signals through rymple SA, Murray R, Manolagas SC 1995 Regulation of inter- membrane-anchored gp130. Blood 82:1120–1126 leukin-6, osteoclastogenesis and bone mass by androgens: the role 49. Udagawa N, Takahashi N, Katagiri T, Tamura T, Wada S, Findlay of the androgen receptor. J Clin Invest 95:2886–2895 DM, Martin TJ, Hirota H, Tada T, Kishimoto T, Suda T 1995 68. Lakatos P, Foldes J, Horvath C, Kiss L, Tatrai A, Takacs I, Tarjan Interleukin (IL)-6 induction of osteoclast differentiation depends G, Stern PH 1997 Serum interleukin-6 and bone metabolism in on IL-6 receptors expressed on osteoblastic cells but not on oste- patients with thyroid function disorders. J Clin Endocrinol Metab oclast progenitors. J Exp Med 182:1461–1468 82:78–81 50. O’Brien CA, Gubrij I, Lin S-C, Saylors RL, Manolagas SC 1999 69. Ray A, LaForge KS, Sehgal PB 1990 On the mechanism for efficient STAT3 activation in stromal/osteoblastic cells is required for in- repression of the interleukin-6 promoter by glucocorticoids: En- duction of the receptor activator of NF-kB ligand and stimulation hancer, TATA box, and RNA start site (Inr motif) occlusion. Mol of osteoclastogenesis by gp130-utilizing cytokines or interleukin-1 Cell Biol 10:5736–5746 but not 1,25-dihydroxyvitamin D3 or parathyroid hormone. J Biol 70. Grzesik WJ, Gehron Robey P 1994 Bone matrix RGD glycopro- Chem 274:19301–19308 teins: immunolocalization and interaction with human primary 51. Jilka RL, Hangoc G, Girasole G, Passeri G, Williams DC, Abrams osteoblastic bone cells in vitro. J Bone Miner Res 9:487–496 JS, Boyce B, Broxmeyer H, Manolagas SC 1992 Increased oste- 71. Rodan GA 1995 Osteopontin overview. Ann NY Acad Sci 760:1–5 oclast development after estrogen loss: mediation by interleukin-6. 72. Horton MA, Townsend P, Nesbitt S 1996 Cell surface attachment Science 257:88–91 molecules in bone. In: Bilezikian JP, Raisz LG, Rodan GA (eds) 52. Poli V, Balena R, Fattori E, Markatos A, Yamamoto A, Tanaka H, Principles of Bone Biology. Academic Press, San Diego, CA, pp Ciliberto G, Rodan GA, Costantini F 1994 Interleukin-6 deficient 217–230 mice are protected from bone loss caused by estrogen depletion. 73. Yamate T, Mocharla H, Taguchi Y, Igietseme JU, Manolagas SC, EMBO J 13:1189–1196 Abe E 1997 Osteopontin expression by osteoclast and osteoblast 53. Bellido T, Stahl N, Farruggella TJ, Borba V, Yancopoulos GD, progenitors in the murine bone marrow: demonstration of its re- Manolagas SC 1996 Detection of receptors for interleukin-6, inter- quirement for osteoclastogenesis and its increase after ovariec- leukin-11, leukemia inhibitory factor, oncostatin M, and ciliary tomy. Endocrinology 138:3047–3055 neurotrophic factor in bone marrow stromal/osteoblastic cells. 74. Takeuchi Y, Suzawa M, Kikuchi T, Nishida E, Fujita T, Matsu- J Clin Invest 97:431–437 moto T 1997 Differentiation and transforming growth factor-␤ re- April, 2000 BIRTH AND DEATH OF BONE CELLS 133

ceptor down-regulation by collagen-␣2␤1 integrin interaction is myoblasts by the adipogenic transcription factors PPAR ␥ and mediated by focal adhesion kinase and its downstream signals in C/EBP ␣. Proc Natl Acad Sci USA 92:9856–9860 murine osteoblastic cells. J Biol Chem 272:29309–29316 96. Chen D, Ji X, Harris MA, Feng JQ, Karsenty G, Celeste AJ, Rosen 75. Zhao W, Krane S 1998 Inability of collagenase to cleave type I V, Mundy GR, Harris SE 1998 Differential roles for bone mor- collagen in vivo is associated with osteocyte apoptosis. Bone 23:S185 phogenetic protein (BMP) receptor type IB and IA in differentiation (Abstract) and specification of mesenchymal precursor cells to osteoblast and 76. Globus RK, Doty SB, Lull JC, Holmuhamedov E, Humphries MJ, adipocyte lineages. J Cell Biol 142:295–305 Damsky CH 1998 Fibronectin is a survival factor for differentiated 97. Ott SM 1996 Theoretical and methodological approach. In: Bilez- osteoblasts. J Cell Sci 111:1385–1393 ikian JP, Raisz LG, Rodan GA (eds) Principles of Bone Biology. 77. Xiao G, Wang D, Benson MD, Karsenty G, Franceschi RT 1998 Academic Press, San Diego, CA pp 231–241 Role of the alpha2-integrin in osteoblast-specific gene expression 98. Mohan S, Baylink DJ 1991 The role of IGF-1 in the coupling of bone and activation of the osf2 transcription factor. J Biol Chem 273: formation to resorption. In: Spencer EM (ed) Modern Concepts of 32988–32994 Insulin-Like Growth Factors. Elsevier, New York, pp 169–184 78. Ross FP, Chappel J, Alvarez JI, Sander D, Butler WT, Farach- 99. Bonewald LF, Wakefield L, Oreffo ROC, Escobedo A, Twardzik Carson MC, Mintz KA, Robey PG, Teitelbaum SL, Cheresh DA DR, Mundy GR 1991 Latent forms of transforming growth factor-␤ ␤ 1993 Interactions between the bone matrix proteins osteopontin (TGF ) derived from bone cultures: identification of a naturally and bone sialoprotein and the osteoclast integrin ␣ ␤ potentiate occurring 100-kDa complex with similarity to recombinant latent v 3 ␤ bone resorption. J Biol Chem 268:9901–9907 TGF . Mol Endocrinol 5:741–751 79. Owen M, Friedenstein AJ 1988 Stromal stem cells: marrow- 100. Rodan GA 1996 Coupling of bone resorption and formation during derived osteogenic precursors. Ciba Found Symp 136:42–60 bone remodeling. In: Marcus R, Feldman D, Kelsey J (eds) Osteo- 80. Westen H, Bainton DF 1979 Association of alkaline-phosphatase- porosis. Academic Press, San Diego, CA, pp 289–299 positive reticulum cell in bone marrow with granulocyte precur- 101. Corral DA, Amling M, Priemel M, Loyer E, Fuchs S, Ducy P, sors. J Exp Med 150:919–937 Baron R, Karsenty G 1998 Dissociation between bone resorption 81. Weiss L 1988 Bone marrow. In: Weiss L (ed) Cell and Tissue and bone formation in osteopenic transgenic mice. Proc Natl Acad Biology. Urban and Schwarzenberg, Baltimore, pp 469–478 Sci USA 95:13835–13840 82. Bianco P, Costantini M, Dearden LC, Bonucci E 1988 Alkaline 102. Udagawa N, Takahashi N, Akatsu T, Sasaki T, Yamaguchi A, phosphatase positive precursors of adipocytes in the human bone Kodama H, Martin TJ, Suda T 1989 The bone marrow-derived marrow. Br J Haematol 68:401–403 stromal cell lines MC3T3–G2/PA6 and ST2 support osteoclast-like 83. Rouleau MF, Mitchell J, Goltzman D 1988 In vivo distribution of cell differentiation in cocultures with mouse spleen cells. Endocri- parathyroid hormone receptors in bone: evidence that a predom- nology 125:1805–1813 inant osseous target cell is not the mature osteoblast. Endocrinol- 103. Dorheim MA, Sullivan M, Dandapani V, Wu X, Hudson J, Segarini ogy 123:187–192 PR, Rosen DM, Aulthouse AL, Gimble JM 1993 Osteoblastic gene 84. Bianco P, Bradbeer JN, Riminucci M, Boyde A 1993 Marrow expression during adipogenesis in hematopoietic supporting murine bone marrow stromal cells. J Cell Physiol 154:317–328 stromal (Western-Bainton) cells: identification, morphometry, con- 104. Kelly KA, Tanaka S, Baron R, Gimble JM 1998 Murine bone focal imaging and changes in disease. Bone 14:315–320 marrow stromally derived BMS2 adipocytes support differentia- 85. Bennett JH, Joyner CJ, Triffitt JT, Owen ME 1991 Adipocytic cells tion and function of osteoclast-like cells in vitro. Endocrinology cultured from marrow have osteogenic potential. J Cell Sci 99: 139:2092–2101 131–139 105. Jilka RL, Weinstein RS, Takahashi K, Parfitt AM, Manolagas SC 86. Diascro Jr DD, Vogel RL, Johnson TE, Witherup KM, Pitzen- 1996 Linkage of decreased bone mass with impaired osteoblasto- berger SM, Rutledge SJ, Prescott DJ, Rodan GA, Schmidt A 1998 genesis in a murine model of accelerated senescence. J Clin Invest High fatty acid content in rabbit serum is responsible for the dif- 97:1732–1740 ferentiation of osteoblasts into adipocyte-like cells. J Bone Miner 106. Weinstein RS, Jilka RL, Parfitt AM, Manolagas SC 1997 The Res 13:96–106 effects of androgen deficiency on murine bone remodeling and 87. Yeh WC, Cao Z, Classon M, McKnight SL 1995 Cascade regulation bone mineral density are mediated via cells of the osteoblastic of terminal adipocyte differentiation by three members of the lineage. Endocrinology 138:4013–4021 C/EBP family of leucine zipper proteins. Genes Dev 9:168–181 107. Suda T, Takahashi N, Udagawa N, Jimi E, Gillespie MT, Martin 88. Spiegelman BM, Flier JS 1996 Adipogenesis and obesity: rounding TJ 1999 Modulation of osteoclast differentiation and function by the out the big picture. Cell 87:377–389 new members of the tumor necrosis factor receptor and ligand 89. Wu Z, Bucher NL, Farmer SR 1996 Induction of peroxisome pro- families. Endocr Rev 20:345–357 ␥ liferator-activated receptor during the conversion of 3T3 fibro- 108. Anderson DM, Maraskovsky E, Billingsley WL, Dougall WC, ␤ ␦ blasts into adipocytes is mediated by C/EBP , C/EBP , and glu- Tometsko ME, Roux ER, Teepe MC, DuBose RF, Cosman D, cocorticoids. Mol Cell Biol 16:4128–4136 Galibert L 1997 A homologue of the TNF receptor and its ligand 90. Shao D, Lazar MA 1997 Peroxisome proliferator activated receptor enhance T-cell growth and dendritic-cell function. Nature 390: ␥ ␣ , CCAAT/enhancer-binding protein , and cell cycle status reg- 175–179 ulate the commitment to adipocyte differentiation. J Biol Chem 109. Lacey DL, Timms E, Tan HL, Kelley MJ, Dunstan CR, Burgess T, 272:21473–21478 Elliott R, Colombero A, Elliott G, Scully S, Hsu H, Sullivan J, 91. Samuelsson L, Stromberg K, Vikman K, Bjursell G, Enerback S Hawkins N, Davy E, Capparelli C, Eli A, Qian YX, Kaufman S, 1991 The CCAAT/enhancer binding protein and its role in adipo- Sarosi I, Shalhoub V, Senaldi G, Guo J, Delaney J, Boyle WJ 1998 cyte differentiation: evidence for direct involvement in terminal Osteoprotegerin ligand is a cytokine that regulates osteoclast dif- adipocyte development. EMBO J 10:3787–3793 ferentiation and activation. Cell 93:165–176 92. Freytag SO, Paielli DL, Gilbert JD 1994 Ectopic expression of the 110. Yasuda H, Shima N, Nakagawa N, Yamaguchi K, Kinoshaki M, CCAAT/enhancer-binding protein ␣ promotes the adipogenic pro- Mochizuki S, Tomoyasu A, Yano K, Goto M, Murakami A, Tsuda gram in a variety of mouse fibroblastic cells. Genes Dev 8:1654– E, Morinaga T, Higashio K, Udagawa N, Takahashi N, Suda T 1663 1998 Osteoclast differentiation factor is a ligand for osteoprote- 93. Tontonoz P, Hu E, Spiegelman BM 1994 Stimulation of adipo- gerin/osteoclastogenesis-inhibitory factor and is identical to genesis in fibroblasts by PPAR ␥ 2, a lipid-activated transcription TRANCE/RANKL. Proc Natl Acad Sci USA 95:3597–3602 factor. Cell 79:1147–1156 111. Simonet WS, Lacey DL, Dunstan CR, Kelley M, Chang MS, Luthy 94. Lecka-Czernik B, Gubrij I, Moerman E, Kajkenova O, Lipschitz R, Nguyen HQ, Wooden S, Bennett L, Boone T, Shimamoto G, D, Manolagas S, Jilka RL 1999 Inhibition of Osf2/Cbfa1 expression DeRose M, Elliott R, Colombero A, Tan HL, Trail G, Sullivan J, and terminal osteoblast differentiation by PPAR ␥2. J Cell Biochem Davy E, Bucay N, Renshaw-Gegg L, Hughes TM, Hill D, Pattison 74:357–371 W, Campbell P, Boyle WJ 1997 Osteoprotegerin: a novel secreted 95. Hu E, Tontonoz P, Spiegelman BM 1995 Transdifferentiation of protein involved in the regulation of bone density. Cell 89:309–319 134 MANOLAGAS Vol. 21, No. 2

112. Bucay N, Sarosi I, Dunstan CR, Morony S, Tarpley J, Capparelli 135. Nesbitt SA, Horton MA 1997 Trafficking of matrix collagens C, Scully S, Tan HL, Xu WL, Lacey DL, Boyle WJ, Simonet WS through bone-resorbing osteoclasts. Science 276:266–269 1998 Osteoprotegerin-deficient mice develop early onset osteopo- 136. Salo J, Lehenkari P, Mulari M, Metsikko¨K,Va¨a¨na¨nen HK 1997 rosis and arterial calcification. Genes Dev 12:1260–1268 Removal of osteoclast bone resorption products by transcytosis. 113. Kong YY, Yoshida H, Sarosi I, Tan HL, Timms E, Capparelli C, Science 276:270–273 Morony S, Oliveira-dos-Santos AJ, Van G, Itie A, Khoo W, Wake- 137. Udagawa N, Takahashi N, Akatsu T, Tanaka H, Sasaki T, Nishi- ham A, Dunstan CR, Lacey DL, Mak TW, Boyle WJ, Penninger hara T, Koga T, Martin TJ, Suda T 1990 Origin of osteoclasts: JM 1999 OPGL is a key regulator of osteoclastogenesis, lymphocyte mature monocytes and macrophages are capable of differentiating development and lymph-node organogenesis. Nature 397:315–323 into osteoclasts under a suitable microenvironment prepared by 114. Degli-Esposti M 1999 To die or not to die–the quest of the TRAIL bone marrow-derived stromal cells. Proc Natl Acad Sci USA 87: receptors. J Leukoc Biol 65:535–542 7260–7264 115. Emery JG, McDonnell P, Burke MB, Deen KC, Lyn S, Silverman 138. Hayman AR, Jones SJ, Boyde A, Foster D, Colledge WH, Carlton C, Dul E, Appelbaum ER, Eichman C, DiPrinzio R, Dodds RA, MB, Evans MJ, Cox TM 1996 Mice lacking tartrate-resistant acid James IE, Rosenberg M, Lee JC, Young PR 1998 Osteoprotegerin phosphatase (Acp 5) have disrupted endochondral ossification and is a receptor for the cytotoxic ligand TRAIL. J Biol Chem 273: mild osteopetrosis. Development 122:3151–3162 14363–14367 139. Hughes DE, Dai A, Tiffee JC, Li HH, Mundy GR, Boyce BF 1996 116. O’Brien CA, Farrar N, Manolagas SC 1998 Identification of an Estrogen promotes apoptosis of murine osteoclasts mediated by ␤ OSF-2 binding site in the murine RANKL/OPGL gene promoter: TGF- . Nat Med 2:1132–1136 a potential link between osteoblastogenesis and osteoclastogenesis. 140. Jilka RL, Weinstein RS, Bellido T, Parfitt AM, Manolagas SC 1998 Bone 23:S149 (Abstract) Osteoblast programmed cell death (apoptosis): modulation by 117. Manolagas SC 1999 Cell number vs. cell vigor–what really matters growth factors and cytokines. J Bone Miner Res 13:793–802 to a regenerating skeleton? Endocrinology 140:4377–4381 141. Steller H 1995 Mechanisms and genes of cellular suicide. Science 118. Manolagas SC, Weinstein RS 1999 New developments in the 267:1445–1449 pathogenesis and treatment of steroid-induced osteoporosis. J Bone 142. Parfitt AM 1990 Bone-forming cells in clinical conditions. In: Hall Miner Res 14:1061–1066 BK (ed) Bone. The Osteoblast and Osteocyte. Telford Press and CRC 119. Reddy SV, Roodman GD 1998 Control of osteoclast differentia- Press, Boca Raton, FL, vol 1:351–429 tion. Crit Rev Eukaryot Gene Expr 8:1–17 143. Frost HM 1960 In vivo osteocyte death. J Bone Joint Surg [Am] 120. Robey PG, Boskey AL 1995 The biochemistry of bone. In: Marcus 42:138–143 R, Feldman D, Bilezikian JP, Kelsey J (eds) Osteoporosis. Academic 144. Noble BS, Stevens H, Loveridge N, Reeve J 1997 Identification of Press, New York, pp 95–183 apoptotic changes in osteocytes in normal and pathological human 121. Ducy P, Desbois C, Boyce B, Pinero G, Story B, Dunstan C, Smith bone. Bone 20:273–282 E, Bonadio J, Goldstein S, Gundberg C, Bradley A, Karsenty G 145. Hughes DE, Boyce BF 1997 Apoptosis in bone physiology and 1996 Increased bone formation in osteocalcin-deficient mice. Na- disease. Mol Pathol 50:132–137 146. Bellido T, O’Brien CA, Roberson PK, Manolagas SC 1998 Tran- ture 382:448–452 scriptional activation of the p21WAF1,CIP1,SDI1 gene by interleukin-6 122. Delany A, Amling M, Priemel M, Delling G, Howe C, Baron R, type cytokines—a prerequisite for their pro-differentiating and Canalis E 1998 Osteonectin-null mice develop severe osteopenia. anti-apoptotic effects on human osteoblastic cells. J Biol Chem Bone 23:S199 (Abstract) 273:21137–21144 123. Boskey AL 1998 Biomineralization: conflicts, challenges, and op- 147. Quarles LD, Siddhanti SR 1996 Guanine nucleotide binding- portunities. J Cell Biochem Suppl 30–31:83–91 protein coupled signaling pathway regulation of osteoblast-medi- 124. Boskey AL 1996 Matrix proteins and mineralization: an overview. ated bone formation (editorial). J Bone Miner Res 11:1375–1383 Connect Tissue Res 35:357–363 148. Teitelbaum SL, Tondravi MM, Ross FP 1996 Osteoclast biology. 125. Whyte MP 1994 Hypophosphatasia and the role of alkaline phos- In: Marcus R, Feldman D, Kelsey J (eds) Osteoporosis. Academic phatase in skeletal mineralization. Endocr Rev 15:439–461 Press, San Diego, CA, pp 61–94 126. Marotti G, Cane V, Palazzini S, Palumbo C 1990 Structure-func- 149. Suda T, Nakamura I, Jimi E, Takahashi N 1997 Regulation of tion relationships in the osteocyte. Ital J Miner Electro Metab osteoclast function. J Bone Miner Res 12:869–879 4:93–106 150. Lian JB, Stein GS, Stein JL, Van Wijnen AJ 1998 Osteocalcin gene 127. Nijweide PJ, Burger EH, Klein-Nulend J, van der Pluijm G 1996 promoter: unlocking the secrets for regulation of osteoblast growth The Osteocyte. In: Bilezikian JP, Raisz LG, Rodan GA (eds) Prin- and differentiation. J Cell Biochem Suppl 30–31:62–72 ciples of Bone Biology. Academic Press, San Diego, CA, pp 115–126 151. Greenfield EM, Bi Y, Miyauchi A 1999 Regulation of osteoclast 128. Marotti G 1996 The structure of bone tissues and the cellular activity. Life Sci 65:1087–1102 control of their deposition. Ital J Anat Embryol 101:25–79 152. Lajtha L 1983 Stem cell concepts. In: Potten CS (ed) Stem Cells— 129. Aarden EM, Burger EH, Nijweide PJ 1994 Function of osteocytes Their Identification and Characterisation. Churchill Livingston, in bone. J Cell Biochem 55:287–299 New York, pp 1–11 130. Weinstein RS, Jilka RL, Parfitt AM, Manolagas SC 1998 Inhibition 153. Chambers TJ, Magnus CJ 1982 Calcitonin alters behavior of iso- of osteoblastogenesis and promotion of apoptosis of osteoblasts lated osteoclasts. J Pathol 136:27–39 and osteocytes by glucocorticoids: potential mechanisms of their 154. Fuller K, Wong B, Fox S, Choi Y, Chambers TJ 1998 TRANCE is deleterious effects on bone. J Clin Invest 102:274–282 necessary and sufficient for osteoblast-mediated activation of bone 131. Tomkinson A, Gevers EF, Wit JM, Reeve J, Noble BS 1998 The role resorption in osteoclasts. J Exp Med 188:997–1001 of estrogen in the control of rat osteocyte apoptosis. J Bone Miner 155. Wani MR, Fuller K, Kim NS, Choi Y, Chambers T 1999 Prosta- Res 13:1243–1250 glandin E2 cooperates with TRANCE in osteoclast induction from 132. Tomkinson A, Reeve J, Shaw RW, Noble BS 1997 The death of hemopoietic precursors: synergistic activation of differentiation, osteocytes via apoptosis accompanies estrogen withdrawal in hu- cell spreading, and fusion. Endocrinology 140:1927–1935 man bone. J Clin Endocrinol Metab 82:3128–3135 156. Reid IR, Nicholson GC, Weinstein RS, Hosking DJ, Cundy T, 133. Parfitt AM, Mundy GR, Roodman GD, Hughes DE, Boyce BF Kotowicz MA, Murphy WAJ, Yeap S, Dufresne S, Lombardi A, 1996 A new model for the regulation of bone resorption, with Musliner TA, Thompson DE, Yates AJ 1996 Biochemical and ra- particular reference to the effects of bisphosphonates. J Bone Miner diologic improvement in Paget’s disease of bone treated with alen- Res 11:150–159 dronate: a randomized, placebo-controlled trial. Am J Med 101: 134. Bossard MJ, Tomaszek TA, Thompson SK, Amegadzie BY, Han- 341–348 ning CR, Jones C, Kurdyla JT, McNulty DE, Drake FH, Gowen 157. Burgess TL, Qian YX, Kaufman S, Ring BD, Van G, Capparelli C, M, Levy MA 1996 Proteolytic activity of human osteoclast cathep- Kelley M, Hsu HL, Boyle WJ, Dunstan CR, Hu S, Lacey DL 1999 sin K—expression, purification, activation, and substrate identifi- The ligand for osteoprotegerin (OPGL) directly activates mature cation. J Biol Chem 271:12517–12524 osteoclasts. J Cell Biol 145:527–538 April, 2000 BIRTH AND DEATH OF BONE CELLS 135

158. Lean J, Matsuo K, Wong B, Wagner EF, Choi Y, Chambers T 1999 activity and the resulting production of c-Jun and JunD. J Clin Infection with TRANCE-expressing retrovirus for the generation of Invest 104:503–513 osteoclastic cell lines. Bone 23:S165 (Abstract) 178. Sunyer T, Lewis J, Collin-Osdoby P, Osdoby P 1999 Estrogen’s 159. Dobnig H, Turner RT 1997 The effects of programmed adminis- bone-protective effects may involve differential IL-1 receptor reg- tration of human parathyroid hormone fragment (1–34) on bone ulation in human osteoclast-like cells. J Clin Invest 103:1409–1418 histomorphometry and serum chemistry in rats. Endocrinology 179. Kimble RB, Matayoshi AB, Vannice JL, Kung VT, Williams C, 138:4607–4612 Pacifici R 1995 Simultaneous block of interleukin-1 and tumor 160. Manolagas SC 1998 Cellular and molecular mechanisms of osteo- necrosis factor is required to completely prevent bone loss in the porosis. Aging Clin Exp Res 10:182–190 early postovariectomy period. Endocrinology 136:3054–3061 161. Jilka RL 1998 Cytokines, bone remodeling, and estrogen defi- 180. Kitazawa R, Kimble RB, Vannice JL, Kung VT, Pacifici R 1994 ciency: a 1998 update. Bone 23:75–81 Interleukin-1 receptor antagonist and tumor necrosis factor binding 162. Pacifici R 1998 Cytokines, estrogen, and postmenopausal osteo- protein decrease osteoclast formation and bone resorption in ovari- porosis—the second decade. Endocrinology 139:2659–2661 ectomized mice. J Clin Invest 94:2397–2406 163. Lin SC, Yamate T, Taguchi Y, Borba VZ, Girasole G, O’Brien CA, 181. Ammann P, Rizzoli R, Bonjour JP, Bourrin S, Meyer JM, Vassalli Bellido T, Abe E, Manolagas SC 1997 Regulation of the gp80 and P, Garcia I 1997 Transgenic mice expressing soluble tumor necrosis gp130 subunits of the IL-6 receptor by sex steroids in the murine factor-receptor are protected against bone loss caused by estrogen bone marrow. J Clin Invest 100:1980–1990 deficiency. J Clin Invest 99:1699–1703 164. McDonnell DP, Norris JD 1997 Analysis of the molecular phar- 182. Hofbauer LC, Khosla S, Dunstan CR, Lacey DL, Spelsberg TC, macology of estrogen receptor agonists and antagonists provides Riggs BL 1999 Estrogen stimulates gene expression and protein insights into the mechanism of action of estrogen in bone. Osteo- production of osteoprotegerin in human osteoblastic cells. Endo- poros Int 7 Suppl 1:S29–S34 crinology 140:4367–4370 165. Miyaura C, Kusano K, Masuzawa T, Chaki O, Onoe Y, Aoyagi M, 183. Eriksen EF, Langdahl B, Vesterby A, Rungby J, Kassem M 1999 Sasaki T, Tamura T, Koishihara Y, Ohsugi Y, Suda T 1995 En- Hormone replacement therapy prevents osteoclastic hyperactivity: dogenous bone-resorbing factors in estrogen deficiency: coopera- a histomorphometric study in early postmenopausal women. tive effects of IL-1 and IL-6. J Bone Miner Res 10:1365–1373 J Bone Miner Res 14:1217–1221 166. Bismar H, Diel I, Ziegler R, Pfeilschifter J 1995 Increased cytokine 184. Manolagas SC, Weinstein RS, Bellido T, Bodenner DL 1999 Op- secretion by human bone marrow cells after menopause or dis- posite effects of estrogen on the life span of osteoblasts/osteocytes continuation of estrogen replacement. J Clin Endocrinol Metab vs. osteoclasts in vivo and in vitro: an explanation of the imbalance 80:3351–3355 between formation and resorption in estrogen deficiency. J Bone 167. Cheleuitte D, Mizuno S, Glowacki J 1998 In vitro secretion of Miner Res 14:S169 (Abstract) cytokines by human bone marrow: effects of age and estrogen 185. Smith EP, Boyd J, Frank GR, Takahashi H, Cohen RM, Specker status. J Clin Endocrinol Metab 83:2043–2051 B, Williams TC, Lubahn DB, Korach KS 1994 Estrogen resistance 168. Kassem M, Khosla S, Spelsberg TC, Riggs BL 1996 Cytokine caused by a mutation in the estrogen-receptor gene in a man. production in the bone marrow microenvironment: failure to dem- N Engl J Med 331:1056–1061 onstrate estrogen regulation in early postmenopausal women. 186. Carani C, Qin K, Simoni M, Faustini-Fustini M, Serpente S, Boyd J Clin Endocrinol Metab 81:513–518 J, Korach KS, Simpson ER 1997 Effect of testosterone and estradiol 169. Roodman GD, Kurihara N, Ohsaki Y, Kukita A, Hosking D, in a man with aromatase deficiency. N Engl J Med 337:91–95 Demulder A, Smith JF, Singer FR 1992 Interleukin 6. A potential 187. Bilezikian JP, Morishima A, Bell J, Grumbach MM 1998 Increased autocrine/paracrine factor in Paget’s disease of bone. J Clin Invest bone mass as a result of estrogen therapy in a man with aromatase 89:46–52 deficiency. N Engl J Med 339:599–603 170. Klein B, Wijdenes J, Zhang X-G, Jourdan M, Boiron J-M, Brochier 188. Riggs BL, Khosla S, Melton III LJ 1998 Unitary model of osteo- J, Liautard J, Merlin M, Clement C, Morel-Fournier B, Lu Z-Y, porosis revisited. J Bone Miner Res 13:1954–1955 Mannoni P, Sany J, Bataille R 1991 Murine anti-interleukin-6 189. Schwartz BD, Zhu YS, Cordero J, Imperato-McGinley J, 5␣- monoclonal antibody therapy for a patient with plasma cell leu- Reductase deficiency and complete androgen insenstitivity: natural kemia. Blood 78:1198–1204 models to suggest a direct role for androgens on bone density in 171. Kotake S, Sato K, Kim KJ, Takahashi N, Udagawa N, Nakamura men. Program of the 81st Annual Meeting of The Endocrine Society, I, Yamaguchi A, Kishimoto T, Suda T, Kashiwazaki S 1996 San Diego, CA, 1999, p 93 (Abstract) Interleukin-6 and soluble interleukin-6 receptors in the synovial 190. Weinstein RS, Bellido T, Chambers TM, Crawford JA, Swain FL, fluids from rheumatoid arthritis patients are responsible for Han L, Manolagas SC 1999 Like estrogen, androgen exert potent osteoclast-like cell formation. J Bone Miner Res 11:88–95 and direct anti-apoptotic effects on osteoblasts and osteocytes in 172. Takayanagi H, Juji T, Miyazaki T, Iizuka H, Takahashi T, Isshiki vivo and in vitro. J Bone Miner Res 14:S451 (Abstract) M, Okada M, Tanaka Y, Koshihara Y, Oda H, Kurokawa T, Na- 191. Lips P, Courpron P, Meunier PJ 1978 Mean wall thickness of kamura K, Tanaka S 1999 Suppression of arthritic bone destruction trabecular bone packets in the human iliac crest: changes with age. by adenovirus-mediated csk gene transfer to synoviocytes and Calcif Tissue Res 26:13–17 osteoclasts. J Clin Invest 104:137–146 192. Parfitt AM, Villanueva AR, Foldes J, Rao DS 1995 Relations be- 173. Siddiqi A, Monson JP, Wood DF, Besser GM, Burrin JM 1999 tween histologic indices of bone formation: implications for the Serum cytokines in thyrotoxicosis. J Clin Endocrinol Metab 84: pathogenesis of spinal osteoporosis. J Bone Miner Res 10:466–473 435–439 193. Parfitt AM, Han ZH, Palnitkar S, Rao DS, Shih MS, Nelson D 174. Yamamoto T, Ozono K, Kasayama S, Yoh K, Hiroshima K, Takagi 1997 Effects of ethnicity and age or menopause on osteoblast func- M, Matsumoto S, Michigami T, Yamaoka K, Kishimoto T, Okada tion, bone mineralization, and osteoid accumulation in iliac bone. S 1996 Increased IL-6 production by cells isolated from the fibrous J Bone Miner Res 12:1864–1873 bone dysplasia tissues in patients with McCune-Albright syn- 194. Eastell R, Delmas PD, Hodgson SF, Eriksen EF, Mann KG, Riggs drome. J Clin Invest 98:30–35 BL 1988 Bone formation rate in older normal women: concurrent 175. Jilka RL, Takahashi K, Munshi M, Williams DC, Roberson PK, assessment with bone histomorphometry, calcium kinetics, and Manolagas SC 1998 Loss of estrogen upregulates osteoblastogen- biochemical markers. J Clin Endocrinol Metab 67:741–748 esis in the murine bone marrow: evidence for autonomy from 195. Melton LJ, Khosla S, Atkinson EJ, O’Fallon WM, Riggs BL 1997 factors released during bone resorption. J Clin Invest 101:1942–1950 Relationship of bone turnover to bone density and fractures. J Bone 176. Srivastava S, Neale WM, Kimble RB, Rizzo M, Zahner M, Mil- Miner Res 12:1083–1091 brandt J, Patrick RF, Pacifici R 1998 Estrogen blocks M-CSF gene 196. Garnero P, Hausherr E, Chapuy MC, Marcelli C, Grandjean H, expression and osteoclast formation by regulating phosphorylation Muller C, Cormier C, Breart G, Meunier PJ, Delmas PD 1996 of egr-1 and its interaction with Sp-1. J Clin Invest 102:1850–1859 Markers of bone resorption predict hip fracture in elderly women: 177. Srivastava S, Weitzmann MN, Cenci S, Ross FP, Adler S, Pacifici the EPIDOS Prospective Study. J Bone Miner Res 11:1531–1538 R 1999 Estrogen decreases TNF gene expression by blocking JNK 197. Garnero P, Sornay-Rendu E, Chapuy MC, Delmas PD 1996 In- 136 MANOLAGAS Vol. 21, No. 2

creased bone turnover in late postmenopausal women is a major tion of osteoprotegerin production by glucocorticoids in human os- determinant of osteoporosis. J Bone Miner Res 11:337–349 teoblastic lineage cells: potential paracrine mechanisms of glucocor- 198. D’ippolito G, Schiller PC, Ricordi C, Roos BA, Howard GA 1999 ticoid-induced osteoporosis. Endocrinology 140:4382–4389 Age-related osteogenic potential of mesenchymal stromal stem 218. Jilka RL, Weinstein RS, Bellido T, Roberson P, Parfitt AM, Mano- cells from human vertebral bone marrow. J Bone Miner Res 14: lagas SC 1999 Increased bone formation by prevention of osteoblast 1115–1122 apoptosis with PTH. J Clin Invest 104:439–446 199. Mueller S, Glowacki J 1999 The effect of age on the osteogenic 219. Plotkin L, Weinstein RS, Parfitt AM, Roberson PK, Manolagas potential of human bone marrow stromal cells. J Bone Miner Res SC, Bellido T 1999 Prevention of osteocyte and osteoblasts apo- 14:S354 (Abstract) ptosis by bisphosphonates and calcitonin. J Clin Invest 104:1363– 200. Kajkenova O, Lecka-Czernik B, Gubrij I, Hauser SP, Takahashi 1374 K, Parfitt AM, Jilka RL, Manolagas SC, Lipschitz DA 1997 In- 220. Mankin HJ 1992 Nontraumatic necrosis of bone (osteonecrosis). creased adipogenesis and myelopoiesis in the bone marrow of N Engl J Med 326:1473–1479 SAMP6, a murine model of defective osteoblastogenesis and low 221. Weinstein RS, Nicholas RW, Kirchner JR, Crawford JA, Skinner turnover osteopenia. J Bone Miner Res 12:1772–1779 RA, Swain FL, Manolagas SC 1998 Anatomic juxtaposition of 201. Nagy L, Tontonoz P, Alvarez JG, Chen H, Evans RM 1998 Oxi- apoptotic osteocytes and avascular necrosis in femurs from patients dized LDL regulates macrophage gene expression through ligand with glucocorticoid-excess. Bone 23:S461 (Abstract) activation of PPAR␥. Cell 93:229–240 222. Chang DJ, Ji C, Kim KK, Casinghino S, McCarthy TL, Centrella 202. Tontonoz P, Nagy L, Alvarez JG, Thomazy VA, Evans RM 1998 M 1998 Reduction in transforming growth factor beta receptor I PPAR␥ promotes monocyte/macrophage differentiation and up- expression and transcription factor CBFa1 on bone cells by glu- take of oxidized LDL. Cell 93:241–252 cocorticoid. J Biol Chem 273:4892–4896 203. Nuttall ME, Patton AJ, Olivera DL, Nadeau DP, Gowen M 1998 223. Centrella M, Rosen V, Wozney JM, Casinghino SR, McCarthy TL Human trabecular bone cells are able to express both osteoblastic 1997 Opposing effects by glucocorticoid and bone morphogenetic and adipocytic phenotype: implications for osteopenic disorders. protein-2 in fetal rat bone cell cultures. J Cell Biochem 67:528–540 J Bone Miner Res 13:371–382 224. Canalis E 1998 Inhibitory actions of glucocorticoids on skeletal 204. Parhami F, Jackson SM, Tintut Y, Le V, Balucan JP, Territo M, growth. Is local insulin-like growth factor I to blame? Endocrinol- Demer LL 1999 Atherogenic diet and minimally oxidized low ogy 139:3041–3042 density lipoprotein inhibit osteogenic and promote adipogenic dif- 225. Shi XM, Chang ZJ, Blair HC, McDonald JM, Cao X 1998 Glu- ferentiation of marrow stromal cells. J Bone Miner Res 14:2067–2078 cocorticoids induce adipogenesis of stromal cells by transcription- 205. Banks LM, Lees B, MacSweeney JE, Stevenson JC 1994 Effect of ally activating PPARy2. Bone 23:S454 (Abstract) degenerative spinal and aortic calcification on bone density mea- 226. Dempster DW, Cosman F, Parisien M, Shen V 1993 Anabolic surements in post-menopausal women: links between osteoporosis actions of parathyroid hormone on bone. Endocr Rev 14:690–709 and cardiovascular disease? Eur J Clin Invest 24:813–817 227. Tam CS, Heersche JNM, Murray TM, Parsons JA 1982 Parathyroid 206. Laroche M, Pouilles JM, Ribot C, Bendayan P, Bernard J, Boccalon hormone stimulates the bone apposition rate independently of its H, Mazzaferro S 1994 Comparison of the bone mineral content of resorptive action: differential effects of intermittent and continuous the lower limbs in men with ischaemic atherosclerotic disease. Clin administration. Endocrinology 110:506–512 Rheumatol 13:611–614 228. Reeve J, Meunier PJ, Parsons JA, Bernat M, Bijvoet OL, Courpron 207. Shmookler Reis RJ, Benes H, McClure T, Zheng W, Weinstein RS, P, Edouard C, Klenerman L, Neer RM, Renier JC, Slovik D, Shelton R, Jilka RL, Manolagas SC 1999 Genetic mapping of loci Vismans FJ, Potts JTJ 1980 Anabolic effect of human parathyroid conferring osteopenia using closely-related mouse strains. J Bone hormone fragment on trabecular bone in involutional osteoporosis: Miner Res 14:S141 (Abstract) a multicentre trial. Br Med J 280:1340–1344 208. Klein RF, Mitchell SR, Phillips TJ, Belknap JK, Orwoll ES 1998 229. Finkelstein JS, Klibanski A, Arnold AL, Toth TL, Hornstein MD, Quantitative trait loci affecting peak bone mineral density in mice. Neer RM 1998 Prevention of estrogen deficiency-related bone loss J Bone Miner Res 13:1648–1656 with human parathyroid hormone-(1–34): a randomized controlled 209. Shimizu M, Higuchi K, Bennett B, Xia C, Tsuboyama T, Kasai S, trial. JAMA 280:1067–1073 Chiba T, Fujisawa H, Kogishi K, Kitado H, Kimoto M, Takeda N, 230. Lane NE, Sanchez S, Modin GW, Genant HK, Pierini E, Arnaud Matsushita M, Okumura H, Serikawa T, Nakamura T, Johnson C 1998 Parathyroid hormone treatment can reverse corticosteroid- TE, Hosokawa M 1999 Identification of peak bone mass QTL in a induced osteoporosis. J Clin Invest 102:1627–1633 spontaneously osteoporotic mouse strain. Mamm Genome 231. Reeve J, Hesp R, Williams D, Hulme P, Klenerman L, Zanelli JM, 10:81–87 Darby AJ, Tregear GW, Parsons JA 1976 Anabolic effect of low 210. Mohan S, Baylink DJ 1997 Serum insulin-like growth factor bind- doses of a fragment of human parathyroid hormone on the skeleton ing protein (IGFBP)-4 and IGFBP-5 levels in aging and age- in postmenopausal osteoporosis. Lancet 1:1035–1038 associated diseases. Endocrine 7:87–91 232. Stewart AF 1996 PTHrP(1–36) as a skeletal anabolic agent for the 211. Rajaram S, Baylink DJ, Mohan S 1997 Insulin-like growth factor- treatment of osteoporosis. Bone 19:303–306 binding proteins in serum and other biological fluids: regulation 233. Machwate M, Rodan SB, Rodan GA, Harada SI 1998 Sphingosine and functions. Endocr Rev 18:801–831 kinase mediates cyclic AMP suppression of apoptosis in rat peri- 212. Rozman C, Feliu E, Berga L, Reverter JC, Climent C, Ferran MJ osteal cells. Mol Pharmacol 54:70–77 1989 Age-related variations of fat tissue fraction in normal human 234. Turner PR, Bencsik M, Malecz N, Christakos S, Nissenson RA bone marrow depend both on size and number of adipocytes: a 1998 Apoptosis mediated by the PTH/PTHrP receptor: role of JNK stereological study. Exp Hematol 17:34–37 and calcium signaling pathways. Bone 23:S155 (Abstract) 213. Gimble JM, Robinson CE, Wu X, Kelley KA 1996 The function of 235. Leaffer D, Sweeney M, Kellerman LA, Avnur Z, Krstenansky JL, adipocytes in the bone marrow stroma: an update. Bone 19:421–428 Vickery BH, Caulfield JP 1995 Modulation of osteogenic cell ul- 214. Tavassoli M 1989 Fatty involution of marrow and the role of trastructure by RS-23581, an analog of human parathyroid hor- adipose tissue in hemopoiesis. In: Tavassoli M (ed) Handbook of mone (PTH)-related peptide-(1–34), and bovine PTH-(1–34). En- the Hematopoietic Microenvironment. Humana Press, Clifton, NJ, docrinology 136:3624–3631 pp 157–187 236. Dobnig H, Turner RT 1995 Evidence that intermittent treatment 215. Kajkenova O, Gubrij I, Hauser SP, Takahashi K, Jilka RL, Mano- with parathyroid hormone increases bone formation in adult rats lagas SC, Lipschitz DA 1995 Increased hematopoiesis accompanies by activation of bone lining cells. Endocrinology 136:3632–3638 reduced osteoblastogenesis in the senescence-accelerated mouse 237. Hock JM 1999 Stemming bone loss by suppressing apoptosis. J Clin (SAM-P/6). J Bone Miner Res 10[Suppl 1]:S431 (Abstract) Invest 104:371–373 216. Fitzpatrick LA 1994 Glucocorticoid-induced osteoporosis. In: Mar- 238. Papapoulos SE 1996 Bisphosphonates: pharmacology and use in cus R (ed) Osteoporosis. Blackwell Science, Boston, pp 202–226 the treatment of osteoporosis. In: Marcus R, Feldman D, Kelsey J 217. Hofbauer LC, Gori F, Riggs BL, Lacey DL, Dunstan CR, Spelsberg (eds) Osteoporosis. Academic Press, San Diego, CA, pp 1209–1234 TC, Khosla S 1999 Stimulation of osteoprotegerin ligand and inhibi- 239. Azria M, Avioli LV 1996 Calcitonin. In: Bilezikian JP, Raisz LG, April, 2000 BIRTH AND DEATH OF BONE CELLS 137

Rodan GA (eds) Principles of Bone Biology. Academic Press, San T, Kato S, Kawashima H 1997 Rapid activation of MAP kinase by Diego, CA, pp 1083–1097 estrogen in the bone cell line. Biochem Biophys Res Commun 240. Hughes DE, Wright KR, Uy HL, Sasaki A, Yoneda T, Roodman 235:99–102 GD, Mundy GR, Boyce BF 1995 Bisphosphonates promote apo- 253. Aronica SM, Kraus WL, Katzenellenbogen BS 1994 Estrogen ac- ptosis in murine osteoclasts in vitro and in vivo. J Bone Miner Res tion via the cAMP signaling pathway: stimulation of adenylate 10:1478–1487 cyclase and cAMP-regulated gene transcription. Proc Natl Acad Sci 241. Fisher JE, Rogers MJ, Halasy JM, Luckman SP, Hughes DE, USA 91:8517–8521 Masarachia PJ, Wesolowski G, Russell RGG, Rodan GA, Reszka 254. McEwen BS, Alves SE 1999 Estrogen actions in the central nervous AA 1999 Alendronate mechanism of action: geranylgeraniol, an system. Endocr Rev 20:279–307 intermediate in the mevalonate pathway, prevents inhibition of 255. Chen Z, Yuhanna IS, Galcheva-Gargova Z, Karas RH, Mendel- osteoclast formation, bone resorption, and kinase activation in vitro. sohn ME, Shaul PW 1999 Estrogen receptor alpha mediates the Proc Natl Acad Sci USA 96:133–138 nongenomic activation of endothelial nitric oxide synthase by es- 242. Fleisch H 1998 Bisphosphonates: mechanisms of action. Endocr trogen. J Clin Invest 103:401–406 Rev 19:80–100 256. Castoria G, Barone MV, Di Domenico M, Bilancio A, Ametrano 243. Cummings S, Black D, Vogt TM 1996 Changes in BMD substan- D, Migliaccio A, Auricchio F 1999 Non-transcriptional action of tially underestimate the anti-fracture effects of alendronate and oestradiol and progestin triggers DNA synthesis. EMBO J 18:2500– other antiresorptive drugs. J Bone Miner Res 11:S102 (Abstract) 2510 244. Chavassieux PM, Arlot ME, Reda C, Wei L, Yates AJ, Meunier PJ 257. Pietras RJ, Szego CM 1977 Specific binding sites for oestrogen at 1997 Histomorphometric assessment of the long-term effects of the outer surfaces of isolated endometrial cells. Nature 265:69–72 alendronate on bone quality and remodeling in patients with os- 258. Pappas TC, Gametchu B, Watson CS 1995 Membrane estrogen teoporosis. J Clin Invest 100:1475–1480 receptors identified by multiple antibody labeling and impeded- 245. Balena R, Toolan BC, Shea M, Markatos A, Myers ER, Lee SC, ligand binding. FASEB J 9:404–410 Opas EE, Seedor JG, Klein H, Frankenfield D, Quartuccio H, 259. Razandi M, Pedram A, Greene GL, Levin ER 1999 Cell membrane Fioravanti C, Clair J, Brown E, Hayes WC, Rodan GA 1993 The and nuclear estrogen receptors (ERs) originate from a single tran- effects of 2-year treatment with the aminobisphosphonate alen- script: Studies of ER␣ and ER␤ expressed in Chinese hamster ovary dronate on bone metabolism, bone histomorphometry, and bone cells. Mol Endocrinol 13:307–319 strength in ovariectomized nonhuman primates. J Clin Invest 92: 260. Norfleet AM, Thomas ML, Gametchu B, Watson CS 1999 Estrogen 2577–2586 receptor-alpha detected on the plasma membrane of aldehyde- 246. Storm T, Steiniche T, Thamsborg G, Melsen F 1993 Changes in fixed GH3/B6/F10 rat pituitary tumor cells by enzyme-linked im- bone histomorphometry after long-term treatment with intermit- munocytochemistry. Endocrinology 140:3805–3814 tent, cyclic etidronate for postmenopausal osteoporosis. J Bone 261. Manolagas SC, Weinstein RS, Bellido T, Bodenner DL, Jilka RL, Miner Res 8:199–208 Parfitt AM 1999 Activators of non-genomic estrogen-like signalling 247. Mohan S, Baylink D 1996 Therapeutic potential of TGF-beta, BMP, (ANGELS): a novel class of small molecules with bone anabolic and FGF in the treatment of bone loss. In: Bilezikian JP, Raisz LG, properties. J Bone Miner Res 14:S180 (Abstract) Rodan GA (eds) Principles of Bone Biology, pp 1111–1123 262. VanderKuur JA, Hafner MS, Christman JK, Brooks SC 1993 Ef- 248. Rosen C, Wuster C 1996 Growth hormone, insulin-like growth fects of estradiol-17␤ analogues on activation of estrogen response factors. In: Marcus R, Feldman D, Kelsey J (eds) Osteoporosis. element regulated chloramphenicol acetyltransferase expression. Academic Press, San Diego, CA, pp 1313–1333 Biochemistry 32:7016–7021 249. Bodenner DL, Yamamoto M, Kozlowski M, Manolagas SC 1999 263. Christman JK, Nehls S, Polin L, Brooks SC 1995 Relationship Essential requirement of the estrogen receptor alpha or beta for between estrogen structure and conformational changes in estro- (non-genomic) anti-apoptotic effects of estrogen. J Bone Miner Res gen receptor/DNA complexes. J Steroid Biochem Mol Biol 54: 14:S227 (Abstract) 201–210 250. Bellido T, Plotkin LI, Han L, Manolagas SC 1999 Estrogen inhibit 264. Green PS, Gridley KE, Simpkins JW 1998 Nuclear estrogen re- apoptosis of osteoblasts and osteocytes through rapid (non-genom- ceptor-independent neuroprotection by estratrienes: a novel inter- ic) activation of extracellular signal-regulated kinases (ERKs). action with glutathione. Neuroscience 84:7–10 J Bone Miner Res 14:S342 (Abstract) 265. Green PS, Gordon K, Simpkins JW 1997 Phenolic A ring require- 251. Lieberherr M, Grosse B, Kachkache M, Balsan S 1993 Cell sig- ment for the neuroprotective effects of steroids. J Steroid Biochem naling and estrogens in female rat osteoblasts: a possible involve- Mol Biol 63:229–235 ment of unconventional nonnuclear receptors. J Bone Miner Res 266. Shi J, Zhang YQ, Simpkins JW 1997 Effects of 17␤-estradiol on 8:1365–1376 glucose transporter 1 expression and endothelial cell survival fol- 252. Endoh H, Sasaki H, Maruyama K, Takeyama K, Waga I, Shimizu lowing focal ischemia in the rats. Exp Brain Res 117:200–206

STADY II-International Symposium on Signal Transduction in Health and Disease September 12–15, 2000 Tel Aviv, Israel

For further information, contact: Professor Zvi Naor Department of Biochemistry Tel Aviv University Tel Aviv, Israel Telephone: 972-3-640-9032/641-7057 Fax: 972-3-640-6834 E-mail: [email protected] or [email protected] IBMS BoneKEy. 2011 February;8(2):74-83 http://www.bonekey-ibms.org/cgi/content/full/ibmske;8/2/74 doi: 10.1138/20110493

PERSPECTIVES

The Osteoclast Cytoskeleton: How Does It Work?

Steven L. Teitelbaum and Wei Zou Washington University School of Medicine, St. Louis, Missouri, USA

Abstract

The capacity of the osteoclast to resorb bone is distinctive, as is the cell’s appearance. Both characteristics reflect cytoskeletal organization that yields structures such as the sealing zone and ruffled border. The unique nature of these organelles and their dependence upon contact with bone have been appreciated for some time but insights into the mechanisms by which they are generated come from more recent studies. These insights include the role of integrins, particularly αvβ3, in cytoskeletal organization and the canonical signaling pathway they activate. Investigators now appreciate that the sealing zone isolates the resorptive microenvironment from the general extracellular space, permitting secretion of matrix- degrading molecules on the bone surface. Thus, the osteoclast is a secretory cell that depends upon polarization of exocytic vesicles to the bone-apposed plasma membrane into which they insert under the aegis of vesicle/membrane fusion proteins. This process focally expands and convolutes the plasmalemma included within the sealing zone, eventuating in formation of the ruffled border. Many of these events are now better understood and are the focus of this Perspective. IBMS BoneKEy. 2011 February;8(2):74-83. 2011 International Bone & Mineral Society

Isolating the Resorptive gasket-like, sealing zone. A single Microenvironment osteoclast, being a large cell, enjoys multiple contacts with bone and therefore generates Osteoclasts are polykaryons and members numerous sealing zones and resorptive of the macrophage lineage with the unique microenvironments. capacity to degrade the inorganic and organic matrices of bone. If excessive, the When most other cells attach to matrix they bone resorptive activity of osteoclasts generate focal adhesions. These stable causes osteoporosis. Conversely, the structures contain integrins and signaling osteoclast initiates remodeling that likely and cytoskeletal molecules that, upon removes structurally compromised bone, contact with matrix, promote formation of thereby maintaining mechanical integrity (1). actin stress fibers. Consistent with the lack Skeletal health, therefore, requires optimal of actin stress fibers in mammalian osteoclast function. osteoclasts, they lack focal adhesions but in their place develop podosomes (2;3). These Resorption is initiated by attachment of punctuate structures contain an F-actin core osteoclasts to bone. They then develop a and a peripheral “cloud” of a loose network compartment between their plasma of radial actin cables (4-7). In contrast to membrane and the bone surface into which focal adhesions, podosomes are transient the cells transport matrix-degrading but mediate substrate adhesion and thus molecules including H+ and Cl-, which, in formation of the resorptive concert, demineralize the target bone, and microenvironment. , which degrades the exposed collagen fibers and associated proteins. To Most past studies of osteoclast podosomes isolate this resorptive microenvironment utilized cells resident on plastic or glass. In from the general extracellular space, these circumstances, the punctuate osteoclasts reorganize their cytoskeleton to structures initially appear in clusters but generate an encompassing, actin-rich, ultimately coalesce, first into an

74

Copyright 2011 International Bone & Mineral Society

IBMS BoneKEy. 2011 February;8(2):74-83 http://www.bonekey-ibms.org/cgi/content/full/ibmske;8/2/74 doi: 10.1138/20110493 intracytoplasmic actin ring and then a degrading bone and its absence indicates peripheral actin belt (4). The physiological the cell is not doing so. Reflecting multiple relevance of this observation was contacts with bone and attendant sealing challenged by the absence of apparent zones, ruffled borders are also numerous in podosomes or an actin belt in non-stressed a given osteoclast. osteoclasts on bone (4). While the relevance of the peripheral belt is not established, it is This complex enfolding of the plasma clear that, like the actin ring, the sealing membrane, unique to the osteoclast, abuts zone of bone-residing osteoclasts also and extends into the resorptive space. It is consists of podosome-containing structural surrounded by the sealing zone and is the units (8). venue by which the cell secretes matrix- degrading molecules on the bone surface Microtubules Are Important (17). It is therefore the resorptive organelle and is absent or deranged in many forms of Depending upon their state of acetylation, osteopetrosis. As osteoclast resorption microtubules in osteoclasts are transient or alternates with migration, the ruffled border polymerized and relatively stable (9-11). is a transient structure. Unlike actin ring formation by glass-residing cells, sealing zone generation in bone- The ruffled border, which forms only upon resorbing osteoclasts is characterized by contact with mineralized substrate, is microtubule acetylation, again illustrating the initiated by transport of cathepsin K – and/or influence of substrate on the cell’s H+ATPase – and Cl- channel-bearing cytoskeleton (10). vesicles to the bone-apposed plasma membrane (17), likely under the aegis of The histone deacetylase HDAC6 GTPases such as Rabs 7, 9 and 3 (18-22). depolymerizes tubulin, thereby destabilizing While unproven, studies in chicken microtubules. Cbl proteins compete with the osteoclasts raise the possibility that ruffled deacetylase for tubulin binding and thus border-forming vesicles may polarize to the promote polymerization (12). While resorptive surface via microtubules (23). The destabilizing microtubules in other cells (13), polarized vesicles fuse with the bone- RhoA appears to activate HDAC6 in glass- apposed plasma membrane, to increase its generated osteoclasts, suggesting the complexity, via a process mirroring GTPase negatively regulates the cell. When exocytosis (17). Similar to that occurring in on bone, however, osteoclasts in which the context of neurotransmitter exocytosis, RhoA is inhibited lose their apical-basal vesicle/plasmalemma fusion is regulated by polarity and, consequently, are incapable of v- (vesicular) and t- (target) SNAREs optimal resorption (6). Furthermore, RhoA (soluble N-ethylmaleimide-sensitive fusion promotes actin ring and podosome protein (NSF) attachment protein (SNAP) formation and osteoclast motility (6;14). receptors) (24). When osteoclasts contact bone, RhoA is activated and localizes to the cytoskeleton Ruffled border formation requires a (15;16), The fact that RhoA activation is synaptotagmin (Syt) linking the vesicle and diminished, in osteoclasts lacking αvβ3, target plasma membrane. Fifteen Syt establishes that the GTPase is regulated by isoforms have been identified in mammalian the integrin (3). Because of the conflicting cells but Syt VII generates ruffled borders phenotypes of osteoclasts on plastic or and is essential for bone degradation (17). bone, the impact of active RhoA on stability Syt VII also enables osteoblasts to secrete of their microtubules remains unknown. bone matrix proteins and as such, both resorption and formation are repressed in The Ruffled Border Is King Syt VII-deficient mice.

The ruffled border is the morphological sine Autophagy is a cellular degradative process qua non of the resorbing osteoclast as only by which cells recycle organelles and long- its presence assures that the cell is lived proteins. Autophagosomes, which are

75

Copyright 2011 International Bone & Mineral Society

IBMS BoneKEy. 2011 February;8(2):74-83 http://www.bonekey-ibms.org/cgi/content/full/ibmske;8/2/74 doi: 10.1138/20110493 double-membrane bound vesicles, envelop β3(-/-) mice are hypocalcemic and and then deliver cellular components to osteoclast number is substantially increased lysosomes for degradation. While the in these animals, likely reflecting secondary process promotes survival in starved or hyperparathyroidism and an abundance of stressed cells as well as maintenance of osteoclastogenic cytokines in the marrow organelle quality (25-27), recent evidence (35;37;38). However, in contrast, absence of indicates it may also participate in regulated αv/β3 diminishes the abundance of the exocytosis (28). In fact, Atg5, Atg7 and polykaryons in vitro (33). As differentiation, LC3β autophagy proteins, representing the apoptosis and precursor proliferation are not two ubiquitin-like conjugation systems, are compromised, a reasonable hypothesis important for generation of the osteoclast holds that the paucity of osteoclasts, in ruffled border and the secretory function of culture, reflects cytoskeletal dysfunction, osteoclasts both in vitro and in vivo. Thus, and specifically impairment of migration osteoclasts lacking these proteins do not necessary for cell fusion. efficiently polarize cathepsin K to the resorptive microenvironment and are αv/β3 signaling in osteoclasts is initiated by incapable of optimal bone resorption (29). changing the integrin’s conformation from a low to a high affinity state by outside-in or Integrin Activity Starts It All inside-out activation (3;39). Outside-in activation is characterized by integrin Skeletal resorption requires osteoclast-bone clustering, thereby increasing avidity and recognition that is mediated by α/β affinity. Inside-out activation is an indirect heterodimers known as integrins. β1- event wherein signals emanating from containing heterodimers probably participate liganded growth factor or cytokine receptors in the process but αv/β3 is the key integrin target the integrin’s intracellular region, regulating skeletal degradation (30). While changing its conformation and consequently, the αv subunit is constitutively expressed that of the extracellular domain (40). As will throughout osteoclastogenesis, the be discussed, the adaptor protein, talin, is associated β chains alter with differentiation. essential for inside-out αv/β3 activation in Specifically, immature osteoclast precursors, osteoclasts and its absence arrests bone in the form of bone marrow macrophages, resorption. express abundant αvβ5 and little αvβ3. As the cells commit to the osteoclast Resorption is a cyclical event wherein a phenotype, the magnitude of expression of portion of the osteoclast migrates to a the two heterodimers reverses (31). Hence, candidate bone resorptive site and forms an αv/β3 is a relatively specific marker of actin ring and ruffled border. Following osteoclastogenesis. This integrin is liganded matrix degradation, the cell detaches and re- by the amino acids Arg-Gly-Asp (RGD), a initiates the cycle. Prior to bone recognition, motif present in bone proteins such as the integrin is predominantly in a low affinity osteopontin and bone sialoprotein. Small state and confined to podosomes within the molecules mimicking this sequence sealing zone (3;41). Activated αv/β3 leaves suppress osteoclast activity and are the podosome and transits to lamellipodia candidate anti-resorptive drugs (32-34). that mediate motility, compromised in the absence of the integrin (3). During In keeping with its governance of osteoclast resorption, the heterodimer appears in the function, absence of αv/β3, globally and ruffled membrane (3;41). conditionally in osteoclasts, increases bone mass and protects against estrogen- Integrins serve as attachment molecules but deficient osteoporosis (35;36). Reflecting the their intracellular transmission of matrix- integrin’s role in cytoskeletal organization, derived signals is at least as important. For αv/β3 deficiency yields deranged ruffled example, αv/β3 substrate robustly activates borders, failure of cell spreading and sub- ERKs in wild-type osteoclastic cells but not optimal bone resorption (35). Consequently, those lacking the integrin (3). Since

76

Copyright 2011 International Bone & Mineral Society

IBMS BoneKEy. 2011 February;8(2):74-83 http://www.bonekey-ibms.org/cgi/content/full/ibmske;8/2/74 doi: 10.1138/20110493 activation of these MAP kinases typically regulated by the ubiquitinating activity of c- stimulates proliferation, their absence in β3 Cbl (50;51). knockout osteoclasts may contribute to their reduced numbers in vitro. The ITAM-bearing adaptors, Dap 12 and FcRγ, are expressed by osteoclasts and How Does αv/β3 Do It? their combined, but not individual deletion prompts severe osteopetrosis (45;52-54). In 1991, Soriano et al. determined that c-src While deletion of both co-stimulatory deletion eventuates in severe osteopetrosis molecules is reported to arrest due to osteoclast dysfunction (42). osteoclastogenesis (55), we find such is not Interestingly, c-src-deficient mice, like those the case (56), suggesting their resorptive lacking αv/β3, have increased numbers of abnormality reflects deranged osteoclast osteoclasts that fail to organize their function but not generation. The same cytoskeleton. c-src is a tyrosine kinase and obtains regarding osteoblast-mediated an adaptor protein and both functions are generation of osteoclasts lacking Dap12, necessary for optimal cytoskeletal with or without FcRγ. These mutant cells organization (43). form in normal numbers but fail to organize their cytoskeleton or resorb bone. Among Because c-src- and αv/β3-deficient the most dramatic consequences of this osteoclasts share qualitatively similar dysfunction is the inability of Dap12(-/-) cytoskeletal features, the kinase presents as osteoclasts to migrate through a layer of a mediator of integrin signaling. In fact, osteoblasts, required to attach to a under the aegis of phospholipase Cγ candidate resorptive bone surface (56;57). (PLCγ2) (44), c-src binds directly to the β3 Thus the dominant role of ITAM proteins in subunit in the bone resorptive cell, and we the osteoclast appears to be cytoskeletal have found this to be a constitutive event organization and not differentiation (56;58). (45). Others, however, propose that αv/β3 occupancy phosphorylates the focal Syk-mediated organization of the osteoclast adhesion kinase family member, Pyk2, cytoskeleton involves Vav3. This guanine which recruits c-src to the integrin (44;46). nucleotide exchange factor (GEF) is uniquely expressed in abundance in the cell αv/β3-associated c-src phosphorylates c-Cbl and activated upon v 3 occupancy in a which, in turn, inhibits c-src’s activity (3;47). α /β Regardless of the mechanism of SLP-76-dependent manner (15;59). Vavs association, c-src activation requires integrin transit cytoskeleton-organizing Rho GTPases from their inactive GDP- to their occupancy and again, signaling via PLCγ2. active GTP-associated conformation. Thus, Activated c-src prompts podosomal Vav3(-/-) osteoclasts are dysfunctional and disassembly, most probably by the mice from which they are derived are phosphorylating cortactin (48;49). Hence, osteopetrotic. podosomes are more abundant in c-src(-/-) than wild-type osteoclasts and the mutant Vavs are Rac GEFs and it is therefore not cells are less motile. In keeping with Pyk2 surprising that this Rho GTPase regulates regulating the cell’s cytoskeleton, osteoclasts lacking the kinase are unable to the osteoclast cytoskeleton in an αv/β3- generate normal sealing zones on bone dependent manner (60;61). The two (11). The cytoskeletal effects of Pyk2, isoforms expressed in osteoclasts, Rac1 however, may reflect its promotion of tubulin and Rac2, are mutually compensatory (62). acetylation. Effective deletion of both, however, produces severe osteopetrosis in which Syk is another non-receptor tyrosine kinase osteoclasts fail to organize their cytoskeleton. Absence of the related Rho mediating αv/β3 signaling in osteoclasts. family GTPase, cdc42, also causes Upon integrin occupancy it binds the β3 osteopetrosis but in this circumstance the cytoplasmic domain close to c-src, which dominant mechanism is arrested osteoclast activates it (45). Syk is also negatively

77

Copyright 2011 International Bone & Mineral Society

IBMS BoneKEy. 2011 February;8(2):74-83 http://www.bonekey-ibms.org/cgi/content/full/ibmske;8/2/74 doi: 10.1138/20110493

Fig. 1. Proposed mechanism organizing the cytoskeleton of resorbing osteoclasts. 1). M-CSF occupying its receptor, c-fms, stimulates inside-out αvβ3 activation by inducing talin association with the β3 cytoplasmic domain that binds c-src constitutively. 2). Clustering of the integrin by RGD ligand increases avidity as well as affinity by outside-in activation. The liganded integrin activates c-src as evidenced by Y416 phosphorylation. Activated c-src tyrosine phosphorylates ITAM proteins that recruit Syk to the integrin by binding Syk-SH2 domains. c-src activates β3-associated Syk that phosphorylates Vav3 in the context of SLP-76. Vav3 then shuttles Rac-GDP to its activated GTP-associated state. 3). Rac-GTP prompts association of lysosome-derived secretory vesicles with microtubules (MTs) that deliver them to the bone- apposed plasma membrane into which they insert under the influence of Syt VII and LC3. Rac-GTP and MTs also promote sealing zone (SZ) formation. Secretory vesicle fusion focally expands the plasma membrane forming the ruffled border and eventuating in discharge of cathepsin K (CTK) and HCl into the resorptive microenvironment. recruitment due to inhibited precursor which M-CSF structures the osteoclast proliferation and accelerated apoptosis of cytoskeleton may, therefore, be independent the mature polykaryon (63). of the integrin, or alternatively, represent inside-out αv/β3 activation. In fact, M-CSF M-CSF Helps αv/β3 transits αv/β3 from its default low affinity to its high affinity conformation by inducing RANK ligand (RANKL) and M-CSF are the talin binding to the β3 cytoplasmic domain requisite osteoclastogenic cytokines but (39). Absence of talin, in osteoclast each also promotes the resorptive activity of precursors, does not arrest differentiation the mature polykaryon. In the case of M- but blocks substrate adherence and motility. CSF, the cytokine interacting with its The impaired function of talin-deficient receptor, c-fms, stimulates a signaling osteoclasts results in a 5-fold increase in the pathway remarkably similar to that induced bone mass of mutant mice. Interestingly, the by αv/β3, thereby organizing the osteopetrotic phenotype of mice with talin cytoskeleton (37;58;64). The means by (-/-) osteoclasts is more severe than of those

78

Copyright 2011 International Bone & Mineral Society

IBMS BoneKEy. 2011 February;8(2):74-83 http://www.bonekey-ibms.org/cgi/content/full/ibmske;8/2/74 doi: 10.1138/20110493 lacking αv/β3, which likely represents arrest 3. Faccio R, Novack DV, Zallone A, Ross of compensatory integrins, particularly those FP, Teitelbaum SL. Dynamic changes in bearing β1 (30;35). the osteoclast cytoskeleton in response to growth factors and cell attachment Conclusion are controlled by beta3 integrin. J Cell Biol. 2003 Aug 4;162(3):499-509. The magnitude of bone resorption reflects osteoclast number and function of the 4. Saltel F, Chabadel A, Bonnelye E, individual cell, the latter dependent upon Jurdic P. Actin cytoskeletal organisation cytoskeletal organization. The osteoclast in osteoclasts: a model to decipher cytoskeleton is a unique structure whose transmigration and matrix degradation. conversion to its active state depends upon Eur J Cell Biol. 2008 Sep;87(8-9):459- contact with mineralized matrix (Fig. 1). 68. These extracellular signals, which polarize the resorptive machinery to the bone-cell 5. Chabadel A, Bañon-Rodríguez I, Cluet interface, are transmitted intracellularly by D, Rudkin BB, Wehrle-Haller B, Genot integrins dominated by αv/β3. In conjunction E, Jurdic P, Anton IM, Saltel F. CD44 with M-CSF-stimulated inside-out activation, and beta3 integrin organize two a canonical signaling pathway emanates functionally distinct actin-based domains from the αv/β3 integrin. Occupancy of the in osteoclasts. Mol Biol Cell. 2007 heterodimer phosphorylates constitutively Dec;18(12):4899-910. associated c-src which in turn targets Dap 12. The ITAM’s phosphotyrosines serve to 6. Saltel F, Destaing O, Bard F, Eichert D, recruit Syk to the β3 cytoplasmic domain Jurdic P. Apatite-mediated actin where it is also phosphorylated by c-src. dynamics in resorbing osteoclasts. Mol Utilizing SLP-76, Syk activates Vav3, Biol Cell. 2004 Dec;15(12):5231-41. eventuating in formation of Rac-GTP and organization of the resorptive cytoskeleton. 7. Destaing O, Sanjay A, Itzstein C, Horne Given current concerns regarding long- WC, Toomre D, De Camilli P, Baron R. acting anti-resorptive agents, such as The tyrosine kinase activity of c-Src bisphosphonates, short-acting counterparts regulates actin dynamics and are in demand. Delineating the molecular organization of podosomes in mechanism by which osteoclasts organize osteoclasts. Mol Biol Cell. 2008 their cytoskeleton to degrade bone has Jan;19(1):394-404. provided an array of candidate therapeutic targets. 8. Luxenburg C, Geblinger D, Klein E, Anderson K, Hanein D, Geiger B, Conflict of Interest: None reported. Addadi L. The architecture of the adhesive apparatus of cultured Peer Review: This article has been peer-reviewed. osteoclasts: from podosome formation to sealing zone assembly. PLoS ONE. References 2007 Jan 31;2(1):e179.

1. Teitelbaum SL. Bone resorption by 9. Waterman-Storer CM, Salmon E. osteoclasts. Science. 2000 Sep Positive feedback interactions between 1;289(5484):1504-8. microtubule and actin dynamics during cell motility. Curr Opin Cell Biol. 1999 2. Marchisio PC, Bergui L, Corbascio GC, Feb;11(1):61-7. Cremona O, D'Urso N, Schena M, Tesio L, Caligaris-Cappio F. Vinculin, talin, 10. Destaing O, Saltel F, Gilquin B, and integrins are localized at specific Chabadel A, Khochbin S, Ory S, Jurdic adhesion sites of malignant B P. A novel Rho-mDia2-HDAC6 pathway lymphocytes. Blood. 1988 controls podosome patterning through Aug;72(2):830-3.

79

Copyright 2011 International Bone & Mineral Society

IBMS BoneKEy. 2011 February;8(2):74-83 http://www.bonekey-ibms.org/cgi/content/full/ibmske;8/2/74 doi: 10.1138/20110493

microtubule acetylation in osteoclasts. J and osteoblast secretion. Dev Cell. 2008 Cell Sci. 2005 Jul 1;118(Pt 13):2901-11. Jun;14(6):914-25.

11. Gil-Henn H, Destaing O, Sims NA, Aoki 18. Abu-Amer Y, Teitelbaum SL, Chappel K, Alles N, Neff L, Sanjay A, Bruzzaniti JC, Schlesinger P, Ross FP. Expression A, De Camilli P, Baron R, Schlessinger and regulation of RAB3 proteins in J. Defective microtubule-dependent osteoclasts and their precursors. J Bone podosome organization in osteoclasts Miner Res. 1999 Nov;14(11):1855-60. leads to increased bone density in Pyk2(-/-) mice. J Cell Biol. 2007 Sep 10; 19. Zhao H, Laitala-Leinonen T, Parikka V, 178(6):1053-64. Väänänen HK. Downregulation of small GTPase Rab7 impairs osteoclast 12. Purev E, Neff L, Horne WC, Baron R. c- polarization and bone resorption. J Biol Cbl and Cbl-b act redundantly to protect Chem. 2001 Oct 19;276(42):39295-302. osteoclasts from apoptosis and to displace HDAC6 from beta-tubulin, 20. Van Wesenbeeck L, Odgren PR, Coxon stabilizing microtubules and FP, Frattini A, Moens P, Perdu B, podosomes. Mol Biol Cell. 2009 MacKay CA, Van Hul E, Timmermans Sep;20(18):4021-30. JP, Vanhoenacker F, Jacobs R, Peruzzi B, Teti A, Helfrich MH, Rogers MJ, Villa 13. Cook TA, Nagasaki T, Gundersen GG. A, Van Hul W. Involvement of Rho guanosine triphosphatase mediates PLEKHM1 in osteoclastic vesicular the selective stabilization of transport and osteopetrosis in incisors microtubules induced by absent rats and humans. J Clin Invest. lysophosphatidic acid. J Cell Biol. 1998 2007 Apr;117(4):919-30. Apr 6;141(1):175-85. 21. Pavlos NJ, Xu J, Riedel D, Yeoh JS, 14. Chellaiah MA, Soga N, Swanson S, Teitelbaum SL, Papadimitriou JM, Jahn McAllister S, Alvarez U, Wang D, Dowdy R, Ross FP, Zheng MH. Rab3D SF, Hruska KA. Rho-A is critical for regulates a novel vesicular trafficking osteoclast podosome organization, pathway that is required for osteoclastic motility, and bone resorption. J Biol bone resorption. Mol Cell Biol. 2005 Chem. 2000 Apr 21;275(16):11993- Jun;25(12):5253-69. 2002. 22. Zhao H, Ettala O, Väänänen HK. 15. Faccio R, Teitelbaum SL, Fujikawa K, Intracellular membrane trafficking Chappel J, Zallone A, Tybulewicz VL, pathways in bone-resorbing osteoclasts Ross FP, Swat W. Vav3 regulates revealed by cloning and subcellular osteoclast function and bone mass. Nat localization studies of small GTP- Med. 2005 Mar;11(3):284-90. binding rab proteins. Biochem Biophys Res Commun. 2002 May 16. Lakkakorpi PT, Wesolowski G, Zimolo 10;293(3):1060-5. Z, Rodan GA, Rodan SB. Phosphatidylinositol 3-kinase 23. Abu-Amer Y, Ross FP, Schlesinger P, association with the osteoclast Tondravi MM, Teitelbaum SL. Substrate cytoskeleton, and its involvement in recognition by osteoclast precursors osteoclast attachment and spreading. induces C-src/microtubule association. J Exp Cell Res. 1997 Dec 15;237(2):296- Cell Biol. 1997 Apr 7;137(1):247-58. 306. 24. Jahn R, Scheller RH. SNAREs--engines 17. Zhao H, Ito Y, Chappel J, Andrews NW, for membrane fusion. Nat Rev Mol Cell Teitelbaum SL, Ross FP. Biol. 2006 Sep;7(9):631-43. Synaptotagmin VII regulates bone remodeling by modulating osteoclast

80

Copyright 2011 International Bone & Mineral Society

IBMS BoneKEy. 2011 February;8(2):74-83 http://www.bonekey-ibms.org/cgi/content/full/ibmske;8/2/74 doi: 10.1138/20110493

25. Mizushima N, Yoshimori T, Levine B. 33. Engleman VW, Nickols GA, Ross FP, Methods in mammalian autophagy Horton MA, Griggs DW, Settle SL, research. Cell. 2010 Feb 5;140(3):313- Ruminski PG, Teitelbaum SL. A 26. peptidomimetic antagonist of the alpha(v)beta3 integrin inhibits bone 26. Virgin HW, Levine B. Autophagy genes resorption in vitro and prevents in immunity. Nat Immunol. 2009 osteoporosis in vivo. J Clin Invest. 1997 May;10(5):461-70. May 1;99(9):2284-92.

27. Levine B, Kroemer G. Autophagy in the 34. Murphy MG, Cerchio K, Stoch SA, pathogenesis of disease. Cell. 2008 Jan Gottesdiener K, Wu M, Recker R; L- 11;132(1):27-42. 000845704 Study Group. Effect of L- 000845704, an alphaVbeta3 integrin 28. Cadwell K, Liu JY, Brown SL, Miyoshi H, antagonist, on markers of bone turnover Loh J, Lennerz JK, Kishi C, Kc W, and bone mineral density in Carrero JA, Hunt S, Stone CD, Brunt postmenopausal osteoporotic women. J EM, Xavier RJ, Sleckman BP, Li E, Clin Endocrinol Metab. 2005 Mizushima N, Stappenbeck TS, Virgin Apr;90(4):2022-8. HW 4th. A key role for autophagy and the autophagy gene Atg16L1 in mouse 35. McHugh KP, Hodivala-Dilke K, Zheng and human intestinal Paneth cells. MH, Namba N, Lam J, Novack D, Feng Nature. 2008 Nov 13;456(7219):259-63. X, Ross FP, Hynes RO, Teitelbaum SL. Mice lacking beta3 integrins are 29. DeSelm C, Miller B, Zou W, Virgin H, osteosclerotic because of dysfunctional Teitelbaum S. Autophagy related osteoclasts. J Clin Invest. 2000 proteins mediate osteoclast function in Feb;105(4):433-40. vitro and in vivo. J Bone Miner Res. 2010;25(Suppl 1). [Abstract] 36. Zhao H, Kitaura H, Sands MS, Ross FP, Teitelbaum SL, Novack DV. Critical role 30. Helfrich MH, Nesbitt SA, Lakkakorpi PT, of beta3 integrin in experimental Barnes MJ, Bodary SC, Shankar G, postmenopausal osteoporosis. J Bone Mason WT, Mendrick DL, Väänänen Miner Res. 2005 Dec;20(12):2116-23. HK, Horton MA. Beta 1 integrins and osteoclast function: involvement in 37. Faccio R, Takeshita S, Zallone A, Ross collagen recognition and bone FP, Teitelbaum SL. c-Fms and the resorption. Bone. 1996 Oct;19(4):317- alphavbeta3 integrin collaborate during 28. osteoclast differentiation. J Clin Invest. 2003 Mar;111(5):749-58. 31. Inoue M, Ross FP, Erdmann JM, Abu- Amer Y, Wei S, Teitelbaum SL. Tumor 38. Weir EC, Lowik CW, Paliwal I, Insogna necrosis factor alpha regulates KL. Colony stimulating factor-1 plays a alpha(v)beta5 integrin expression by role in osteoclast formation and function osteoclast precursors in vitro and in in bone resorption induced by vivo. Endocrinology. 2000 parathyroid hormone and parathyroid Jan;141(1):284-90. hormone-related protein. J Bone Miner Res. 1996 Oct;11(10):1474-81. 32. Yamamoto M, Fisher JE, Gentile M, Seedor JG, Leu CT, Rodan SB, Rodan 39. Zou W, Petrich B, Monkley S, Critchley GA. The integrin ligand echistatin D, Ginsberg M, Teitelbaum S. Talin is prevents bone loss in ovariectomized critical for osteoclast function by mice and rats. Endocrinology. 1998 mediating inside-out integrin activation. Mar;139(3):1411-9. J Bone Miner Res. 2010;25(Suppl 1). [Abstract]

81

Copyright 2011 International Bone & Mineral Society

IBMS BoneKEy. 2011 February;8(2):74-83 http://www.bonekey-ibms.org/cgi/content/full/ibmske;8/2/74 doi: 10.1138/20110493

40. Shattil SJ, Kim C, Ginsberg MH. The localized in podosomes and activated by final steps of integrin activation: the end beta(2)-integrin ligation. Cell Motil game. Nat Rev Mol Cell Biol. 2010 Cytoskeleton. 2000 Nov;47(3):174-88. Apr;11(4):288-300. 48. Wu H, Reynolds AB, Kanner SB, Vines 41. Faccio R, Grano M, Colucci S, Villa A, RR, Parsons JT. Identification and Giannelli G, Quaranta V, Zallone A. characterization of a novel cytoskeleton- Localization and possible role of two associated pp60src substrate. Mol Cell different alpha v beta 3 integrin Biol. 1991 Oct;11(10):5113-24. conformations in resting and resorbing osteoclasts. J Cell Sci. 2002 Jul 49. Tehrani S, Faccio R, Chandrasekar I, 15;115(Pt 14):2919-29. Ross FP, Cooper JA. Cortactin has an essential and specific role in osteoclast 42. Soriano P, Montgomery C, Geske R, actin assembly. Mol Biol Cell. 2006 Bradley A. Targeted disruption of the c- Jul;17(7):2882-95. src proto-oncogene leads to osteopetrosis in mice. Cell. 1991 Feb 50. Shao Y, Yang C, Elly C, Liu YC. 22;64(4):693-702. Differential regulation of the B cell receptor-mediated signaling by the E3 43. Schwartzberg PL, Xing L, Hoffmann O, ubiquitin ligase Cbl. J Biol Chem. 2004 Lowell CA, Garrett L, Boyce BF, Varmus Oct 15;279(42):43646-53. HE. Rescue of osteoclast function by transgenic expression of kinase- 51. Zou W, Reeve JL, Zhao H, Ross FP, deficient Src in src-/- mutant mice. Teitelbaum SL. Syk tyrosine 317 Genes Dev. 1997 Nov 1;11(21):2835- negatively regulates osteoclast function 44. via the ubiquitin-protein isopeptide ligase activity of Cbl. J Biol Chem. 2009 44. Epple H, Cremasco V, Zhang K, Mao D, Jul 10;284(28):18833-9. Longmore GD, Faccio R. Phospholipase Cgamma2 modulates integrin signaling 52. Faccio R, Zou W, Colaianni G, in the osteoclast by affecting the Teitelbaum SL, Ross FP. High dose M- localization and activation of Src kinase. CSF partially rescues the Dap12-/- Mol Cell Biol. 2008 Jun;28(11):3610-22. osteoclast phenotype. J Cell Biochem. 2003 Dec 1;90(5):871-83. 45. Zou W, Kitaura H, Reeve J, Long F, Tybulewicz VL, Shattil SJ, Ginsberg MH, 53. Mócsai A, Humphrey MB, Van Ziffle JA, Ross FP, Teitelbaum SL. Syk, c-Src, the Hu Y, Burghardt A, Spusta SC, alphavbeta3 integrin, and ITAM Majumdar S, Lanier LL, Lowell CA, immunoreceptors, in concert, regulate Nakamura MC. The immunomodulatory osteoclastic bone resorption. J Cell Biol. adapter proteins DAP12 and Fc receptor 2007 Mar 12;176(6):877-88. gamma-chain (FcRgamma) regulate development of functional osteoclasts 46. Sanjay A, Houghton A, Neff L, through the Syk tyrosine kinase. Proc DiDomenico E, Bardelay C, Antoine E, Natl Acad Sci U S A. 2004 Apr Levy J, Gailit J, Bowtell D, Horne WC, 20;101(16):6158-63. Baron R. Cbl associates with Pyk2 and Src to regulate Src kinase activity, 54. Tomasello E, Vivier E. alpha(v)beta3 integrin-mediated KARAP/DAP12/TYROBP: three names signaling, cell adhesion, and osteoclast and a multiplicity of biological functions. motility. J Cell Biol. 2001 Jan Eur J Immunol. 2005 Jun;35(6):1670-7. 8;152(1):181-95. 55. Koga T, Inui M, Inoue K, Kim S, 47. Duong LT, Rodan GA. PYK2 is an Suematsu A, Kobayashi E, Iwata T, adhesion kinase in macrophages, Ohnishi H, Matozaki T, Kodama T,

82

Copyright 2011 International Bone & Mineral Society

IBMS BoneKEy. 2011 February;8(2):74-83 http://www.bonekey-ibms.org/cgi/content/full/ibmske;8/2/74 doi: 10.1138/20110493

Taniguchi T, Takayanagi H, Takai T. Costimulatory signals mediated by the 60. Razzouk S, Lieberherr M, Cournot G. ITAM motif cooperate with RANKL for Rac-GTPase, osteoclast cytoskeleton bone homeostasis. Nature. 2004 Apr and bone resorption. Eur J Cell Biol. 15;428(6984):758-63. 1999 Apr;78(4):249-55.

56. Zou W, Zhu T, Craft CS, Broekelmann 61. Hall A. G proteins and small GTPases: TJ, Mecham RP, Teitelbaum SL. distant relatives keep in touch. Science. Cytoskeletal dysfunction dominates in 1998 Jun 26;280(5372):2074-5. DAP12-deficient osteoclasts. J Cell Sci. 2010 Sep 1;123(Pt 17):2955-63. 62. Croke M, Ross F, Teitelbaum S. Rac deletion in osteoclasts causes severe 57. Saltel F, Chabadel A, Zhao Y, Lafage- osteopetrosis. J Bone Miner Res. Proust MH, Clézardin P, Jurdic P, 2010;25(Suppl 1). [Abstract] Bonnelye E. Transmigration: a new property of mature multinucleated 63. Ito Y, Teitelbaum SL, Zou W, Zheng Y, osteoclasts. J Bone Miner Res. 2006 Johnson JF, Chappel J, Ross FP, Zhao Dec;21(12):1913-23. H. Cdc42 regulates bone modeling and remodeling in mice by modulating 58. Zou W, Reeve JL, Liu Y, Teitelbaum SL, RANKL/M-CSF signaling and osteoclast Ross FP. DAP12 couples c-Fms polarization. J Clin Invest. 2010 Jun activation to the osteoclast cytoskeleton 1;120(6):1981-93. by recruitment of Syk. Mol Cell. 2008 Aug 8;31(3):422-31. 64. Ross FP, Teitelbaum SL. alphavbeta3 and macrophage colony-stimulating 59. Reeve JL, Zou W, Liu Y, Maltzman JS, factor: partners in osteoclast biology. Ross FP, Teitelbaum SL. SLP-76 Immunol Rev. 2005 Dec;208:88-105. couples Syk to the osteoclast cytoskeleton. J Immunol. 2009 Aug 1;183(3):1804-12.

83

Copyright 2011 International Bone & Mineral Society

REVIEW

Osteoclast Activity and Subtypes as a Function of Physiology and Pathology—Implications for Future Treatments of Osteoporosis

K. Henriksen, J. Bollerslev, V. Everts, and M. A. Karsdal

Nordic Bioscience A/S (K.H., M.A.K.), DK-2730 Herlev, Denmark; Section of Endocrinology (J.B.), Department of Medicine, Rikshospitalet, Oslo University Hospital and the University of Oslo, 0450 Oslo, Norway; and Department Oral Cell Biology (V.E.), Academic Centre of Dentistry Amsterdam, University of Amsterdam and VU University Amsterdam, Research Institute Move, 1066 EA Amsterdam, The Netherlands

Osteoclasts have traditionally been associated exclusively with catabolic functions that are a prerequisite for bone resorption. However, emerging data suggest that osteoclasts also carry out functions that are important for optimal bone formation and bone quality. Moreover, recent findings indicate that osteoclasts have different subtypes depending on their location, genotype, and possibly in response to drug intervention.

The aim of the current review is to describe the subtypes of osteoclasts in four different settings: 1) physiological, in relation to turnover of different bone types; 2) pathological, as exemplified by monogenomic disorders; 3) pathological, as identified by different disorders; and 4) in drug-induced situations.

The profiles of these subtypes strongly suggest that these osteoclasts belong to a heterogeneous cell population, namely, a diverse macrophage-associated cell type with bone catabolic and anabolic functions that are depen- dent on both local and systemic parameters. Further insight into these osteoclast subtypes may be important for understanding cell–cell communication in the bone microenvironment, treatment effects, and ultimately bone quality. (Endocrine Reviews 32: 31–63, 2011)

I. Introduction F. Other diseases characterized by increased oste- II. Bone Remodeling oclast activity III. The Classical Osteoclast VI. Drug-Induced Osteoclast Subtypes A. Existing drugs IV. Osteoclast Subtypes in Physiological Situations B. Future treatments A. Endochondral vs. intramembranous bone VII. The Bone Anabolic Effects of the Osteoclasts osteoclasts VIII. Conclusions and Future Perspectives B. Chondroclasts C. Osteoclasts involved in targeted and stochastic remodeling D. Trabecular and cortical osteoclasts I. Introduction E. Diurnal variation in osteoclasts or osteoclast activity? steoclasts are multinucleated bone-resorbing cells V. Osteoclast Subtypes in Pathological Situations O that are unique in their ability to degrade mineral- A. Osteoporotic osteoclasts ized matrices, such as bone and calcified cartilage (1). Os- B. Changes in osteoclast activities with increasing teoclasts have for a long time been considered bone- bone matrix age C. Osteoclast-rich osteopetrosis resorbing “machines,” yet some years ago it was D. Osteoclast-poor osteopetrosis demonstrated that not all osteoclasts are the same and that E. Pycnodysostotic osteoclasts careful elucidation of the osteoclast subtype may prove

ISSN Print 0021-972X ISSN Online 1945-7197 Abbreviations: AGE, Advanced glycation end-product; BMD, bone mineral density; CAII, car- Printed in U.S.A. bonic anhydrase II; ClC-7, chloride channel 7; CT-1, cardiotrophin-1; CTX, C-terminal Copyright © 2011 by The Endocrine Society crosslinked telopeptide of type I collagen; ER, estrogen receptor; GLP, glucagon-like peptide; doi: 10.1210/er.2010-0006 Received March 19, 2010. Accepted August 10, 2010. HRT, hormone replacement therapy; ICTP, carboxyterminal peptide of type I collagen; MMP, First Published Online September 17, 2010 matrix metalloproteinase; ONJ, ; OPG, osteoprotegerin; RA, rheuma- toid arthritis; RANK, receptor activator of nuclear factor ␬B; RANKL, RANK ligand; SERM, selective estrogen receptor modulator; TRACP, tartrate-resistant acid phosphatase; V-ATPase, vacuolar type ATPase.

Endocrine Reviews, February 2011, 32(1):31–63 edrv.endojournals.org 31 32 Henriksen et al. Osteoclast Subtypes Endocrine Reviews, February 2011, 32(1):31–63 beneficial (1–4). As illustrated in Table 1, under normal that cannot be completely countered by an increase in circumstances, osteoclasts are influenced by a complex bone formation. This results in low bone mass, in deteri- combination of systemic hormones and local mediators oration of the microarchitecture of the skeleton, and often present in the different bones. Under pathological condi- in fractures (25). Osteoporotic fractures are associated tions, such as cessation of estrogen production or inflam- with increased morbidity and mortality and give rise to a matory conditions, additional cytokines are present. Even significant public health problem (24). the bone type and age may influence the phenotype of the Osteopetrosis, on the other hand, is a rare, inherited osteoclast (2, 5–7). Finally and importantly, different disease in various species including man, which was orig- classes of drugs used for treatment of osteoporosis and inally identified by Albers-Scho¨ nberg in 1904 (26). In the other diseases also influence the osteoclasts significantly. majority of cases, it is caused by defective resorption by the The aim of this review is to provide a thorough descrip- osteoclasts, resulting in high bone mass with poor bone tion of the complex nature of osteoclasts under healthy quality and increased fracture frequency due to defective and diseased states and to describe their modulation by bone remodeling (1, 26, 27). However, recent studies also drugs that have been approved for use or are under de- characterized patients with osteopetrosis due to dys- velopment. The paper will also emphasize the role of os- functional osteoclastogenesis either directly affecting teoclasts in initiating bone formation, a recently discov- the osteoclast precursors or indirectly through the os- ered activity of these cells that has gained much attention teoblasts, and thus the phenotype was caused by the (3, 8–13). absence of osteoclasts, rather than inactivity of these cells (28–31). Interestingly, the studies of osteopetrotic patients have indicated that the presence of osteoclasts, II. Bone Remodeling but not their activity, is essential for bone formation, indicating that some aspects of the coupling principle Bone remodeling is required for optimal control of cal- should be revised (1, 3). cium homeostasis and strength of the bones and is essential Because hypogonadal osteoporosis is associated with for the continued maintenance of a healthy skeleton (14). increased numbers and activity of osteoclasts (16), most Bone remodeling is performed by three cell types: 1) the treatments developed so far, such as bisphosphonates and osteoclasts, which are the sole cells in the body possessing hormone replacement therapy (HRT)/selective estrogen the ability to degrade both the inorganic calcium matrix receptor modulators (SERMs), have focused on eliminat- and the organic collagen matrix; 2) the osteoblasts, which ing or reducing the number of osteoclasts and thereby are the bone-forming cells; and 3) the osteocytes, which reducing bone resorption (32). These treatments are as- appear to regulate the activity of both osteoclasts and os- sociated with secondary decreases in bone formation due teoblasts (14, 15). In healthy adults, under normal cir- to the coupled nature of the bone remodeling process, cumstances, bone resorption is always followed by an which naturally limits their efficacy (3, 24). However, as equal degree of bone formation, a tightly balanced process seen in the osteopetrotic syndromes, there are indications referred to as coupling (9, 16). The modulation of activ- that bone resorption and bone formation can be dissoci- ities of the cells involved in the remodeling cycle was re- ated, and from recent studies it appears that the osteoclast cently described in detail (15). itself, whether it is a physiological, pathological, or drug- Coupling was initially discovered in the 1960s when induced subtype, is highly important for a secondary effect Frost and co-workers (17, 18) demonstrated that osteo- on bone formation (1, 3). blasts filled the resorption pits created by osteoclasts in In this review, we describe differences in osteoclast more than 97% of the cases (17–21). Since then, coupling activity and subtypes in relation to physiology, patho- has been understood as a coordinated and balanced in- physiology, and medication, with special attention to duction of osteoblastic bone formation in response to coupling in the bone remodeling process. Ultimately, prior bone resorption (19). Uncoupling occurs when the this review highlights potential directions for new treat- balance between resorption and formation is disrupted, ment modalities. which often leads to pathological situations such as os- teoporosis or osteopetrosis (3, 9, 14, 22, 23). However, uncoupling also occurs under physiological conditions, III. The Classical Osteoclast i.e., during skeletal growth in children, where bone for- mation exceeds bone resorption (10). Osteoclastogenesis is a complex process requiring both the Hypogonadal osteoporosis is usually caused by a de- correct extracellular stimuli and the correct cellular mol- crease or loss of sex steroid production, which results in ecules to interact without impediment (22, 33). Osteoclasts accelerated osteoclastogenesis and bone resorption (24) arise from hematopoietic stem cells that, in the presence of Endocrine Reviews, February 2011, 32(1):31–63 edrv.endojournals.org 33

TABLE 1. A simplified summary of osteoclast phenotypes as a function of physiology, pathology, and drugs, also indicating areas of osteoclast biology that are not well-understood

Osteoclast no. Bone formation Resorptive process Bone resorption Acid Cat K MMP Classical osteoclast Normal Normal ϩϩϩ ϩϩϩ ϩ/Ϫ Balanced Physiology Targeted Recruitment to specific Increased ϩϩ ? ? Balanced areas increased Stochastic Not clear Not clear ϩ ? ? Not clear Night Normal Increased ϩϩ ϩϩ ϩ/Ϫ Minor up-regulation Day Normal Decreased ϩ ? ? Minor down-regulation Chondroclast Normal Normal ϩϩϩϩBalanced Endochondral Normal Normal ϩϩ ϪBalanced Intramembraneous Normal Normal ϩϪ ϩNot balanced, opposite side of bone than resorption Trabecular Normal Normal ϩϩ ϩϩ Ϫ Balanced Cortical Normal Normal ϩϩ ϩϩ Ϫ Balanced Pathology Osteoporosis Increased Increased ϩϩϩϩ Ϫ Increased, but less than resorption Age Increased Increased ϩϩ ϩϩ ? Decreased OC-rich OP Greatly increased Decreased ϪϪ ϪIncreased according to increased OC number OC-poor OP No osteoclasts Decreased ϪϪ ϪDecreased? Pycnodysostosis Unchanged/increased Decreased ϩ Ϫ ϩϩϩ Not clear osteoclast size Paget’s Greatly increased at Increased at local sites ϩϩ ϩ ϩϩϩ Increased locally, but local sites does not compensate resorption RA Greatly increased at Not known ϩ ϩ ϩϩϩ Not known local sites Lytic metastases Greatly increased at Increased at local sites ϩ ϩ ϩϩϩ Increased locally, but local sites does not compensate resorption Drug-induced BPs Reduced Decreased ϪϪ ? Decreased secondary to resorption HRT/SERMs Reduced Decreased ϪϪ ? Decreased secondary to resorption Calcitonin Unchanged Decreased ϪϪ ? Not changed or minor decrease PTH Increased/unchanged Decreased ϩϩ ϩϩ ? Increased, but only temporarily Strontium ranelate Unchanged Decreased? ϪϪ ? Increased GCs Unchanged/increased Unchanged/increased ϩϩ ? Decreased strongly Denosumab Greatly reduced Decreased ϪϪ ϪDecreased secondary to resorption Cat K inhibitors Unchanged Decreased ϩ Ϫ ϩϩϩ Decreased secondary to resorption GLP-2 Unchanged Decreased ϪϪ ? Not changed, but long term effects are not known Acidification inhibitors Increased Decreased ϪϪ ϪIncreased, but so far only in animal models

The table shows the subtype of osteoclasts, the number of osteoclasts, the effect on bone resorption, which part of the resorption machinery that is active/affected, and the effect on bone formation. OC, Osteoclast; OP, osteopetrosis; Cat K, cathepsin K; BPs, bisphosphonates; GCs, glucocorticoids; MMP, matrix metalloproteinase; RA, rheumatoid arthritis; PTH, parathyroid hormone; HRT, hormone replacement therapy; SERMs, selective estrogen receptor modulators; GLP-2, glucagon-like peptide 2. 34 Henriksen et al. Osteoclast Subtypes Endocrine Reviews, February 2011, 32(1):31–63

FIG. 1. Schematic illustration of the molecules involved in osteoclastogenesis and function. (See Refs. 1, 33, and 42). NF␬B, nuclear factor ␬ B; TRAF6, TNF receptor-associated factor 6; nuclear factor of activated T-cells, cytoplasmic, calcineurin-dependent 1; PYK2, Proline-rich Tyrosine Kinase 2; MitF, microphthalmia-associated transcription factor; ClC-7; Chloride Channel 7; PLEKHM1, pleckstrin homology domain containing, family M (with RUN domain) member 1; osteopetrosis associated transmembrane protein 1; CA2, carbonic anhydrase II; AE2, anion exchanger 2. receptor activator of nuclear factor ␬B (RANK) ligand teolysis, although these processes likely occur at the same (RANKL) and macrophage-colony stimulating factor, un- time (44, 51). dergo differentiation and fusion resulting in large multinu- Bone resorption is initiated by active secretion of pro- cleated cells characterized by expression of a series of oste- tons through a vacuolar type ATPase (V-ATPase) and pas- oclast markers, such as tartrate-resistant acid phosphatase sive transport of chloride through a chloride channel (52, (TRACP), matrix metalloproteinase (MMP)-9, cathepsin K, 53). The secretion of hydrochloric acid leads to dissolution carbonic anhydrase II (CAII), the a3 subunit of the vacuolar of the inorganic matrix of the bones (54). The osteoclastic [Hϩ]-ATPase, chloride channel 7 (ClC-7), osteopetrosis-as- V-ATPase is functionally specific and contains the a3 sub- sociated transmembrane protein 1, and the calcitonin recep- unit, and accordingly, loss of a3 leads to osteopetrosis (37, tor (34–41). Osteoclastogenesis and the molecules involved 55–57). In both mice and man, the chloride channel ClC-7, in this process are summarized in Fig. 1, but are not has been shown to mediate chloride transport, thereby discussed in any further detail because several excellent ensuring the electrochemical balance required for intense reviews have been published recently on this topic (1, acidification (Fig. 2) (8, 39, 58). Recent data showed that 22, 33, 42). ClC-7 functions as a proton-chloride antiporter (59, 60). Polarization and formation of the sealing zone, which To generate the necessary levels of Hϩ and ClϪ, the is a specialized ring structure containing a high number of enzyme CAII catalyzes conversion of CO2 and H2O into ␤ ϩ Ϫ -actin filaments, are the next steps in the life span of an H2CO3, which ionizes into H and HCO3 (35), thereby ␣ ␤ osteoclast (43, 44). These processes require the v 3 in- providing the protons for the V-ATPase (27). Meanwhile, Ϫ Ϫ tegrin and the intracellular signal transducers c-src, Syk, basolateral exchange of HCO3 ions for Cl by anion ex- and proline-rich tyrosine kinase 2, as well as the microph- changer 2 (61–63) provides ClϪ ions required for the in- thalmia transcription factor (33, 45, 46), which appears to tense acidification occurring in the resorption lacuna. be an important regulator of osteoclastic gene transcrip- Interestingly, long bones differ from flat bones with tion (47–49). respect to the molecular nature of the acidification ma- The final step of osteoclastogenesis is the activation of chinery (62). resorption, a process that is characterized by the forma- Proteolysis of the type I collagen matrix in bones is tion of a ruffled border that is an intensely convoluted mainly mediated by the cysteine proteinase, cathepsin K. membrane present inside the sealing zone (43, 44). The This enzyme is active at low pH in the resorption lacuna formation of the ruffled border is not well-characterized; (Fig. 2) (64–68). The neutral MMPs also appear to play a however, the signaling molecule Rab7 is required (50). minor role during organic matrix degradation; however, Bone resorption takes place at the ruffled border local- the exact role of MMPs is still being investigated (69) and ized at the apical side of the osteoclasts, and it can be is highly dependent on the bone type (38, 70, 71). The divided into two processes, namely acid secretion and pro- resorbed material is removed from the resorption pit by Endocrine Reviews, February 2011, 32(1):31–63 edrv.endojournals.org 35

FIG. 2. Top, Schematic illustration of the differences between acid secretion and proteolysis during osteoclastic bone resorption, illustrating that the collagen matrix is removed by proteolysis after acidification. Bottom, Mutations/knockout in genes/proteins involved in bone resorption, phenotypes, and effect on osteoclasts. Ae2, Anion exchanger 2; a3 V-ATPase, a3 subunit of the osteoclast-specific V-ATPase. ClC-7, chloride channel 7; OSTM1, osteopetrosis associated transmembrane protein 1; CA2, carbonic anhydrase 2; MMP-9, matrix metalloproteinase 9. uptake and transcytosis through the osteoclast (72, 73). IV. Osteoclast Subtypes in After completing resorption, osteoclasts either undergo Physiological Situations apoptosis or perform a further round of resorption (44) Osteoclast activities are essential for development, as well (Table 1). as remodeling of bone in response to aging and stress (6, In summary, the osteoclasts are highly specialized for 14, 15). Under normal physiological circumstances, the both dissolution of the inorganic matrix and degradation osteoclasts can be categorized into subgroups depending of the organic matrix of the bones. These highly polarized on the matrix on which they are positioned, the time of cells are characterized by a unique set of membrane-bound day, and the type of remodeling in which they participate. molecules that ensure an efficient resorption of bone and These different groups of osteoclasts have provided key other mineralized tissues. This complex machinery may be information on skeletal maintenance. affected by a range of important parameters in physiology and pathology, and importantly in drug-induced situa- A. Endochondral vs. intramembranous bone osteoclasts tions that are important to identify and advance osteoclast Anatomically, two types of bones are present in the research and biology. body, the long bones (e.g., the femur and tibia) and the 36 Henriksen et al. Osteoclast Subtypes Endocrine Reviews, February 2011, 32(1):31–63 flat bones (e.g., the calvarium), with the main difference chondroclasts may also play a role in both rheumatoid between these two types of bone being their develop- arthritis (RA) and osteoarthritis (83–85). The term chon- ment (74). Studies also indicate that osteoclasts on these droclast derives from the localization of these cells on cal- two types of bones are functionally different with re- cified cartilage as seen in the expanding growth plates spect to both acid secretion and proteases involved in during endochondral ossification (80, 86). For their for- degradation (2). mation, these cells are dependent on the presence of Evidence that differences between resorption of flat and macrophage-colony stimulating factor and RANKL, as long bone exist was presented in 1999 (4). However, in- are bone-resorbing osteoclasts (87–90). dications that even the acidification process is different Most of the evidence for the functionality of chondro- have been published only recently (62). Data from mice clasts is derived from studies of the longitudinal growth of deficient in the bicarbonate-anion exchanger Ae2 (Slc4a2) long bones, i.e., metatarsals and tibias isolated from have shown that it is essential for bone resorption in long mouse embryos (38, 80, 91–93). First, bone/cartilage re- bones (61, 63), whereas it is not involved in bone resorp- sorption in these models is still dependent on acid secretion tion in calvariae (62), showing that distinct acid transport as evidenced by mice with mutations in the acid secretion mechanisms are present in different subsets of osteoclasts. process, i.e., ClC-7-deficient and a3 V-ATPase-deficient With respect to acid secretion into the resorption lacunae, mice, as well as mice unable to form sealing zones, i.e., it is presently not known whether any differences exist, c-src-deficient mice, in which the massive bones mainly although the absence of calvarial thickening in patients consist of calcified cartilage due to the defective resorption with defective ClC-7 strongly suggests that ClC-7 is not process (56, 94, 95). Similar findings have been noted in involved in resorption of the flat bones (75). the corresponding human disease(s) (27, 96, 97). Interest- Extensive research into the proteolytic processes in- volved in resorption of flat and long bones clearly dem- ingly, the ruffled border is less prominent in the chondro- onstrates that the proteolytic processes involved in deg- clasts than osteoclasts, potentially suggesting that lower radation of these two types of bone matrix are distinct (4, levels of acid secretion are required for dissolution of this 71). Osteoclasts in flat bones preferentially appear to en- matrix (98, 99). The main difference between chondro- gage in MMP-mediated bone resorption, although cathep- clasts and osteoclasts is in the profiles of enzymes neces- sin L seems to be involved, too. Osteoclasts in long bones sary for tissue degradation. Resorption by chondroclasts primarily depend on cysteine proteinases, in particular ca- does not appear to depend as much on cathepsin K as does thepsin K (4, 71). TRACP also appears to be involved in resorption by osteoclasts. Cathepsin K-deficient mice bone resorption, and more so in calvarial bone (76–78). show no evidence of calcified cartilage in the marrow cav- When osteoclasts generated from human peripheral blood ity of long bones, indicating that the removal of calcified are seeded on cortical bone, they primarily depend on ca- cartilage during endochondral ossification occurs, al- thepsin K, whereas when cathepsin K activity is blocked, though there are indications that the process is delayed there is some compensatory bone resorption mediated by (65, 100). Of importance is the observation of a massive MMPs (69). How these osteoclasts behave on bone sub- compensation by MMPs in the absence of cathepsin K, strates other than cortical bone is presently not known which obscures the interpretation of data from cathepsin (Table 1). K-deficient systems (69, 70, 101–103). Finally, one study Data suggesting that the bone matrix could play a role has indicated that TRACP is activated and secreted at the in the control of osteoclastic activities were presented in a ruffled border in cathepsin K-deficient osteoclasts, and study showing compositional differences between long only in cells that play a role in the removal of calcified bone and flat bone matrices, including differences in the cartilage (104) (Table 1). presence of putative cysteine proteinase inhibitors (79). Although chondroclasts are involved in the degrada- The functional significance of these data still remains to be tion of a different matrix than osteoclasts, an interesting fully elucidated, although they clearly illustrate the im- observation is that bone formation is tightly coupled to portance of understanding how a given context affects the resorption of the mineralized matrix, as has clearly been osteoclasts. demonstrated in studies of endochondral ossification (86). B. Chondroclasts It is more likely that bone formation is coupled to chon- It has long been discussed whether chondroclasts are a droclast numbers because release of molecules from deg- “real” cell type or whether they simply are osteoclasts that radation of cartilage, which in composition is far from reside on cartilage instead of bone (77, 80–82). Chon- bone, would be expected to be different from molecules droclasts are mainly important in endochondral bone de- released during bone resorption; however, this has never velopment, and in addition there is some evidence that been studied in detail. Endocrine Reviews, February 2011, 32(1):31–63 edrv.endojournals.org 37

FIG. 3. Schematic illustration of stochastic vs. targeted remodeling. The figure illustrates the local nature of targeted remodeling, which is activated at specific sites after the formation of microcracks and leads to removal of the microcrack and restoration of the damaged bone. Stochastic remodeling, on the other hand, is of a systemic nature and is activated by low calcium levels in the circulation leading to PTH release. Other hormones, such as vitamin D3 (VitD3), and potentially calcitonin also play roles in stochastic remodeling. It appears that there are two levels of calcium homeostasis—one mediated by osteocytes independent of osteoclasts, and one including the osteoclasts—although the balance between these two ways of releasing calcium still remains to be fully understood.

C. Osteoclasts involved in targeted and ized by high numbers of apoptotic osteocytes, are prefer- stochastic remodeling entially and rapidly degraded by osteoclasts. This further Two different modes of remodeling have been pro- supports the possibility that changes in the bone matrix posed: targeted and stochastic. Targeted bone remodeling and the balance between live and dead osteocytes deter- takes place at specific sites, whereas stochastic remodeling mine which areas will be remodeled (108). Finally, a recent occurs more randomly (19, 105, 106). The first type is study demonstrated that targeted ablation of osteocytes primarily performed to replace microdamaged bone and led to a dramatic increase in osteoclast activity (109) (Ta- thus to maintain the load-bearing capacity of the skeleton. ble 1). These data indicate that death of osteocytes is a key The second type of remodeling appears random with re- point in induction of osteoclast activity (Fig. 3). spect to localization, although it may be involved in main- Taken together with the finding that bones of different taining integrity of the bones, independent of damage. age lead to different levels of osteoclastogenesis (7), it ap- This process is hormonally regulated (105, 107). pears that osteoclast functionality is at least partially con- The balance between these two modes of bone remod- trolled by osteocyte-derived molecules, which are seques- eling has not been fully elucidated yet, but studies in dogs indicate that approximately 30% of all remodeling is tar- tered in the bone matrix. geted and the remaining 70% is stochastic (105). With Stochastic remodeling, while occurring at random sites, respect to the osteoclasts mediating these two types of is centrally regulated by hormones such as PTH, vitamin remodeling, most studies have focused on how targeted D3, and potentially calcitonin, and its main role is the remodeling is controlled. regulation of calcium homeostasis (110–112). It has even It appears that the bone matrix contains signals regu- been questioned to what extent this process depends on the lating the activity of the osteoclasts (108). A recent study presence of osteoclasts because patients with nonfunc- demonstrated that aged bones were more readily resorbed tional osteoclasts have normal calcium homeostasis (110– than young bones, thus supporting the hypothesis that the 112). Yet, when they are calcium-deprived, osteopetrotic bone matrix composition influences remodeling rates (7). patients fail to correct their calcium levels, indicating that Furthermore, areas of microdamage, which are character- osteoclasts, which are either absent or nonfunctional in 38 Henriksen et al. Osteoclast Subtypes Endocrine Reviews, February 2011, 32(1):31–63 these patients, do play a role in calcium homeostasis, and number and activity (32) (Table 1). An interesting aspect of thus stochastic remodeling (Fig. 3) (113). this is that targeting nocturnal resorptive activity appears to In summary, targeted remodeling is beginning to be lead to inhibition of bone resorption, whereas not attenuat- understood in detail, and it is a tightly regulated and cou- ing bone formation (125–127), thereby highlighting an in- pled process involving osteocytes, osteoclasts, and cells of teresting prospect of reducing bone resorption in a specific, the osteoblast lineage. On the other hand, stochastic re- nocturnal manner. modeling and the role it plays in calcium homeostasis are In summary, studies of osteoclasts under different phys- still not very well understood, although there are indica- iological conditions, such as those listed above, have high- tions that there is a level of regulation by the activity of lighted the heterogeneity of these cells. Furthermore, these osteoclasts. studies highlighted the importance of the balance between bone resorption and bone formation, a tightly regulated D. Trabecular and cortical osteoclasts phenomenon that rarely is disturbed under physiological Bone remodeling does not occur with the same fre- conditions. Finally, how the heterogeneity of the oste- quency in cortical and trabecular bone. Every year, 25% oclasts affects bone formation is presently not well under- of the trabecular bone matrix, but only 4% of the cortical stood, but a further understanding of this process could matrix, is remodeled (114). Interestingly, most in vitro help optimal treatment of diseases involving alterations in osteoclast experiments are based on cortical bone (or den- bone remodeling. tine) substrates, which are either slowly remodeled or not remodeled at all (7, 102, 114–116). Studies have shown that bones endogenously contain signals regulating osteo- V. Osteoclast Subtypes in clastogenesis and resorption and that these signals appear Pathological Situations to be related to the age of the bone (7, 117) (Table 1). Thus, an interesting question is whether osteoclasts themselves Changes in osteoclast activity and number have been de- are indeed different when derived from different matrices tected in several diseases, ranging from illnesses involv- or whether the difference is matrix related. Furthermore, ing excessive bone resorption, such as osteoporosis and systemic regulation is likely to be involved in controlling Paget’s disease; to those involving secondary activation of which bones are resorbed to some extent. osteoclasts, such as osteolytic metastases and RA; to dis- These data also correlate with evidence indicating that eases involving defective osteoclast differentiation and/or remodeling of different bone compartments can be either function, such as osteopetrosis. These different types of primarily targeted, such as in the cortex, or primarily sto- diseases have shed important light on osteoclastic function chastic, as seen in some parts of trabecular bone (106). A with respect to obtaining the right type of treatment. They further understanding of this could provide directions for have also shed light on a very central aspect in bone biol- the development of novel drugs producing optimal benefit ogy, the coupling principle. The coupling principle de- at the sites where it is most needed, i.e., leading to a better scribes the phenomenon that bone formation follows reduction than that presently obtained. resorption, which leads to a complete restoration of the bone removed during bone resorption (17).

E. Diurnal variation in osteoclasts or osteoclast activity? Bone resorption markers measured in serum may be A. Osteoporotic osteoclasts interpreted as indicating the net result of all osteoclast sub- 1. Changes in osteoclastogenic potential in osteoporosis types and activity levels at one particular time. A wide range An important aspect of osteoporosis is whether the of factors, known and unknown, may influence the inter- number of osteoclast precursors in the circulation in- pretation (15). Diurnal variation is a well-established and creases, and, if so, whether the osteoclastogenic potential important parameter of bone turnover. Postprandially, bone of these cells is increased. Eghbali-Fatourechi et al. (128) resorption decreases by approximately 50% compared with showed that the overall number of cells expressing that of fasting individuals, but during the night, bone resorp- RANKL is increased in postmenopausal women com- tion increases to an equally large degree (118–120). Several pared with premenopausal or estrogen-treated women, investigations have demonstrated that the circadian varia- clearly indicating that the bone marrow microenviron- tion in bone resorption is induced in part by food intake ment, including stromal, T, and B cells, changes in a proos- (121–123), which, at least partially, involves the peptide hor- teoclastic direction when estrogen is reduced. These data mone glucagon-like peptide (GLP) 2 (124). Interestingly, the were supported by a recent study from the same authors osteoclast number does not appear to depend on the time of showing that bone marrow cells isolated from estrogen- day, further emphasizing differences between osteoclast treated or control postmenopausal women displayed Endocrine Reviews, February 2011, 32(1):31–63 edrv.endojournals.org 39 reduced osteoclastogenic potential (129) (Table 1). Es- of osteoclastogenesis (137). Again, there appear to be dif- trogen was shown to have a dual mode of action—the ferent osteoclastic subtypes, which also appear to be rel- first leading to overall lower RANKL expression by evant in the context of bone loss rates in different bone bone marrow cells, and the second reducing the oste- compartments during osteoporosis (114). As indicated oclastogenic response to RANKL (130). Interestingly, above, an important point is the difference between gen- aging of mice was also shown to increase the osteoclas- ders (139). In mice, the gender-based difference between togenic potential of bone marrow cells, both by up- cortical apposition and endocortical resorption that be- regulation of RANKL production and by increasing comes more apparent with increasing age might be ex- precursor sensitivity to RANKL (131). plained by differences in ER expression (142). With re- In vitro studies of the changes in cellular activity of spect to changes in the osteoclasts after menopause, a osteoclasts from osteoporosis patients are limited, but couple of studies have clearly demonstrated that bone re- these have indicated both an accelerated osteoclastogen- sorption, as well as bone formation, increases in women esis and resorption (132, 133). Furthermore, a key cell in after menopause (143–145), and these changes become the up-regulation of osteoclastogenesis appears to be the more explicit in high- and low-turnover patients (145). T cell, which responds to lowered estrogen levels by in- Although bone formation increases as a function of the creasing RANKL production (134). increased resorption, it does not match bone resorption, The main issue with all the studies of osteoporotic os- thereby illustrating the importance of understanding the teoclasts and their precursors is the use of mixed cell pop- interplay between osteoclasts and osteoblasts in detail. ulations, which clouds the interpretation of the results, and therefore these aspects of osteoclastic function still B. Changes in osteoclast activities with increasing bone require further investigation. Furthermore, with the recent matrix age publication of the possibility of assessing the “anabolic Numerous studies have investigated the control of os- potential” of osteoclasts (11, 13), it would be of interest to teoclast activity as a function of changes in biochemical investigate the anabolic capacity of osteoporotic oste- properties of the bone matrix. Aging leads to accumula- oclasts and thus shed light on the imbalance between bone tion of different biochemical modifications of the bone resorption and bone formation in osteoporosis. matrix, such as advanced glycation end-products (AGEs), 2. A direct role for sex steroids on osteoclasts homocysteine, increased calcium concentration, as well as The role of estrogen on cells belonging to the osteoclas- some modifications of the collagen matrix (146). tic lineage has been studied extensively, with several find- Recent studies have indicated that these modifications ings indicating that estrogen suppresses osteoclastogenesis of the bone matrix itself actually modulate the activity of but not the resorptive activity of mature osteoclasts (135– the osteoclasts to a certain extent (7, 117). Homocysteine, 137). Androgens, such as dihydrotestosterone, exhibit which accumulates in bone and in circulation with age, similar effects to estrogen on osteoclasts in vitro, although was shown to activate osteoclastogenesis and bone resorp- this has not been studied in great detail (138). Finally, a tion (147) (Table 1). AGEs are modifications of proteins recent study using mice deficient for the estrogen receptor that accumulate in various tissues with age, and they have (ER)-␣, specifically in mature osteoclasts, showed bone been implicated in the pathology of osteoporosis (146, loss in female, but not male, mice (139). This demon- 148). Some evidence indicating a direct regulation of os- strated that estrogen likely plays a direct role in bone re- teoclast activity by AGEs has been published, but these sorption by even mature osteoclasts (139). Although the studies are contradictory. One study shows activation of authors used the cathepsin K promoter to ensure specific resorption by AGE-modified proteins (149), whereas the knock-down of the ER-␣ in osteoclasts, cathepsin K is also other study shows the opposite (117); however, quite dif- expressed in preosteoclasts, albeit to a lower extent (139). ferent techniques were used. More studies are needed to investigate the role of ER-␣ in Interestingly, AGEs are accumulated in diabetes, and mature osteoclasts specifically. Interestingly, osteoclasts they have been speculated to be involved in the increased in different bone sites preferentially express different ERs, fracture rates observed in patients with this disease (150, with cortical osteoclasts mainly expressing ER-␣ and 151). Another intriguing finding is the induction of trabecular osteoclasts mainly expressing ER-␤ (140), apoptosis in osteoblasts by AGEs (152), which potentially whereas from a functional point of view ER-␣ appears to could play a role in the imbalance between osteoclast and be more relevant for trabecular, not cortical, bone (141). osteoblast function during osteoporosis and aging. These Furthermore, the expression pattern also differs between findings are all preliminary in nature, and they await con- mature and differentiating osteoclasts; ER-␣ is mainly ex- firmation from independent research groups. However, pressed in immature cells, and ER-␤ is present at all stages once again they illustrate the heterogeneity of osteoclasts, 40 Henriksen et al. Osteoclast Subtypes Endocrine Reviews, February 2011, 32(1):31–63 in this case as a function of matrix age, and the importance that osteopetrosis was due to nonfunctional osteoclasts or of understanding this phenomenon. the absence of osteoclasts, respectively (162, 163). Inter- estingly, these two groups of mice have opposing pheno- C. Osteoclast-rich osteopetrosis types with respect to bone formation. The osteoclast-rich The most frequently occurring forms of osteopetrosis c-src knockouts have increased bone formation (164), and are those caused by mutations in either the a3 subunit of the osteoclast-poor c-fos knockouts have decreased bone the V-ATPase, ClC-7, or osteopetrosis-associated trans- formation (165). The anabolic effects of PTH are present Ϫ Ϫ Ϫ Ϫ membrane protein 1. Osteoclasts from patients with mu- in the c-src / mice but are blunted in the c-fos / mice tations in these genes or proteins and from knockout mice (166), indicating that osteoclasts are central for bone for- have been studied quite extensively (8, 39, 56, 58, 95, mation (Table 1). 153–156). Two recent studies identified mutations in the genes for Microscopic analyses of cells from patients with defec- RANK and RANKL as the causes of osteopetrosis in a tive acid secretion by osteoclasts indicated defective ruf- novel group of patients (28, 29). No indications of osteo- fled border formation, but also accumulation of material clasts were found in these patients (28, 29), which is con- in vesicles, indicating hampered transcytosis (157). Apart sistent with previous observations in mice deficient in both from confirming the defective acid secretion and thereby RANKL and RANK (87, 90). Patients with mutations in bone resorption, when either ClC-7 or the a3 subunit is either RANKL or RANK have a pronounced osteopetrotic mutated (8, 39, 56, 58, 95, 153, 154), these studies also phenotype and classical histological hallmarks of osteo- shed light on important aspects of bone remodeling. petrosis including unresorbed primary spongiosa. How- In vitro studies indicate that osteoclasts with impaired ever, although limited data have been published, the os- acid secretion have higher survival rates than cells with teopetrotic phenotype appears to be less severe than the normal secretion, due to the reduced release of proapop- one observed in the osteoclast-rich forms (1). Thus, muta- totic signals during resorption (159). This observation tions within the RANK/RANKL/osteoprotegerin (OPG) sys- correlates well with the high numbers of osteoclasts ob- tem can lead to osteoclast-poor osteopetrosis with low bone served in vivo in this group of patients, as well as with formation in mice and men. findings in mice with attenuated acidification of the re- Interestingly, alterations in osteoblast function, such as sorption lacunae (95, 97) (Table 1). Furthermore, signif- changes in the production of RANKL and OPG, may have icantly increased resorbed areas are seen during impaired the same effect. Stabilizing osteoblastic ␤-catenin in trans- acid secretion, but the resorption pits are shallow, indi- genic mice, thus mimicking constitutive activation of the cating a disturbed activity of the osteoclasts (97). More importantly, these studies highlighted that bone forma- canonical Wnt signaling pathway, was followed by an tion in these patients is ongoing—a process that appears to up-regulation of OPG in relation to RANKL (31, 167). As be correlated to the increased number of osteoclasts rather expected, the mice developed osteoclast-poor osteopetro- than bone resorption (96, 154, 160, 161). These findings sis with failure of tooth eruption, a classical phenomenon contrast with the classical perception that bone formation in murine osteopetrosis. Mutations within LRP5 related always follows bone resorption in a tightly coordinated to the Wnt signaling pathway have underscored the fun- manner and illustrate the importance of the actual pres- damental importance of this pathway for regulation of ence of osteoclasts to maintain bone formation. bone mass. The osteoporosis pseudoglioma syndrome was found to be caused by loss of function mutations in the gene for LRP5 (168). In contrast, mutations affecting the D. Osteoclast-poor osteopetrosis Several murine forms of osteoclast-poor osteopetrosis first propeller of the coreceptor, presumed to be followed have been described in the literature (1, 42). In general, by chronic activation of the Wnt pathway, were found in whereas the mutations express a pronounced osteope- various forms of monogenic human osteosclerotic pheno- trotic phenotype and few or no osteoclasts are present, the types (169). Among these, autosomal dominant osteope- phenotypes are less severe than the phenotypes of the dif- trosis type 1 has been well characterized clinically, bio- ferent osteoclast-rich osteopetrotic mutations (1). These chemically, histomorphometrically, and biomechanically data strongly suggest that the osteoclasts are indeed in- (75). Autosomal dominant osteopetrosis type 1 is an os- volved in the production of anabolic signals for bone for- teoclast-poor osteopetrotic phenotype with increased bio- mation (3, 15). mechanical competence and no low-energy fractures. Studies of mice deficient in c-src and c-fos, a key mol- Osteoclast profiles are markedly decreased (97), bone ecule involved in ruffled border formation and a key signal formation seems to be normal, and OPG levels in the cir- transducer for osteoclastogenesis, clearly demonstrated culation increased (170). However, when investigating os- Endocrine Reviews, February 2011, 32(1):31–63 edrv.endojournals.org 41 teoclasts ex vivo from these patients, they express normal F. Other diseases characterized by increased bone resorptive capacity (30). osteoclast activity In summary, osteoclast-poor osteopetrosis can arise in Apart from hypogonadal osteoporosis, several diseases murine mutations/transgenics or humans when the OPG/ are characterized by accelerated osteoclastogenesis and RANKL/RANK system is affected directly or indirectly. function. Although the etiology of these diseases is differ- These findings underscore this cytokine system as a key ent, there are interesting overlaps and discrepancies that regulator of osteoclastogenesis. Moreover, the pheno- provide highly useful information about osteoclastic func- types seem to be less affected than the osteoclast-rich tion and secondary effects on bone formation under dif- forms, the reason for which is so far unresolved, although ferent circumstances (182). there are indications that reductions in bone formation are involved (1, 28, 29). 1. Pagetic osteoclasts Paget’s disease is a late-onset disease that is quite com- E. Pycnodysostotic osteoclasts mon in the elderly Caucasian population, where it affects An interesting subtype of osteoclasts with defective approximately 3% of individuals (182). The disease is bone resorption is observed in patients with pycnodysos- characterized by focal increases in osteoclast numbers, tosis. Pycnodysostosis is caused by loss of function or loss nuclearity, and size, which leads to localized bone destruc- of expression mutations in the cysteine proteinase cathep- tion, although surrounding osteoblasts also are activated sin K, which in humans causes dwarfism and poor bone (183) (Table 1). The identified causes of the disease in- quality due to defective remodeling of the bones (36, 171– clude mutations in four different genes, TNFRSF11A, 173). Few studies examining the phenotype of pycnodys- TNFRSF11B, VCP, and SQSTM1 (182, 184–186). These ostotic osteoclasts have been published. Microscopic anal- genes encode RANK, OPG, p97, and p62, all of which are yses of the osteoclasts have shown significantly increased involved in the regulation of osteoclastogenesis. The mu- amounts of demineralized collagen matrix in the resorp- tations all result in different subtypes of Paget’s (182, tion pit, but also inside the osteoclasts, indicating dis- 184–186). These mutations render the osteoclast precur- turbed resorption and trafficking of resorbed components sors more sensitive to RANKL stimulation, resulting in a (174, 175). A study of biochemical markers of bone turn- higher number of osteoclasts, and potentially also explain- over showed that C-terminal crosslinked telopeptide of ing the presence of giant osteoclasts (185, 187, 188). In- type I collagen (CTX-I) release was reduced, whereas pro- terestingly, a recent study in mice indicated that the most duction of the MMP-generated type I collagen fragment common mutation in p62 does not make the osteoclasts carboxyterminal telopeptide of type I collagen (ICTP) was Pagetic alone, although it sensitizes them to other yet-to- increased (67) (Table 1). Several studies of cathepsin K- be-described causes of Paget’s (189, 190). deficient mice have been published, and whereas they con- In Paget’s patients, biochemical markers of both bone firm that cathepsin K is essential for degradation of the resorption and bone formation are increased, showing an organic matrix in bone (65, 66, 176), there are also several overall increase in bone turnover at the affected sites. differences between the human and mouse phenotypes However, bone resorption clearly exceeds bone formation (103). Furthermore, in cathepsin K-deficient mice, bone (182). Whether the osteoclasts in Paget’s behave differ- formation parameters are highly increased (100). These ently from those in healthy individuals during bone re- findings have not been replicated in pycnodysostosis pa- sorption is presently unknown. In particular, it is not tients in whom the bone matrix is disordered (177), and a known whether osteoclasts in Paget’s require acidification clinical case study indicated that anabolic response to PTH to resorb bone, or whether cathepsin K is the main pro- was absent (174). Two recently published clinical studies tease, although answers to these questions might be of have shown that whereas bone resorption markers are value in the development of new therapies for Paget’s. strongly reduced, bone formation is also suppressed in Furthermore, an explanation for the localized nature of women treated with the cathepsin K inhibitor odanacatib Pagetic lesions has still not been found. Even under the (178). In a monkey study monitoring bone formation by extreme circumstances seen in Pagetic lesions, bone for- histomorphometry reductions in bone formation, rates mation is coupled to osteoclastic parameters, although were shown in the trabecular bone compartment, whereas whether this is due to increased osteoclast numbers or bone formation was increased in the cortical compartment activities is not known. Moreover, in this case a treatment (179–181). type eliminating the activity of both types of cells is most In conclusion, cathepsin K mediates cleavage of type I likely to be preferred because the increase in bone forma- collagen in the resorption lacunae, but its secondary ef- tion occurring is part of the pathology, and likely will fects on bone formation are bone type-dependent and still provide no benefit for the bones if maintained. Thus, need to be investigated further. bisphosphonate, which strongly attenuated overall bone 42 Henriksen et al. Osteoclast Subtypes Endocrine Reviews, February 2011, 32(1):31–63 turnover, appears to be highly relevant in the context of 3. Arthritic osteoclasts Paget’s disease (191). Later stages of RA are characterized by massive bone destruction caused by osteoclasts (212, 213). However, 2. Osteolytic osteoclasts there are several indications that these osteoclasts are not Several forms of cancer can metastasize to bone and classical bone-resorbing osteoclasts but include cells that form osteolytic metastases (192–196). Once the cancer degrade calcified cartilage (83, 214) (Table 1). Several has reached the bone, tumor and bone interact in a vicious studies have indicated that TNF-␣ at least partially drives cycle in which tumor-secreted factors, such as PTHrP, osteoclastogenesis in RA (215), as exemplified by mice stimulate bone cells, which in turn release growth factors overexpressing human TNF-␣ with massive joint destruc- and cytokines that promote further tumor cell growth tion including bone erosion (216). Furthermore, TNF-␣- (192, 197). The activation of osteoclastogenesis induced neutralizing antibodies, such as infliximab, or soluble by tumor cells has been shown to involve a switch in the TNF-␣ receptor antagonists, such as etanercept, provide RANKL/OPG ratio favoring osteoclastogenesis and acti- amelioration of RA in humans (217, 218). Apart from vation, leading to release of the tumorigenic factor TGF-␤, TNF-␣, RANKL is, not surprisingly, a crucial factor in and thereby inducing the vicious cycle (198). As a function osteoclastogenesis during RA (87), and mice deficient in of the increased numbers of osteoclasts and accelerated RANKL are protected against bone, but not cartilage, ero- bone resorption, a marked up-regulation of osteoblast ac- sion (219). Treatment with OPG of mice with collagen- tivities is also observed (199, 200). induced arthritis also leads to amelioration of bone de- The activity of osteoclasts in metastases has been mon- struction, while having a markedly lower effect on itored closely using biochemical markers of bone turnover cartilage degradation (220). In addition, a study in which (199, 200), and these studies have indicated that bone TNF-␣ overexpressing mice were crossed with mice defi- resorption by tumor-induced osteoclasts to some extent cient in c-fos (i.e., deficient in osteoclasts), showed no bone depends on MMP activity, rather than cathepsin K, be- cause the type I collagen fragment ICTP is released in high destruction but clear evidence of cartilage destruction amounts (199, 201) (Table 1). Animal models of breast (221). Furthermore, human clinical trials using deno- cancer bone metastases are to some extent sensitive to sumab have demonstrated that the RANKL/RANK axis is inhibitors of both cathepsin K and MMPs (101, 202–204); a key player in RA and that inhibition of RANKL signaling however, clinical data for MMP inhibitors have been dis- may provide a useful treatment option (222). Finally, ␣ ␤ appointing (205, 206). An interesting question is whether more recent evidence has indicated that IL-1 and IL-1 these agents, to be effective, have to inhibit MMPs before the both play a partial role in bone resorption and cartilage tumors actually metastasize. For cathepsin K inhibitors, the degradation (223). Anakinra, which is a soluble IL-1 data indicate a beneficial effect on the release of the bone receptor antagonist, is also used for treatment of RA, ␣ resorption marker N-terminal crosslinked peptide of type although it appears to be less effective than the TNF- I collagen, and an increase in ICTP levels (204). However, inhibitors (217). In addition, tocilizumab (anti-inter- further information is needed to draw reliable conclusions leukin-6 receptor inhibitor) has shown promise in pre- on the usefulness of cathepsin K inhibitors for metastatic venting RA progression through an effect including a bone disease. reduction in osteoclast numbers (158, 324). In contrast, treatments ablating both osteoclasts and Because osteoclasts play a significant role in RA, the increased osteoblast activity, such as denosumab and bisphosphonates appear to be an attractive treatment op- bisphosphonates, reduce the destructive capacity of the tion. Early evidence has indicated that zolendronate may metastasis and, importantly, the afflicted pain. How- be useful (224), although this has not been fully estab- ever, they do not appear to affect the cancer cells (192, lished yet (225). Furthermore, interpretation of the effects 207–210), although there are some indications that the of bisphosphonates in RA is often clouded by glucocorti- bisphosphonates affect the life span of the cancer cells coid treatment of the same patients because glucocorti- as well as reducing (211). coids are associated with rapid systemic bone loss, In summary, from a treatment point of view, there are independent of RA (226, 227). However, in both collagen- several similarities between Paget’s and osteolytic metas- induced arthritis in rats and in the TNF-␣ transgenic tases. The optimal approach appears to involve a strategy mouse model, zolendronate was effective in reducing both of reducing overall bone turnover toward the normal bone and cartilage destruction (228, 229). range, such as with the use of denosumab or bisphospho- The bone resorption process in RA is still not com- nates An intriguing possibility would be to target only the pletely understood despite several studies into the molec- areas undergoing destruction, but whether this is feasible ular mechanisms. The role of cathepsin K has been exten- is presently not known. sively studied, and the data are somewhat conflicting Endocrine Reviews, February 2011, 32(1):31–63 edrv.endojournals.org 43

(230–232). Overexpression of cathepsin K has been important secondary role in bone remodeling, which is of shown to accelerate joint destruction in mice (231), and importance when developing novel treatments for osteo- overexpression of cathepsin K has been observed in hu- porosis (15). Second, excessive and local activation of mans with RA (233, 234). However, studies in the TNF-␣ osteoclasts occurs in several diseases, and interestingly overexpression model crossed with cathepsin K-deficient the osteoclasts appear to switch subtype with respect to mice showed that cathepsin K plays only a marginal role their resorption machinery. These findings highlight the in bone resorption in RA (232), a finding supported by a importance of characterizing the function of osteoclasts case study showing severe arthritis in a pycnodysostotic under pathological circumstances to optimize treatment patient (235), although there are still controversies with strategies. respect to the role of cathepsin K in RA (236). Other cathepsins have not been explored in detail, and their expression patterns do not indicate a particular effect VI. Drug-Induced Osteoclast Subtypes on osteoclast function in RA (237). Under some circumstances, MMPs also play a role in A. Existing drugs bone resorption (2, 81). Studies showing that the MMP- Several antiresorptive drugs for the treatment of osteo- derived collagen type I fragment, ICTP, is increased in RA porosis, as well as glucocorticoids and PTH treatment, are could indicate that osteoclasts used MMPs to digest ma- known to alter osteoclasts in various ways. These drugs all trix under these circumstances (238, 239). Infliximab provide critical information on osteoclast function, and treatment has been shown to reduce ICTP levels, as well as furthermore, they have also played a great role in illus- osteoclast numbers (240), further indicating that oste- trating the interplay between osteoclasts and osteoblasts, oclasts utilize MMP-mediated bone degradation in RA. as will be described in the following section. However, a direct link between the production of ICTP and osteoclasts has not been demonstrated yet. 1. Bisphosphonates Whether acid secretion by osteoclasts is needed for Bisphosphonates have long been associated with induc- bone destruction in RA is also not clear. Because bone tion of apoptosis in osteoclasts, and the mechanism of destruction is likely to occur as a result of MMP activity, action underlying the apoptotic effect depends on whether the need for acidification may be reduced when compared or not the bisphosphonates contain nitrogen (242). Both with “classical” bone resorption (69), although this is still classes of bisphosphonates bind to the bone matrix and are not fully understood. Another possibility is that MMP- taken up by the osteoclast during bone resorption. The mediated collagen type I degradation is mediated by an- simple bisphosphonates are metabolized into toxic ATP other cell type, although this still remains to be clarified. analogs, thereby inducing osteoclast apoptosis in vitro A case study of arthritis in a case of autosomal dominant (242). The nitrogen-containing bisphosphonates exert osteopetrosis type II (241) showed a lack of bone degra- their function by inhibiting the mevalonate pathway, dation, whereas cartilage degradation was abundant, which leads to the generation of an ATP analog known to thereby mimicking the situation seen in osteoclast-defi- induce apoptosis in osteoclasts in vitro (242). The antire- cient systems (221) and indicating that bone resorption in sorptive potency of the nitrogen-containing bisphospho- RA depends fully on acid secretion. nates in vivo is controlled by mineral binding affinity and In summary, development of severe RA involves oste- by their ability to inhibit the mevalonate pathway (242). oclasts, and a reduction of bone resorption by these cells Although in vitro data clearly show that bisphospho- is desired. This may be obtained through inhibition of nates induce apoptosis, analyses of osteoclast numbers in inflammation and thereby bone and cartilage destruction, iliac crest biopsies failed to show a reduction in the number as seen with anti-TNF-␣ therapy. Alternatively, therapies of osteoclasts when patients were treated with bisphos- such as denosumab that target the osteoclasts directly may phonates (243–245). On the other hand, bisphosphonates also be useful, although these fail to eliminate inflamma- reduce systemic levels of TRACP 5b and cathepsin K, both tion and only partially prevent cartilage degradation markers of osteoclast number (32, 246–248), potentially (220). The optimal therapy could be a combination of indicating that osteoclasts undergo systemic apoptosis, antiinflammatory and antiosteoclastic measures, al- which correlates well with the expected effects of bisphos- though this is presently not known. phonates (242) (Table 1). Other studies have shown that In summary, studies of osteoclasts under pathological when bisphosphonate therapy continues for more than 1 circumstances have highlighted some important phenom- yr, the number of circulating osteoclast precursors is re- ena. First, osteoclasts themselves, not just their resorptive duced, and these reductions are speculated to be related to activity, mediate bone formation and therefore perform an reduced serum RANKL levels (249, 250). 44 Henriksen et al. Osteoclast Subtypes Endocrine Reviews, February 2011, 32(1):31–63

A recent study of biopsies from alendronate-treated pa- a. Osteonecrosis of the jaw (ONJ) and bisphosphonates. tients showed the presence of giant hypernucleated, de- Bisphosphonate therapy, especially in the case of malig- tached, and frequently apoptotic osteoclasts, and the num- nancy-induced bone loss, has been connected to the oc- ber of these abnormal osteoclasts correlated with the currence of ONJ, mainly due to the ability of bisphospho- cumulative dose of bisphosphonate (251). Although in- nates to strongly suppress bone turnover (260–262). teresting, the biological implications of this finding are not Although the probability of ONJ is very low for the dosing clear yet. regimens used for treatment of osteoporosis, there has still One potential explanation for the discrepancies in scor- been a lot of debate about whether ONJ is the result of the ing osteoclasts in the iliac crest biopsies is the very low massive suppression of bone turnover in the jaw (262). number of osteoclasts observed in general. Recent reports Interestingly, alveolar bone of the jaw is very similar to have also debated the clinical relevance of studying iliac bone matrix in the long bone, i.e., it contains the classical crest biopsies because they are from non-weight-bearing cell types as well as the lamellar structure (263). Further- bones and these are different from weight-bearing bones more, bone remodeling occurs normally in alveolar bone, (252–254), and in general more data are needed to draw although the rate of remodeling has been estimated to be a final conclusion on the osteoclastic response to up to 10-fold higher than the corresponding rate in long bisphosphonates. bones (263–266). In ONJ, the number of osteoclasts has On the other hand, the effect on reduction of bone re- been investigated, and it appears that the osteoclasts are sorption measured both by biochemical markers and by absent from the lesions (267, 268), although opposing evi- bone histomorphometry (activation frequency) confirms a dence also exists (269) and thus more studies are needed. potent reduction in bone resorption, and the level of It has been speculated that massive suppression of os- reduction is often down to the lower range of premeno- teoclast function, and thus bone turnover, in this high- pausal levels, although this depends heavily on the ef- turnover compartment is what causes ONJ to occur; how- ficacy of the individual bisphosphonate (242, 243, 245, ever, there are several other factors involved, such as tooth 255–258). extraction or infections, and the overall causality is still With respect to secondary effects on bone formation, not clear (262). One point of particular interest is measurement of biochemical markers of bone turnover whether this phenomenon is specific for bisphospho- nates or whether it will happen with other very potent shows a marked reduction in bone formation markers, and long-lived antiresorptives; however, this is pres- and the effects are maintained throughout the treatment ently not known. period, although this again is dependent on the individual bisphosphonate (242, 243, 245, 255–258). Biopsy studies 2. Selective estrogen receptor modulators/hormone have confirmed that bone formation is reduced when com- replacement therapy pared with placebo, and although the reduction in bone Because cessation of estrogen production is a major formation rates is dependent on the individual bisphos- cause of osteoporosis (3, 14) and both estrogen and phonate, the data indicate that bone formation is not com- SERMs are used for treatment of osteoporosis, several pletely suppressed but is reduced to the lower postmeno- studies have been conducted to clarify their effect on pausal levels (243, 245, 258). The FLEX study (Fracture osteoclasts. Intervention Trial Long-term Extension), although show- Estrogen has been shown to exert direct antiosteoclas- ing continued reductions in vertebral fractures, increase in tic effects at several stages of osteoclastic differentiation bone mineral density (BMD), and reduction of bone turn- and function, namely osteoclastogenesis, resorption, and over markers with alendronate, did not show a significant apoptosis. Direct inhibition of the formation of multinu- reduction in bone formation rates when comparing pa- cleated osteoclasts is thought to be caused by suppression tients stopping alendronate to patients continuing treat- of RANKL-induced c-Jun and basal c-Jun N-terminal ki- ment; however, the numbers of biopsies were low (259). nase activity in osteoclast precursor cells (135, 136). In All in all, there is no doubt about the fracture-prevent- nonpurified osteoclast-precursor systems, estrogen was ing effects of bisphosphonates; however, knowledge of the found to inhibit osteoclastic differentiation in a human ␣ ␤ effect of bisphosphonates on osteoclasts in vivo is quite system (270), possibly via down-regulation of the v 3 limited. Apoptosis of the osteoclasts most likely explains integrin (271). Two studies of estrogen have been con- the reduction in bone resorption. Furthermore, although ducted using CD14ϩ osteoclast precursors. As mentioned the extent of the secondary reduction in bone formation is earlier, one study showed significant inhibition of oste- still discussed, it appears to be clinically relevant, and it oclastogenesis (137), whereas the other showed no direct most likely is the explanation for the attenuation of the effect on osteoclast precursors (272). To date, there is no BMD increase seen after the first year of treatment. explanation for this discrepancy. Endocrine Reviews, February 2011, 32(1):31–63 edrv.endojournals.org 45

Studies of the effects of SERMs on osteoclasts have vertebral fractures, most likely due to low exposure, the shown that tamoxifen inhibits osteoclastogenesis directly, clinical usefulness is limited (122). Recent studies have whereas raloxifene and ospemifene only inhibited osteo- indicated that a recently developed oral formulation of clasts through up-regulation of the expression of OPG by salmon calcitonin will lead to improved efficacy because it osteoblasts (273). Although early studies showed an effect has been optimized with respect to pharmacokinetic and of raloxifene on osteoclastogenesis, these were conducted pharmacodynamic properties. This has led to a 10-fold using mixed cell populations and therefore most likely higher exposure and thereby a greater reduction in bone reflect the increase in OPG (274). resorption parameters. Thus, this agent will most likely Mature osteoclasts have also been shown to respond provide improved efficacy in preventing fractures (291), directly to estrogen (275, 276). These studies showed that and although it remains to be proven in long-term clinical both the activity and the production of the lysosomal en- trials, the phase II data are promising (127). zymes are down-regulated by estrogen (277, 278), possi- The mode of action of oral calcitonin is a transient bly explaining the reduction in resorption by the down- suppression of the nocturnal rise in bone resorption ob- regulation of cathepsin K and TRACP (36, 66, 77, 279) tained by giving the treatment at the right time of day—in (Table 1). the evening (292), which results in a reduction in bone In summary, in vitro data clearly demonstrate that es- resorption, but no effect or very modest secondary effects trogen and SERMs reduce osteoclast numbers via inhibi- on bone formation (127) (Table 1). These findings are tion of osteoclastogenesis, and potential effects on bone further supported by other clinical studies showing that resorption and apoptosis might add to the in vivo effect. calcitonin may inhibit bone resorption without affecting Although some studies of osteoclasts in patients treated bone formation, a finding observed independent of ad- with either HRT or SERMs have been conducted, the ef- ministration route (293–296). fects of both estrogen and SERMs on bone remodeling There are histological indications that calcitonin atten- indices based on histomorphometry are quite modest uates ruffled border formation by osteoclasts (296–298), (280–284). Overall, these studies show a reduction in ac- and this appears to be the mode of action underlying the tivation, frequency, and depth of resorption, as well as— antiresorptive effects of calcitonin in vivo, thereby elabo- where detectable—a small decrease in osteoclast numbers. rating on the previously described transient reduction in Reduced bone formation rates were also observed, con- bone resorption (292). firming the coupled nature of inhibition mediated by es- Studies of mice lacking the calcitonin receptor indicated trogen and SERMs (280–284). These data are corrobo- that bone formation was increased, and thus that calcito- rated by biochemical markers of bone turnover, which nin is a suppressor of bone formation (299, 300). These clearly demonstrated a coupled reduction in bone resorp- studies were conducted mainly in young mice. A recent tion and bone formation (32, 285–287), and furthermore study in mice deficient in the calcitonin receptor specifi- explain the plateau effect observed in BMD measurements cally in osteoclasts failed to reproduce this finding (301, after 1 yr (285). 302). However, considering the very modest, or nonex- In summary, many of the numerous studies of the in istent, suppression of bone formation in patients treated vitro mode of action of HRT and SERMs show a reduction with calcitonin, the mice data appear of low relevance in in osteoclastogenesis. In alignment, in vivo studies of these the clinical setting (127, 293–296). therapies on osteoclasts confirm that osteoclastogenesis is Further studies are needed to understand this potential lower than in the untreated population, and importantly, dissociation of bone resorption and bone formation. It these also confirm the secondary decrease in bone may be that this dissociation occurs because calcitonin formation. disappears quickly from the circulation and thus is a com- pletely reversible treatment (122). An interesting question 3. Calcitonin is whether calcitonin treatment may result in better bone Calcitonin is a natural peptide hormone produced by quality than potent antiresorptives due to the lack of effect parafollicular cells (C cells) of the thyroid gland. Calcito- on bone formation and the lower suppression of bone nin possesses potent antiresorptive effects (288), and bind- resorption, which is expected to lead to a slow, yet pro- ing of calcitonin to the calcitonin receptor on osteoclasts longed increase in BMD (6, 303). induces a rapid change in the cytoskeletal structure of the osteoclasts in vitro, which in turn leads to a reduction in 4. Parathyroid hormone bone resorption without inducing apoptosis of the cells Although PTH does not appear to affect osteoclasts (102, 289, 290). Calcitonin in either an injectable or a directly because these cells do not appear to express the nasal form has been approved for treatment of osteopo- PTH receptor, PTH nonetheless affects osteoclast func- rosis; however, because it only prevents about 35% of tion on many different levels (10). In vitro studies of the 46 Henriksen et al. Osteoclast Subtypes Endocrine Reviews, February 2011, 32(1):31–63 effects of PTH on osteoclasts all show that PTH induces Furthermore, strontium has also been shown to increase osteoclastogenesis and that induction of a transient OPG expression by osteoblasts (330). RANKL expression is essential for this effect (10). How- In summary, strontium ranelate is a very interesting ever, PTH has mainly been studied in relation to its pow- molecule with respect to effects on osteoclasts, and several erful anabolic effects on osteoblasts (10). Intermittent dos- lines of in vitro evidence indicate that it reduces osteoclast ing of PTH in human subjects results in a marked increase function (326). However, the relevance of the effect on in bone formation markers, and secondarily in activation osteoclasts is still debated, and thus the overall effects of of bone resorption through increased RANKL expression this “uncoupling” molecule are still not fully understood. (9, 304, 305). Bone histomorphometric and biochemical 6. Glucocorticoids marker studies confirm the increase in bone turnover Glucocorticoids are used to overcome inflammatory (306–308) (Table 1). conditions, such as inflammatory bowel diseases and RA The anabolic mode of action of PTH has been debated (331). Glucocorticoid use is associated with severe bone extensively. Studies show that PTH directly activates bone loss due to strongly attenuated bone formation (332). This formation by osteoblasts when given intermittently (309, attenuation of bone formation leads to a rapid accelera- 310). In mouse models that are either deficient in osteo- tion in the number of fractures in glucocorticoid-treated clasts or deficient in bone resorption, data suggest that the patients (332), especially in trabecular bone compart- anabolic effect of PTH is dependent on the presence of ments such as vertebrae (333). Glucocorticoid treatment is mature osteoclasts, but not on their activity (165, 166, the most common cause of secondary osteoporosis (333), 311). Furthermore, initial clinical trials combining alen- and thus patients on glucocorticoids are often treated with dronate and PTH showed that alendronate blunted the antiresorptives (334). anabolic effect of PTH (312, 313), and there were indi- In vivo, glucocorticoids inhibit osteoblastogenesis, the cations that even pretreatment with alendronate led to a generation of bone-forming osteoblasts, and promote ap- blunting of the PTH response (314). On the other hand, optosis of osteoblasts and osteocytes, which is consistent animal studies indicate that PTH can be combined with a with the well-known inhibition of bone formation (335). bisphosphonate (315, 316), but, as noted by Johnston et In contrast, the cellular effects of glucocorticoids on al. (315), there are marked differences in the doses of PTH osteoclasts are a subject of controversy. In vivo, the effects used in rodents and in humans. appear to fall into two categories, one being a short-lived Collectively, PTH exerts marked regulation of bone acceleration of osteoclastogenesis and bone resorption, turnover (15), including the activation of osteoclasts. The whereas the other is a reduction in osteoclast numbers, potential anabolic role of osteoclasts and, especially, how which is not well-characterized with respect to exposure to achieve the right osteoclast subtype are debated in- time to glucocorticoids (335–340) (Table 1). Interestingly, tensely. Future studies will most likely explain this com- a study by Kim et al. (340) showed that the detrimental plex interplay between bone cells and thus guide the right effect of glucocorticoids on bone formation was absent combination of PTH and antiresorptive. when the glucocorticoid receptor was ablated specifically in osteoclasts in mice. 5. Strontium ranelate In vitro studies of glucocorticoids are often conducted Strontium ranelate is approved for treatment of osteo- in the presence of contaminating cells, and because glu- porosis, albeit only in Europe, through its ability to reduce cocorticoid treatment also promotes RANKL and reduces fracture risk in patients (317–321). The mode of action has OPG expression in osteoblasts, it is unclear exactly to been studied extensively, and yet it is not fully clear exactly what extent they influence the osteoclasts (341, 342). Two how it works in vivo. Bone biopsies have been investi- recent studies showed that glucocorticoid treatment hy- gated, and these indicated small increases in bone forma- peractivated osteoclasts and thus suggests that glucocor- tion and mineralization rates but no changes in bone re- ticoids indeed have a direct effect on bone resorption (343, sorption or osteoclast parameters, thus indicating that 344). Yet some studies show the opposite (340). Overall, strontium ranelate stimulates novel bone formation (322). the results appear to be very context-dependent, illus- These data were supported by analysis of biochemical trating the complex nature of the biological effects of markers of bone turnover demonstrating increased bone glucocorticoids. formation (308, 323), while also showing a modest de- Measurements of biochemical markers of bone turn- crease in bone resorption markers (323, 325). over in human subjects on glucocorticoid therapy pro- In vitro studies support the hypothesis that strontium vided diverse results, which appeared to be dependent on ranelate has a dual effect, namely inhibition of bone re- the dose of glucocorticoid used (331, 345, 346). However, sorption while stimulating bone formation (326–329). biochemical marker data indicate that bone resorption Endocrine Reviews, February 2011, 32(1):31–63 edrv.endojournals.org 47 increases short term, whereas bone formation is attenu- resorption by osteoclasts (36, 65, 66). Studies conducted ated long term (345) in response to glucocorticoid ther- in pycnodysostosis patients before the final identification apy, which corresponds well to mouse studies (335–339). of cathepsin K showed massive accumulation of nondi- The short-term increase in bone resorption and long-term gested bone collagen fibers in the resorption pit below the suppression of bone formation are also observed with his- osteoclasts (175). These findings were matched by those tomorphometry in mice (347, 348). from investigations in cathepsin K-deficient mice (66), In summary, glucocorticoids exert detrimental effects demonstrating a critical role for cathepsin K in degrada- on bone, and whereas the effects on osteoclasts are not tion of the organic matrix. Further studies in cathepsin completely clear yet, further investigation of the effect on K-deficient systems have indicated that cells of the osteo- the coupling between osteoclasts and osteoblasts could blast lineage, namely bone-lining cells (68); cells of hema- explain the overall beneficial effect of antiresorptives on a topoietic origin (69); and a general up-regulation of the syndrome mediated primarily by suppressed bone forma- osteoclastic stimuli, osteoclast numbers, and proteases, tion (227, 349). These findings further highlight the im- especially RANKL and MMPs (103), are involved in com- portance of understanding the interplay between bone pensating for the lack of cathepsin K. Interestingly, a hall- cells to provide the optimal treatment. mark of the absence of cathepsin K function is the presence of the MMP-derived collagen fragment ICTP, which is B. Future treatments seen in pycnodysostosis patients, cathepsin K-deficient A series of interesting targets for osteoporosis treat- mice, and cell cultures (67, 69, 103, 356), strongly indi- ment are currently under investigation. The targets of cating a compensation by MMPs in the absence of cathep- these treatments to some extent employ novel modes sin K (Table 1). of action on osteoclasts. These novel modes of action are An interesting study by Fuller et al. (357) showed that of importance when investigating whether they may have inhibition of cathepsin K in cultured osteoclasts led to secondary effects on bone formation, and subsequently on augmented secretion of IGF-I. Furthermore, increased bone quality. numbers of osteoclasts, containing granules of matrix pro- teins, have been observed in monkey studies of cathepsin 1. Denosumab K inhibitors (175, 181), thus indicating the potential of Denosumab is a fully humanized monoclonal antibody this protease for anabolic stimulation of the osteoblasts. to RANKL; it has gone through a phase III fracture effi- Cathepsin K-deficient mice have been studied extensively, cacy trial in which it was shown to reduce fracture rates by and recent experiments indicate that bone formation in 68% in vertebrae and 40% in hip (351); and it was re- trabecular bone is increased after cathepsin K administra- cently accepted for treatment of severe osteoporosis in both the United States and Europe. tion and thus that bone resorption and bone formation are In line with in vitro studies of inhibition of RANKL not coupled (100, 176). However, clinical studies of ca- (210, 352), denosumab prevents osteoclastogenesis, thepsin K inhibitors, such as odanacatib, have shown that blocks bone resorption, and increases osteoclast apopto- whereas a robust reduction in CTX and N-terminal sis. It induces a massive reduction of osteoclasts in vivo crosslinked peptide of type I collagen occurred and no and, thereby, almost complete suppression of bone resorp- changes in TRACP 5b were observed, a significant de- tion in both humans and mice (352–354) (Table 1). De- crease in the bone formation marker pro-peptide of col- nosumab treatment also leads to a marked suppression of lagen type I and nonsignificant reductions in bone forma- bone formation markers in humans (353, 354), as well as tion rates by histomorphometry were seen (178). a marked suppression in bone formation rates measured Furthermore, a study of osteoclast morphology as a func- by histomorphometry in animal models (352, 355). Thus, tion of cathepsin K inhibition in humans indicated in- denosumab treatment is consistent with the classical per- creased size of the osteoclasts and the presence of large ception of coupling. TRACP-positive vacuoles, yet no increase in osteoclast A key point with respect to denosumab is whether the numbers (358). Studies in monkeys clearly demonstrated suppression is too severe and could lead to detrimental that bone formation in the trabecular compartments was effects on bone quality long term (6). However, as is the dose-dependent and significantly reduced by cathepsin K case with bisphosphonate treatment, this is not clear at inhibitors, whereas an induction of bone formation was present. observed at cortical sites (179, 180). Further studies are needed to clarify whether the osteoclasts in cathepsin K- 2. Cathepsin K inhibitors deficient situations indeed signal to the osteoblasts. An Cathepsin K is a critical protease for degradation of the indication came from a pycnodysostosis case study that type I collagen matrix in the resorption pits during bone showed no bone formation response to PTH (174), and 48 Henriksen et al. Osteoclast Subtypes Endocrine Reviews, February 2011, 32(1):31–63 thus indicated that secretion of the coupling signals may be and showing no inhibition of bone formation markers (8, attenuated at least in human systems. A possible expla- 364). Similar findings were published for an inhibitor of nation for the lack of secondary anabolic effects induced the V-ATPase (365). In a study of prosthetic implants by inhibition of cathepsin K is the presence of demineral- coated with bafilomycin, osteoclast numbers were ele- ized collagen fibers in the resorption pit, which are re- vated, and indications of increased bone formation were moved by bone-lining cells (68). Although it is not well observed (366). These studies were the first to provide understood how the presence of fibers and their subse- proof of concept that inhibition of acidification is a really quent removal affect osteoblasts, a study indicated that promising target for osteoporosis treatment. Most inter- RGD sequences, which are numerous in collagen, antag- estingly, bone formation levels, as measured by osteocal- onize osteoblast function (359). cin and by evaluation of the dynamic histomorphometry These findings again illustrate the importance of care- parameters mineral apposition rate and the mineralizing fully investigating the osteoclast subtype as a function of surface vs. bone surface, were not affected. These data cathepsin K inhibition to more accurately predict potential therefore suggest that inhibition of acidification of the secondary effects on bone formation. osteoclastic resorption lacunae results in an uncoupling of bone formation and bone resorption, thereby possibly im- 3. Glucagon-like peptide-2 proving the potential efficacy of the treatment. This is in GLP-2 is a 33-amino acid peptide. GLP-2 is created by contrast to other antiresorptive treatments where a sec- specific posttranslational proteolytic cleavage of proglu- ondary decrease in bone formation is observed (3, 367). cagon in a process that also liberates the related GLP-1 These data also indicate that the subtype of osteoclasts (124). GLP-2 is produced by the intestinal endocrine L cell obtained—nonresorbing yet alive—when targeting acid and by various neurons in the central nervous system secretion is active with respect to bone formation, and thus (124). Intestinal GLP-2 is cosecreted along with GLP-1 might possibly be combined with PTH treatment in the upon nutrient ingestion. future. GLP-2 has in clinical settings been demonstrated to in- Finally, other compounds that appear to modulate the hibit bone resorption (124–126) (Table 1). Reductions in activity of osteoclasts are in development for osteoporosis. bone resorption by exogenous GLP-2 require an intact These include calcilytics, PTHrP, and sclerostin, but their gastrointestinal tract (125, 361, 362). The decreased meal- effects on osteoclasts, which most likely are indirect, are induced inhibition of bone resorption in jejunostomy pa- not clear yet (368–370), and thus these will not be de- tients, who lack a GLP-2 response, supports the view that scribed further. GLP-2 plays a role in postprandial reduction in bone re- sorption (361, 362). GLP-2 has in addition been suggested to inhibit bone VII. The Bone Anabolic Effects of resorption without affecting bone formation (125), high- the Osteoclasts lighting this mode of inhibition of resorption for further Since the early discovery that osteoclast activities were investigation with respect to osteoclast subtypes. involved in regulation of bone formation during targeted remodeling (17, 18, 371, 372), a series of studies have 4. Acid secretion inhibitors investigated the nature of this process. Acid secretion by osteoclasts has been an interesting The early studies focused mainly on the release of mol- therapeutic target since the discovery that this process is ecules from the bone matrix during bone resorption and controlled by the a3 subunit of the V-ATPase and ClC-7, identified molecules such as IGF-I and TGF-␤ (357, 373, both of which are quite specific to osteoclasts (37, 39, 56). 374). However, with the recent discovery that mature os- Furthermore, in vitro studies of osteoclasts treated with teoclasts, not osteoclast precursors and not necessarily inhibitors of these ion transporters have shown that the bone resorption, are needed for stimulation of bone for- osteoclasts are unable to resorb bone and that they there- mation (8, 9), a series of studies have investigated this fore survive longer (8, 159, 363), thereby mimicking the phenomenon. elevated numbers of osteoclasts observed in patients with Zhao et al. (12) demonstrated that osteoclast-mediated mutations in the genes for a3 and ClC-7 (37, 97) (Table 1). expression of EphrinB2 and osteoblast-mediated expres- In aged ovariectomized rats, early low-potency chloride sion of EphB4 were involved in a bidirectional communi- channel inhibitors were able to prevent bone resorption by cation between these cell types. EphrinB2 on osteoclasts approximately 50%, as monitored by both BMD and the stimulated bone formation by the osteoblasts via binding biochemical markers of bone resorption CTX-I or deoxy- to EphB4, while EphB4 expression on osteoblasts in turn pyridinoline, while augmenting the number of osteoclasts inhibited osteoclastogenesis via binding to EphrinB2 (12). Endocrine Reviews, February 2011, 32(1):31–63 edrv.endojournals.org 49

However, ephrin signaling requires close contact between gies; and 2) stimulation of bone formation by the osteo- the osteoclasts and their target cells. This has led to the blasts, a process that as illustrated by studies conducted in speculation that ephrin signaling could be involved in the osteopetrotic patients is, to a large extent, independent of interplay between osteoclasts and bone-lining cells, which bone resorption. It is presently not completely clear when are found in close contact and appear to regulate the ac- the osteoclasts are anabolically active, yet it appears to be tivity of each other (68, 375). related to the presence of large multinuclear osteoclasts Stimulatory signals from osteoclasts directly to mature because bone anabolic responses are seen under these cir- bone-forming osteoblasts are, on the other hand, likely to cumstances (3, 15). be paracrine because these cell types are not found in close Understanding osteoclast functioning may be useful for contact (264). Both TGF-␤ and IGF-I are produced by the developing drugs that not only inhibit bone resorption but osteoclasts and are known to stimulate bone formation also enable bone resorption levels that ensure targeted under various circumstances (376–380). In relation to remodeling and, importantly, support continued anabolic these findings, it is interesting that the anabolic effect of signaling from osteoclasts to osteoblasts in the bone re- PTH in mice was shown to be mediated through IGF-I modeling compartment. This has the triple effect of: (350), an effect that is absent in the absence of osteoclasts 1) maintaining a sufficient resorption level and thereby (166). This confirms that IGF-I is a coupling factor. avoiding excessive aging of the bones; 2) sustaining a local A recent study demonstrated the mature human osteo- stimulation of bone formation at the resorption site only; clasts, independent of their resorptive activity, secrete fac- and 3) not initiating induction of bone formation in oth- tors that activate nodule formation by the osteoblasts (11). erwise quiescent sites. Theoretically, this type of inhibition This study was followed by a study showing that oste- of bone resorption would allow a continuous, ongoing oclasts produce the anabolic factors bone morphogenetic increase in BMD, which is in contrast to the effects of the protein 6, Wnt10b, and sphingosine-1-phosphate, again presently approved antiresorptives where a plateau effect independent of bone resorption (13). Furthermore, inhi- on BMD is observed within the first 12–18 months. This bition of bone morphogenetic protein 6, Wnt10b, and means that even with less powerful suppression of bone sphingosine-1-phosphate led to inhibition of the oste- resorption, such as that seen with the oral formulation of oclast-mediated stimulation of bone formation in vitro. salmon calcitonin, the long-term effects would surpass Finally, osteoclasts have also been shown to produce car- those of the bisphosphonates. diotrophin-1 (CT-1), which activates bone formation by A deeper understanding of both the differences in the osteoblasts, although the role of CT-1 was clearly shown resorption process depending on circumstances and the to be dependent on age because loss of CT-1 in newborn knowledge relating to when the osteoclasts are anaboli- mice caused osteopenia, whereas in larger mice it caused cally active will aid in the identification of novel treatment mild osteopetrosis due to defective bone resorption (360). opportunities for bone diseases. In summary, the presence of mature osteoclasts is as- sociated with the secretion of stimulation of bone anabolic Finally, the use of biochemical markers of bone turn- signals, and whereas several candidate factors have been over is becoming increasingly relevant for the continued identified, a clear demonstration that removal of one of the understanding of osteoclasts. Markers provide systemic molecules specifically in the osteoclasts in vivo leads to loss information on the outcome of a given treatment and can of bone formation is still missing. help answer questions such as whether glucocorticoids exert detrimental effects on bone formation, and whether antiresorptives antagonize bone formation secondary to VIII. Conclusions and Future Perspectives bone resorption because of suppression of osteoclast num- bers or activity. Osteoclasts have traditionally been viewed as bone resorp- tion “machines”; however, studies of osteoclasts have highlighted that these cells are highly context-specific, and the context of the individual osteoclasts is important for Acknowledgments the continued regulation of bone remodeling. The Danish Research Foundation “Den Danske Forskningsfond” is ac- As described in detail in this review, the osteoclasts knowledged for financial support. possess at least two highly important functions: 1) bone resorption, a process that is highly dependent on a series Address all correspondence and requests for reprints to: K. Henriksen, of external stimuli, such as matrix type, remodeling status, Nordic Bioscience A/S, Herlev Hovedgade 207, DK-2730 Herlev, Den- mark. E-mail: [email protected]. hormones involved in calcium homeostasis, genotype, in- Disclosure Summary: M.A.K. owns stocks in Nordic Bioscience A/S. flammation, and importantly also on intervention strate- All other authors have no conflicts of interest. 50 Henriksen et al. Osteoclast Subtypes Endocrine Reviews, February 2011, 32(1):31–63

References chanical properties of bone are compromised by disease. Endocr Rev 28:151–164 1. Segovia-Silvestre T, Neutzsky-Wulff AV, Sorensen MG, 17. Takahashi H, Epker B, Frost HM 1964 Resorption pre- Christiansen C, Bollerslev J, Karsdal MA, Henriksen K cedes formative activity. Surg Forum 15:437–438 2009 Advances in osteoclast biology resulting from 18. Hattner R, Epker BN, Frost HM 1965 Suggested sequen- the study of osteopetrotic mutations. Hum Genet 124: tial mode of control of changes in cell behaviour in adult 561–577 bone remodelling. Nature 206:489–490 2. Everts V, de Vries TJ, Helfrich MH 2009 Osteoclast het- 19. Parfitt AM 1982 The coupling of bone formation to bone erogeneity: lessons from osteopetrosis and inflammatory resorption: a critical analysis of the concept and of its rel- conditions. Biochim Biophys Acta 1792:757–765 evance to the pathogenesis of osteoporosis. Metab Bone 3. Karsdal MA, Martin TJ, Bollerslev J, Christiansen C, Hen- Dis Relat Res 4:1–6 riksen K 2007 Are nonresorbing osteoclasts sources of 20. Martin TJ 1993 Hormones in the coupling of bone resorp- bone anabolic activity? J Bone Miner Res 22:487–494 tion and formation. Osteoporos Int 3(Suppl 1):121–125 4. Everts V, Korper W, Jansen DC, Steinfort J, Lammerse I, 21. Nakamura M, Udagawa N, Matsuura S, Mogi M, Naka- Heera S, Docherty AJ, Beertsen W 1999 Functional heter- mura H, Horiuchi H, Saito N, Hiraoka BY, Kobayashi Y, ogeneity of osteoclasts: matrix metalloproteinases partic- Takaoka K, Ozawa H, Miyazawa H, Takahashi N 2003 ipate in osteoclastic resorption of calvarial bone but not in Osteoprotegerin regulates bone formation through a cou- resorption of long bone. FASEB J 13:1219–1230 pling mechanism with bone resorption. Endocrinology 5. Marks Jr SC 1983 The origin of osteoclasts: evidence, clin- 144:5441–5449 ical implications and investigative challenges of an extra- 22. Teitelbaum SL, Ross FP 2003 Genetic regulation of os- skeletal source. J Oral Pathol 12:226–256 teoclast development and function. Nat Rev Genet 6. Leeming DJ, Henriksen K, Byrjalsen I, Qvist P, Madsen SH, 4:638–649 Garnero P, Karsdal MA 2009 Is bone quality associated 23. Goltzman D 2002 Discoveries, drugs and skeletal disor- with collagen age? Osteoporos Int 20:1461–1470 ders. Nat Rev Drug Discov 1:784–796 7. Henriksen K, Leeming DJ, Byrjalsen I, Nielsen RH, So- 24. Martin TJ, Seeman E 2007 New mechanisms and targets in rensen MG, Dziegiel MH, Martin TJ, Christiansen C, Qvist the treatment of bone fragility. Clin Sci (Lond) 112:77–91 P, Karsdal MA 2007 Osteoclasts prefer aged bone. Osteo- 25. Harvey N, Earl S, Cooper C 2006 Epidemiology of osteo- poros Int 18:751–759 porotic fractures. In: Favus MJ, ed. Primer on the metabolic 8. Karsdal MA, Henriksen K, Sørensen MG, Gram J, Schaller bone diseases and disorders of mineral metabolism. Chap 42. 6th ed. Philadelphia: Lippincott Williams & Wilkins; S, Dziegiel MH, Heegaard AM, Christophersen P, Martin 244–248 TJ, Christiansen C, Bollerslev J 2005 Acidification of the 26. Albers-Scho¨ nberg HE 1904 Ro¨ ntgenbilder einer seltenen osteoclastic resorption compartment provides insight into Knockenerkrankung. 5th ed. Munch. Med. Wochenschr, the coupling of bone formation to bone resorption. Am J Münich: Germany; 365–368 Pathol 166:467–476 27. Tolar J, Teitelbaum SL, Orchard PJ 2004 Osteopetrosis. 9. Martin TJ, Sims NA 2005 Osteoclast-derived activity in the N Engl J Med 351:2839–2849 coupling of bone formation to resorption. Trends Mol Med 28. Sobacchi C, Frattini A, Guerrini MM, Abinun M, Pangra- 11:76–81 zio A, Susani L, Bredius R, Mancini G, Cant A, Bishop N, 10. Martin TJ, Quinn JM, Gillespie MT, Ng KW, Karsdal MA, Grabowski P, Del Fattore A, Messina C, Errigo G, Coxon Sims NA 2006 Mechanisms involved in skeletal anabolic FP, Scott DI, Teti A, Rogers MJ, Vezzoni P, Villa A, Hel- therapies. Ann NY Acad Sci 1068:458–470 frich MH 2007 Osteoclast-poor human osteopetrosis due 11. Karsdal MA, Neutzsky-Wulff AV, Dziegiel MH, Chris- to mutations in the gene encoding RANKL. Nat Genet tiansen C, Henriksen K 2008 Osteoclasts secrete non-bone 39:960–962 derived signals that induce bone formation. Biochem Bio- 29. Guerrini MM, Sobacchi C, Cassani B, Abinun M, Kilic SS, phys Res Commun 366:483–488 Pangrazio A, Moratto D, Mazzolari E, Clayton-Smith J, 12. Zhao C, Irie N, Takada Y, Shimoda K, Miyamoto T, Nishi- Orchard P, Coxon FP, Helfrich MH, Crockett JC, Mellis waki T, Suda T, Matsuo K 2006 Bidirectional ephrinB2- D, Vellodi A, Tezcan I, Notarangelo LD, Rogers MJ, Vez- EphB4 signaling controls bone homeostasis. Cell Metab zoni P, Villa A, Frattini A 2008 Human osteoclast-poor 4:111–121 osteopetrosis with hypogammaglobulinemia due to 13. Pederson L, Ruan M, Westendorf JJ, Khosla S, Oursler MJ TNFRSF11A (RANK) mutations. Am J Hum Genet 83: 2008 Regulation of bone formation by osteoclasts involves 64–76 Wnt/BMP signaling and the chemokine sphingosine-1- 30. Henriksen K, Gram J, Høegh-Andersen P, Jemtland R, phosphate. Proc Natl Acad Sci USA 105:20764–20769 Ueland T, Dziegiel MH, Schaller S, Bollerslev J, Karsdal 14. Seeman E, Delmas PD 2006 Bone quality—the material MA 2005 Osteoclasts from patients with autosomal dom- and structural basis of bone strength and fragility. N Engl inant osteopetrosis type I (ADOI) caused by a T253I mu- J Med 354:2250–2261 tation in LRP5 are normal in vitro, but have decreased 15. Henriksen K, Neutzsky-Wulff AV, Bonewald LF, Karsdal resorption capacity in vivo. Am J Pathol 167:1341–1348 MA 2009 Local communication on and within bone con- 31. Glass 2nd DA, Karsenty G 2006 Canonical Wnt signaling trols bone remodeling. Bone 44:1026–1033 in osteoblasts is required for osteoclast differentiation. Ann 16. Chavassieux P, Seeman E, Delmas PD 2007 Insights into NY Acad Sci 1068:117–130 material and structural basis of bone fragility from diseases 32. Henriksen K, Tanko LB, Qvist P, Delmas PD, Christiansen associated with fractures: how determinants of the biome- C, Karsdal MA 2007 Assessment of osteoclast number and Endocrine Reviews, February 2011, 32(1):31–63 edrv.endojournals.org 51

function: application in the development of new and im- the microphthalmia transcription factor family. Proc Natl proved treatment modalities for bone diseases. Osteoporos Acad Sci USA 98:5798–5803 Int 18:681–685 49. Luchin A, Purdom G, Murphy K, Clark MY, Angel N, 33. Teitelbaum SL 2007 Osteoclasts: what do they do and how Cassady AI, Hume DA, Ostrowski MC 2000 The microph- do they do it? Am J Pathol 170:427–435 thalmia transcription factor regulates expression of the tar- 34. Hayman AR, Jones SJ, Boyde A, Foster D, Colledge WH, trate-resistant acid phosphatase gene during terminal dif- Carlton MB, Evans MJ, Cox TM 1996 Mice lacking tar- ferentiation of osteoclasts. J Bone Miner Res 15:451–460 trate-resistant acid phosphatase (Acp 5) have disrupted en- 50. Zhao H, Laitala-Leinonen T, Parikka V, Va¨a¨na¨nen HK dochondral ossification and mild osteopetrosis. Develop- 2001 Downregulation of small GTPase Rab7 impairs os- ment 122:3151–3162 teoclast polarization and bone resorption. J Biol Chem 35. Sly WS, Hewett-Emmett D, Whyte MP, Yu YS, Tashian RE 276:39295–39302 1983 Carbonic anhydrase II deficiency identified as the 51. Rodan GA, Martin TJ 2000 Therapeutic approaches to primary defect in the autosomal recessive syndrome of os- bone diseases. Science 289:1508–1514 teopetrosis with renal tubular acidosis and cerebral calci- 52. Blair HC, Teitelbaum SL, Ghiselli R, Gluck S 1989 fication. Proc Natl Acad Sci USA 80:2752–2756 Osteoclastic bone resorption by a polarized vacuolar 36. Gelb BD, Shi GP, Chapman HA, Desnick RJ 1996 Pycno- proton pump. Science 245:855–857 dysostosis, a lysosomal disease caused by cathepsin K de- 53. Blair HC, Teitelbaum SL, Tan HL, Koziol CM, Schlesinger ficiency. Science 273:1236–1238 PH 1991 Passive chloride permeability charge coupled to 37. Frattini A, Orchard PJ, Sobacchi C, Giliani S, Abinun M, H(ϩ)-ATPase of avian osteoclast ruffled membrane. Am J Mattsson JP, Keeling DJ, Andersson AK, Wallbrandt P, Physiol 260:C1315–C1324 Zecca L, Notarangelo LD, Vezzoni P, Villa A 2000 Defects 54. Baron R, Neff L, Louvard D, Courtoy PJ 1985 Cell-medi- in TCIRG1 subunit of the vacuolar proton pump are re- ated extracellular acidification and bone resorption: evi- sponsible for a subset of human autosomal recessive os- dence for a low pH in resorbing lacunae and localization of teopetrosis. Nat Genet 25:343–346 a 100-kD lysosomal membrane protein at the osteoclast 38. Vu TH, Shipley JM, Bergers G, Berger JE, Helms JA, ruffled border. J Cell Biol 101:2210–2222 Hanahan D, Shapiro SD, Senior RM, Werb Z 1998 55. Scimeca JC, Franchi A, Trojani C, Parrinello H, MMP-9/gelatinase B is a key regulator of growth plate Grosgeorge J, Robert C, Jaillon O, Poirier C, Gaudray P, angiogenesis and apoptosis of hypertrophic chondro- Carle GF 2000 The gene encoding the mouse homologue of cytes. Cell 93:411–422 the human osteoclast-specific 116-kDa V-ATPase subunit 39. Kornak U, Kasper D, Bo¨ sl MR, Kaiser E, Schweizer M, bears a deletion in osteosclerotic (oc/oc) mutants. Bone Schulz A, Friedrich W, Delling G, Jentsch TJ 2001 Loss of 26:207–213 the ClC-7 chloride channel leads to osteopetrosis in mice 56. Li YP, Chen W, Liang Y, Li E, Stashenko P 1999 Atp6i- and man. Cell 104:205–215 deficient mice exhibit severe osteopetrosis due to loss of 40. Chalhoub N, Benachenhou N, Rajapurohitam V, Pata M, osteoclast-mediated extracellular acidification. Nat Genet Ferron M, Frattini A, Villa A, Vacher J 2003 Grey-lethal 23:447–451 mutation induces severe malignant autosomal recessive os- 57. Kornak U, Schulz A, Friedrich W, Uhlhaas S, Kremens B, teopetrosis in mouse and human. Nat Med 9:399–406 Voit T, Hasan C, Bode U, Jentsch TJ, Kubisch C 2000 41. Findlay DM, Martin TJ 1997 Receptors of calciotropic Mutations in the a3 subunit of the vacuolar H(ϩ)-ATPase hormones. Horm Metab Res 29:128–134 cause infantile malignant osteopetrosis. Hum Mol Genet 42. Del Fattore A, Cappariello A, Teti A 2008 Genetics, 9:2059–2063 pathogenesis and complications of osteopetrosis. Bone 58. Henriksen K, Gram J, Schaller S, Dahl BH, Dziegiel MH, 42:19–29 Bollerslev J, Karsdal MA 2004 Characterization of oste- 43. Va¨a¨na¨nen HK, Horton M 1995 The osteoclast clear zone oclasts from patients harboring a G215R mutation in is a specialized cell-extracellular matrix adhesion structure. ClC-7 causing autosomal dominant osteopetrosis type II. J Cell Sci 108:2729–2732 Am J Pathol 164:1537–1545 44. Roodman GD 1999 Cell biology of the osteoclast. Exp 59. Graves AR, Curran PK, Smith CL, Mindell JA 2008 The Hematol 27:1229–1241 Cl(Ϫ)/H(ϩ) antiporter ClC-7 is the primary chloride per- 45. Boyle WJ, Simonet WS, Lacey DL 2003 Osteoclast differ- meation pathway in lysosomes. Nature 453:788–792 entiation and activation. Nature 423:337–342 60. Weinert S, Jabs S, Supanchart C, Schweizer M, Gimber N, 46. Zou W, Kitaura H, Reeve J, Long F, Tybulewicz VL, Shattil Richter M, Rademann J, Stauber T, Kornak U, Jentsch SJ, Ginsberg MH, Ross FP, Teitelbaum SL 2007 Syk, c-Src, TJ 2010 Lysosomal pathology and osteopetrosis upon the ␣v␤3 integrin, and ITAM immunoreceptors, in con- loss of Hϩ-driven lysosomal ClϪ accumulation. Science cert, regulate osteoclastic bone resorption. J Cell Biol 176: 328:1401–1403 877–888 61. Josephsen K, Praetorius J, Frische S, Gawenis LR, Kwon 47. Meadows NA, Sharma SM, Faulkner GJ, Ostrowski MC, TH, Agre P, Nielsen S, Fejerskov O 2009 Targeted disrup- Hume DA, Cassady AI 2007 The expression of Clcn7 and tion of the ClϪ/HCO3Ϫ exchanger Ae2 results in osteo- Ostm1 in osteoclasts is coregulated by microphthalmia petrosis in mice. Proc Natl Acad Sci USA 106:1638–1641 transcription factor. J Biol Chem 282:1891–1904 62. Jansen ID, Mardones P, Lecanda F, de Vries TJ, Recalde S, 48. Motyckova G, Weilbaecher KN, Horstmann M, Rieman Hoeben KA, Schoenmaker T, Ravesloot JH, van Borren DJ, Fisher DZ, Fisher DE 2001 Linking osteopetrosis and MM, van Eijden TM, Bronckers AL, Kellokumpu S, Me- pycnodysostosis: regulation of cathepsin K expression by dina JF, Everts V, Oude Elferink RP 2009 Ae2a,b-Deficient 52 Henriksen et al. Osteoclast Subtypes Endocrine Reviews, February 2011, 32(1):31–63

mice exhibit osteopetrosis of long bones but not of calvaria. of tartrate-resistant acid phosphatase than long bone os- FASEB J 23:3470–3481 teoclasts and activation does not depend on cathepsin K or 63. Wu J, Glimcher LH, Aliprantis AO 2008 HCO3Ϫ/ClϪ L activity. Calcif Tissue Int 79:245–254 anion exchanger SLC4A2 is required for proper osteoclast 77. Hollberg K, Hultenby K, Hayman A, Cox T, Andersson G differentiation and function. Proc Natl Acad Sci USA 105: 2002 Osteoclasts from mice deficient in tartrate-resistant 16934–16939 acid phosphatase have altered ruffled borders and dis- 64. Bossard MJ, Tomaszek TA, Thompson SK, Amegadzie turbed intracellular vesicular transport. Exp Cell Res 279: BY, Hanning CR, Jones C, Kurdyla JT, McNulty DE, 227–238 Drake FH, Gowen M, Levy MA 1996 Proteolytic activity 78. Roberts HC, Knott L, Avery NC, Cox TM, Evans MJ, of human osteoclast cathepsin K. Expression, purification, Hayman AR 2007 Altered collagen in tartrate-resistant activation, and substrate identification. J Biol Chem 271: acid phosphatase (TRAP)-deficient mice: a role for 12517–12524 TRAP in bone collagen metabolism. Calcif Tissue Int 65. Saftig P, Hunziker E, Wehmeyer O, Jones S, Boyde A, Rom- 80:400–410 merskirch W, Moritz JD, Schu P, von Figura K 1998 Im- 79. van den Bos T, Speijer D, Bank RA, Bro¨ mme D, Everts V paired osteoclastic bone resorption leads to osteopetrosis 2008 Differences in matrix composition between calvaria in cathepsin-K-deficient mice. Proc Natl Acad Sci USA 95: and long bone in mice suggest differences in biomechanical 13453–13458 properties and resorption: special emphasis on collagen. 66. Gowen M, Lazner F, Dodds R, Kapadia R, Feild J, Tavaria Bone 43:459–468 M, Bertoncello I, Drake F, Zavarselk S, Tellis I, Hertzog P, 80. Engsig MT, Chen QJ, Vu TH, Pedersen AC, Therkidsen B, Debouck C, Kola I 1999 Cathepsin K knockout mice de- Lund LR, Henriksen K, Lenhard T, Foged NT, Werb Z, velop osteopetrosis due to a deficit in matrix degradation Delaisse´JM2000 Matrix metalloproteinase 9 and vascular but not demineralization. J Bone Miner Res 14:1654–1663 endothelial growth factor are essential for osteoclast re- 67. Nishi Y, Atley L, Eyre DE, Edelson JG, Superti-Furga A, cruitment into developing long bones. J Cell Biol 151: Yasuda T, Desnick RJ, Gelb BD 1999 Determination of 879–889 bone markers in pycnodysostosis: effects of cathepsin K 81. Delaisse´ JM, Andersen TL, Engsig MT, Henriksen K, deficiency on bone matrix degradation. J Bone Miner Res Troen T, Blavier L 2003 Matrix metalloproteinases 14:1902–1908 (MMP) and cathepsin K contribute differently to osteoclas- 68. Everts V, Delaisse´ JM, Korper W, Jansen DC, Tigchelaar- tic activities. Microsc Res Tech 61:504–513 Gutter W, Saftig P, Beertsen W 2002 The bone lining cell: 82. Shibata S, Yamashita Y 2001 An ultrastructural study of its role in cleaning Howship’s lacunae and initiating bone osteoclasts and chondroclasts in poorly calcified mandible formation. J Bone Miner Res 17:77–90 induced by high doses of strontium diet to fetal mice. Ann 69. Henriksen K, Sørensen MG, Nielsen RH, Gram J, Schaller Anat 183:357–361 S, Dziegiel MH, Everts V, Bollerslev J, Karsdal MA 2006 83. Bromley M, Woolley DE 1984 Chondroclasts and osteoclasts Degradation of the organic phase of bone by osteoclasts: a at subchondral sites of erosion in the rheumatoid joint. Ar- secondary role for lysosomal acidification. J Bone Miner thritis Rheum 27:968–975 Res 21:58–66 84. Karsdal MA, Leeming DJ, Dam EB, Henriksen K, Alex- 70. Everts V, Korper W, Hoeben KA, Jansen ID, Bromme D, Cleutjens KB, Heeneman S, Peters C, Reinheckel T, Saftig andersen P, Pastoureau P, Altman RD, Christiansen C P, Beertsen W 2006 Osteoclastic bone degradation and the 2008 Should subchondral bone turnover be targeted role of different cysteine proteinases and matrix metallo- when treating osteoarthritis? Osteoarthritis Cartilage proteinases: differences between calvaria and long bone. 16:638–646 J Bone Miner Res 21:1399–1408 85. Mansell JP, Collins C, Bailey AJ 2007 Bone, not cartilage, 71. Shorey S, Heersche JN, Manolson MF 2004 The relative should be the major focus in osteoarthritis. Nat Clin Pract contribution of cysteine proteinases and matrix metallo- Rheumatol 3:306–307 proteinases to the resorption process in osteoclasts derived 86. Ortega N, Behonick DJ, Werb Z 2004 Matrix remodeling from long bone and scapula. Bone 35:909–917 during endochondral ossification. Trends Cell Biol 72. Salo J, Lehenkari P, Mulari M, Metsikko¨K,Va¨a¨na¨nen HK 14:86–93 1997 Removal of osteoclast bone resorption products by 87. Kong YY, Yoshida H, Sarosi I, Tan HL, Timms E, Cap- transcytosis. Science 276:270–273 parelli C, Morony S, Oliveira-dos-Santos AJ, Van G, Itie A, 73. Nesbitt SA, Horton MA 1997 Trafficking of matrix col- Khoo W, Wakeham A, Dunstan CR, Lacey DL, Mak TW, lagens through bone-resorbing osteoclasts. Science 276: Boyle WJ, Penninger JM 1999 OPGL is a key regulator of 266–269 osteoclastogenesis, lymphocyte development and lymph- 74. Baron R 2005 General principles of bone biology. In: node organogenesis. Nature 397:315–323 Rosen CJ, Compston JE, Lian JB, eds. Primer on the met- 88. Wiktor-Jedrzejczak W, Bartocci A, Ferrante Jr AW, abolic bone diseases and disorders of mineral metabolism. Ahmed-Ansari A, Sell KW, Pollard JW, Stanley ER 1990 Chap 1. 5th ed. Hoboken, NJ: Wiley; 1–8 Total absence of colony-stimulating factor 1 in the mac- 75. Bollerslev J 1989 Autosomal dominant osteopetrosis: bone rophage-deficient osteopetrotic (op/op) mouse. Proc Natl metabolism and epidemiological, clinical, and hormonal Acad Sci USA 87:4828–4832 aspects. Endocr Rev 10:45–67 89. Wiktor-Jedrzejczak W, Urbanowska E, Aukerman SL, Pol- 76. Perez-Amodio S, Jansen DC, Schoenmaker T, Vogels IM, lard JW, Stanley ER, Ralph P, Ansari AA, Sell KW, Szperl Reinheckel T, Hayman AR, Cox TM, Saftig P, Beertsen W, M 1991 Correction by CSF-1 of defects in the osteopetrotic Everts V 2006 Calvarial osteoclasts express a higher level op/op mouse suggests local, developmental, and humoral Endocrine Reviews, February 2011, 32(1):31–63 edrv.endojournals.org 53

requirements for this growth factor. Exp Hematol sated for by enhanced osteoclastogenesis and increased ex- 19:1049–1054 pression of other proteases via an increased RANKL/OPG 90. Li J, Sarosi I, Yan XQ, Morony S, Capparelli C, Tan HL, ratio. Bone 36:159–172 McCabe S, Elliott R, Scully S, Van G, Kaufman S, Juan SC, 104. Zenger S, Hollberg K, Ljusberg J, Norgård M, Ek- Sun Y, Tarpley J, Martin L, Christensen K, McCabe J, Rylander B, Kiviranta R, Andersson G 2007 Proteolytic Kostenuik P, Hsu H, Fletcher F, Dunstan CR, Lacey DL, processing and polarized secretion of tartrate-resistant Boyle WJ 2000 RANK is the intrinsic hematopoietic cell acid phosphatase is altered in a subpopulation of me- surface receptor that controls osteoclastogenesis and reg- taphyseal osteoclasts in cathepsin K-deficient mice. ulation of bone mass and calcium metabolism. Proc Natl Bone 41:820–832 Acad Sci USA 97:1566–1571 105. Burr DB 2002 Targeted and nontargeted remodeling. Bone 91. Gerber HP, Vu TH, Ryan AM, Kowalski J, Werb Z, Fer- 30:2–4 rara N 1999 VEGF couples hypertrophic cartilage remod- 106. Parfitt AM 2002 Targeted and nontargeted bone remod- eling, ossification and angiogenesis during endochondral eling: relationship to basic multicellular unit origination bone formation. Nat Med 5:623–628 and progression. Bone 30:5–7 92. Blavier L, Delaisse´JM1995 Matrix metalloproteinases are 107. Noble B 2003 Bone microdamage and cell apoptosis. Eur obligatory for the migration of preosteoclasts to the devel- Cell Mater 6:46–55; discussion 55 oping marrow cavity of primitive long bones. J Cell Sci 108. Verborgt O, Gibson GJ, Schaffler MB 2000 Loss of osteo- 108:3649–3659 cyte integrity in association with microdamage and bone 93. Dieudonne´ SC, Foo P, van Zoelen EJ, Burger EH 1991 remodeling after fatigue in vivo. J Bone Miner Res 15: Inhibiting and stimulating effects of TGF-␤ 1 on osteoclas- 60–67 tic bone resorption in fetal mouse bone organ cultures. 109. Tatsumi S, Ishii K, Amizuka N, Li M, Kobayashi T, Kohno J Bone Miner Res 6:479–487 K, Ito M, Takeshita S, Ikeda K 2007 Targeted ablation of 94. Lowe C, Yoneda T, Boyce BF, Chen H, Mundy GR, So- osteocytes induces osteoporosis with defective mechano- riano P 1993 Osteopetrosis in Src-deficient mice is due to transduction. Cell Metab 5:464–475 an autonomous defect of osteoclasts. Proc Natl Acad Sci 110. Talmage RV, Talmage DW 2006 Calcium homeostasis: USA 90:4485–4489 solving the solubility problem. J Musculoskelet Neuronal 95. Neutzsky-Wulff AV, Karsdal MA, Henriksen K 2008 Interact 6:402–407 Characterization of the bone phenotype in ClC-7-deficient 111. Parfitt AM 2003 Misconceptions (3): calcium leaves bone mice. Calcif Tissue Int 83:425–437 only by resorption and enters only by formation. Bone 96. Bollerslev J, Steiniche T, Melsen F, Mosekilde L 1989 33:259–263 Structural and histomorphometric studies of iliac crest tra- 112. Marenzana M, Shipley AM, Squitiero P, Kunkel JG, Ru- becular and cortical bone in autosomal dominant osteo- binacci A 2005 Bone as an ion exchange organ: evidence petrosis: a study of two radiological types. Bone 10:19–24 for instantaneous cell-dependent calcium efflux from bone 97. Bollerslev J, Marks Jr SC, Pockwinse S, Kassem M, Brixen not due to resorption. Bone 37:545–554 K, Steiniche T, Mosekilde L 1993 Ultrastructural investi- 113. Dent CE, Smellie JM, Watson L 1965 Studies in osteope- gations of bone resorptive cells in two types of autosomal trosis. Arch Dis Child 40:7–15 dominant osteopetrosis. Bone 14:865–869 114. Manolagas SC 2000 Birth and death of bone cells: basic 98. Nordahl J, Andersson G, Reinholt FP 1998 Chondroclasts regulatory mechanisms and implications for the patho- and osteoclasts in bones of young rats: comparison of ul- genesis and treatment of osteoporosis. Endocr Rev 21: trastructural and functional features. Calcif Tissue Int 63: 115–137 401–408 115. Karsdal MA, Hjorth P, Henriksen K, Kirkegaard T, 99. Sawae Y, Sahara T, Sasaki T 2003 Osteoclast differen- Nielsen KL, Lou H, Delaisse´ JM, Foged NT 2003 Trans- tiation at growth plate cartilage-trabecular bone junc- forming growth factor-␤ controls human osteoclastogen- tion in newborn rat femur. J Electron Microsc (Tokyo) esis through the p38 MAPK and regulation of RANK ex- 52:493–502 pression. J Biol Chem 278:44975–44987 100. Pennypacker B, Shea M, Liu Q, Masarachia P, Saftig P, 116. Van Wesenbeeck L, Odgren PR, Coxon FP, Frattini A, Rodan S, Rodan G, Kimmel D 2009 Bone density, strength, Moens P, Perdu B, MacKay CA, Van Hul E, Timmermans and formation in adult cathepsin K (Ϫ/Ϫ) mice. Bone 44: JP, Vanhoenacker F, Jacobs R, Peruzzi B, Teti A, Helfrich 199–207 MH, Rogers MJ, Villa A, Van Hul W 2007 Involvement of 101. Garnero P, Ferreras M, Karsdal MA, Nicamhlaoibh R, PLEKHM1 in osteoclastic vesicular transport and osteo- Risteli J, Borel O, Qvist P, Delmas PD, Foged NT, Delaisse´ petrosis in incisors absent rats and humans. J Clin Invest JM 2003 The type I collagen fragments ICTP and CTX 117:919–930 reveal distinct enzymatic pathways of bone collagen deg- 117. Valcourt U, Merle B, Gineyts E, Viguet-Carrin S, Delmas radation. J Bone Miner Res 18:859–867 PD, Garnero P 2007 Non-enzymatic glycation of bone col- 102. Sørensen MG, Henriksen K, Schaller S, Henriksen DB, lagen modifies osteoclastic activity and differentiation. Nielsen FC, Dziegiel MH, Karsdal MA 2007 Character- J Biol Chem 282:5691–5703 ization of osteoclasts derived from CD14ϩ monocytes iso- 118. Schlemmer A, Hassager C, Jensen SB, Christiansen C 1992 lated from peripheral blood. J Bone Miner Metab Marked diurnal variation in urinary excretion of pyri- 25:36–45 dinium cross-links in premenopausal women. J Clin En- 103. Kiviranta R, Morko J, Alatalo SL, NicAmhlaoibh R, Risteli docrinol Metab 74:476–480 J, Laitala-Leinonen T, Vuorio E 2005 Impaired bone re- 119. Gertz BJ, Clemens JD, Holland SD, Yuan W, Greenspan S sorption in cathepsin K-deficient mice is partially compen- 1998 Application of a new serum assay for type I collagen 54 Henriksen et al. Osteoclast Subtypes Endocrine Reviews, February 2011, 32(1):31–63

cross-linked N-telopeptides: assessment of diurnal changes from peripheral blood mononuclear cells in postmeno- in bone turnover with and without alendronate treatment. pausal osteoporosis. FASEB J 19:410–412 Calcif Tissue Int 63:102–106 133. Jevon M, Hirayama T, Brown MA, Wass JA, Sabokbar A, 120. Qvist P, Christgau S, Pedersen BJ, Schlemmer A, Chris- Ostelere S, Athenasou NA 2003 Osteoclast formation tiansen C 2002 Circadian variation in the serum concen- from circulating precursors in osteoporosis. Scand J Rheu- tration of C-terminal telopeptide of type I collagen (serum matol 32:95–100 CTx): effects of gender, age, menopausal status, posture, 134. D’Amelio P, Grimaldi A, Di Bella S, Brianza SZ, Cristofaro daylight, serum cortisol, and fasting. Bone 31:57–61 MA, Tamone C, Giribaldi G, Ulliers D, Pescarmona GP, 121. Bjarnason NH, Henriksen EE, Alexandersen P, Christgau Isaia G 2008 Estrogen deficiency increases osteoclastogen- S, Henriksen DB, Christiansen C 2002 Mechanism of cir- esis up-regulating T cells activity: a key mechanism in os- cadian variation in bone resorption. Bone 30:307–313 teoporosis. Bone 43:92–100 122. Karsdal MA, Byrjalsen I, Riis BJ, Christiansen C 2008 135. Shevde NK, Bendixen AC, Dienger KM, Pike JW 2000 Optimizing bioavailability of oral administration of Estrogens suppress RANK ligand-induced osteoclast dif- small peptides through pharmacokinetic and pharma- ferentiation via a stromal cell independent mechanism in- codynamic parameters: the effect of water and timing of volving c-Jun repression. Proc Natl Acad Sci USA meal intake on oral delivery of salmon calcitonin. BMC 97:7829–7834 Clin Pharmacol 8:5 136. Srivastava S, Toraldo G, Weitzmann MN, Cenci S, Ross 123. Karsdal MA, Byrjalsen I, Azria M, Arnold M, Choi L, Riis FP, Pacifici R 2001 Estrogen decreases osteoclast forma- BJ, Christiansen C 2009 Influence of food intake on the tion by down-regulating receptor activator of NF-␬B li- bioavailability and efficacy of oral calcitonin. Br J Clin gand (RANKL)-induced JNK activation. J Biol Chem 276: Pharmacol 67:413–420 8836–8840 124. Henriksen DB, Alexandersen P, Byrjalsen I, Hartmann B, 137. Sørensen MG, Henriksen K, Dziegiel MH, Tanko´ LB, Bone HG, Christiansen C, Holst JJ 2004 Reduction of noc- Karsdal MA 2006 Estrogen directly attenuates human os- turnal rise in bone resorption by subcutaneous GLP-2. teoclastogenesis, but has no effect on resorption by mature Bone 34:140–147 osteoclasts. DNA Cell Biol 25:475–483 125. Henriksen DB, Alexandersen P, Hartmann B, Adrian CL, 138. Huber DM, Bendixen AC, Pathrose P, Srivastava S, Di- Byrjalsen I, Bone HG, Holst JJ, Christiansen C 2007 Dis- enger KM, Shevde NK, Pike JW 2001 Androgens suppress association of bone resorption and formation by GLP-2: a osteoclast formation induced by RANKL and macroph- 14-day study in healthy postmenopausal women. Bone 40: age-colony stimulating factor. Endocrinology 142:3800– 723–729 3808 126. Henriksen DB, Alexandersen P, Hartmann B, Adrian CL, 139. Nakamura T, Imai Y, Matsumoto T, Sato S, Takeuchi K, Byrjalsen I, Bone HG, Holst JJ, Christiansen C 2009 Four- Igarashi K, Harada Y, Azuma Y, Krust A, Yamamoto Y, month treatment with GLP-2 significantly increases hip Nishina H, Takeda S, Takayanagi H, Metzger D, Kanno J, BMD: a randomized, placebo-controlled, dose-ranging Takaoka K, Martin TJ, Chambon P, Kato S 2007 Estrogen study in postmenopausal women with low BMD. Bone prevents bone loss via estrogen receptor ␣ and induction of 45:833–842 Fas ligand in osteoclasts. Cell 130:811–823 127. Tanko´ LB, Bagger YZ, Alexandersen P, Devogelaer JP, 140. Bord S, Horner A, Beavan S, Compston J 2001 Estrogen Reginster JY, Chick R, Olson M, Benmammar H, Mind- receptors ␣ and ␤ are differentially expressed in developing eholm L, Azria M, Christiansen C 2004 Safety and efficacy human bone. J Clin Endocrinol Metab 86:2309–2314 of a novel salmon calcitonin (sCT) technology-based oral 141. Martin-Millan M, Almeida M, Ambrogini E, Han L, Zhao formulation in healthy postmenopausal women: acute and H, Weinstein RS, Jilka RL, O’Brien CA, Manolagas SC 3-month effects on biomarkers of bone turnover. J Bone 2010 The estrogen receptor-␣ in osteoclasts mediates the Miner Res 19:1531–1538 protective effects of estrogens on cancellous but not cor- 128. Eghbali-Fatourechi G, Khosla S, Sanyal A, Boyle WJ, tical bone. Mol Endocrinol 24:323–334 Lacey DL, Riggs BL 2003 Role of RANK ligand in medi- 142. Riggs BL, Melton Iii 3rd LJ, Robb RA, Camp JJ, Atkinson ating increased bone resorption in early postmenopausal EJ, Peterson JM, Rouleau PA, McCollough CH, Bouxsein women. J Clin Invest 111:1221–1230 ML, Khosla S 2004 Population-based study of age and sex 129. Clowes JA, Eghbali-Fatourechi GZ, McCready L, Oursler differences in bone volumetric density, size, geometry, and MJ, Khosla S, Riggs BL 2009 Estrogen action on bone structure at different skeletal sites. J Bone Miner Res 19: marrow osteoclast lineage cells of postmenopausal women 1945–1954 in vivo. Osteoporos Int 20:761–769 143. Recker R, Lappe J, Davies KM, Heaney R 2004 Bone re- 130. Taxel P, Kaneko H, Lee SK, Aguila HL, Raisz LG, Lorenzo modeling increases substantially in the years after meno- JA 2008 Estradiol rapidly inhibits osteoclastogenesis and pause and remains increased in older osteoporosis patients. RANKL expression in bone marrow cultures in postmeno- J Bone Miner Res 19:1628–1633 pausal women: a pilot study. Osteoporos Int 19:193–199 144. Garnero P, Sornay-Rendu E, Chapuy MC, Delmas PD 131. Cao JJ, Wronski TJ, Iwaniec U, Phleger L, Kurimoto P, 1996 Increased bone turnover in late postmenopausal Boudignon B, Halloran BP 2005 Aging increases stromal/ women is a major determinant of osteoporosis. J Bone osteoblastic cell-induced osteoclastogenesis and alters the Miner Res 11:337–349 osteoclast precursor pool in the mouse. J Bone Miner Res 145. Arlot ME, Delmas PD, Chappard D, Meunier PJ 1990 20:1659–1668 Trabecular and endocortical bone remodeling in post- 132. D’Amelio P, Grimaldi A, Pescarmona GP, Tamone C, menopausal osteoporosis: comparison with normal post- Roato I, Isaia G 2005 Spontaneous osteoclast formation menopausal women. Osteoporos Int 1:41–49 Endocrine Reviews, February 2011, 32(1):31–63 edrv.endojournals.org 55

146. Viguet-Carrin S, Garnero P, Delmas PD 2006 The role of 159. Nielsen RH, Karsdal MA, Sørensen MG, Dziegiel MH, collagen in bone strength. Osteoporos Int 17:319–336 Henriksen K 2007 Dissolution of the inorganic phase of 147. Herrmann M, Widmann T, Colaianni G, Colucci S, Zal- bone leading to release of calcium regulates osteoclast sur- lone A, Herrmann W 2005 Increased osteoclast activity in vival. Biochem Biophys Res Commun 360:834–839 the presence of increased homocysteine concentrations. 160. Alatalo SL, Ivaska KK, Waguespack SG, Econs MJ, Clin Chem 51:2348–2353 Va¨a¨na¨nen HK, Halleen JM 2004 Osteoclast-derived serum 148. Saito M, Marumo K 2010 Collagen cross-links as a deter- tartrate-resistant acid phosphatase 5b in Albers-Schonberg minant of bone quality: a possible explanation for bone disease (type II autosomal dominant osteopetrosis). Clin fragility in aging, osteoporosis, and diabetes mellitus. Os- Chem 50:883–890 teoporos Int 21:195–214 161. Del Fattore A, Peruzzi B, Rucci N, Recchia I, Cappariello 149. Miyata T, Notoya K, Yoshida K, Horie K, Maeda K, Ku- A, Longo M, Fortunati D, Ballanti P, Iacobini M, Luciani rokawa K, Taketomi S 1997 Advanced glycation end prod- M, Devito R, Pinto R, Caniglia M, Lanino E, Messina C, ucts enhance osteoclast-induced bone resorption in cul- Cesaro S, Letizia C, Bianchini G, Fryssira H, Grabowski P, tured mouse unfractionated bone cells and in rats Shaw N, Bishop N, Hughes D, Kapur RP, Datta HK, implanted subcutaneously with devitalized bone particles. Taranta A, Fornari R, Migliaccio S, Teti A 2006 Clinical, J Am Soc Nephrol 8:260–270 genetic, and cellular analysis of 49 osteopetrotic patients: 150. Schwartz AV, Garnero P, Hillier TA, Sellmeyer DE, Strot- implications for diagnosis and treatment. J Med Genet 43: meyer ES, Feingold KR, Resnick HE, Tylavsky FA, Black 315–325 DM, Cummings SR, Harris TB, Bauer DC 2009 Pentosi- 162. Soriano P, Montgomery C, Geske R, Bradley A 1991 Tar- dine and increased fracture risk in older adults with type 2 geted disruption of the c-src proto-oncogene leads to os- diabetes. J Clin Endocrinol Metab 94:2380–2386 teopetrosis in mice. Cell 64:693–702 151. Raska Jr I, Broulík P 2005 The impact of diabetes mellitus 163. Grigoriadis AE, Wang ZQ, Cecchini MG, Hofstetter W, on skeletal health: an established phenomenon with in- Felix R, Fleisch HA, Wagner EF 1994 c-Fos: a key regulator established causes? Prague Med Rep 106:137–148 of osteoclast-macrophage lineage determination and bone 152. Sanguineti R, Storace D, Monacelli F, Federici A, Odetti P remodeling. Science 266:443–448 2008 Pentosidine effects on human osteoblasts in vitro. 164. Marzia M, Sims NA, Voit S, Migliaccio S, Taranta A, Ber- Ann NY Acad Sci 1126:166–172 nardini S, Faraggiana T, Yoneda T, Mundy GR, Boyce BF, 153. Henriksen K, Gram J, Neutzsky-Wulff AV, Jensen VK, Baron R, Teti A 2000 Decreased c-Src expression enhances Dziegiel MH, Bollerslev J, Karsdal MA 2009 Character- osteoblast differentiation and bone formation. J Cell Biol ization of acid flux in osteoclasts from patients harboring 151:311–320 a G215R mutation in ClC-7. Biochem Biophys Res Com- 165. Demiralp B, Chen HL, Koh AJ, Keller ET, McCauley LK mun 378:804–809 2002 Anabolic actions of parathyroid hormone during 154. Taranta A, Migliaccio S, Recchia I, Caniglia M, Luciani M, bone growth are dependent on c-fos. Endocrinology 143: De Rossi G, Dionisi-Vici C, Pinto RM, Francalanci P, Bold- 4038–4047 rini R, Lanino E, Dini G, Morreale G, Ralston SH, Villa A, 166. Koh AJ, Demiralp B, Neiva KG, Hooten J, Nohutcu RM, Vezzoni P, Del Principe D, Cassiani F, Palumbo G, Teti A Shim H, Datta NS, Taichman RS, McCauley LK 2005 2003 Genotype-phenotype relationship in human ATP6i- Cells of the osteoclast lineage as mediators of the anabolic dependent autosomal recessive osteopetrosis. Am J Pathol actions of parathyroid hormone in bone. Endocrinology 162:57–68 146:4584–4596 155. Maranda B, Chabot G, De´carie JC, Pata M, Azeddine B, 167. Holmen SL, Zylstra CR, Mukherjee A, Sigler RE, Faugere Moreau A, Vacher J 2008 Clinical and cellular manifesta- MC, Bouxsein ML, Deng L, Clemens TL, Williams BO tions of OSTM1-related infantile osteopetrosis. J Bone 2005 Essential role of ␤-catenin in postnatal bone acqui- Miner Res 23:296–300 sition. J Biol Chem 280:21162–21168 156. Rajapurohitam V, Chalhoub N, Benachenhou N, Neff L, 168. Gong Y, Slee RB, Fukai N, Rawadi G, Roman-Roman S, Baron R, Vacher J 2001 The mouse osteopetrotic grey- Reginato AM, Wang H, Cundy T, Glorieux FH, Lev D, lethal mutation induces a defect in osteoclast maturation/ Zacharin M, Oexle K, Marcelino J, Suwairi W, Heeger S, function. Bone 28:513–523 Sabatakos G, Apte S, Adkins WN, Allgrove J, Arslan- 157. Semba I, Ishigami T, Sugihara K, Kitano M 2000 Higher Kirchner M, Batch JA, Beighton P, Black GC, Boles RG, osteoclastic demineralization and highly mineralized ce- Boon LM, Borrone C, Brunner HG, Carle GF, Dallapic- ment lines with osteocalcin deposition in a mandibular cola B, De Paepe A, Floege B, Halfhide ML, Hall B, Hen- cortical bone of autosomal dominant osteopetrosis type II: nekam RC, Hirose T, Jans A, Ju¨ ppner H, Kim CA, Keppler- ultrastructural and undecalcified histological investiga- Noreuil K, Kohlschuetter A, LaCombe D, Lambert M, tions. Bone 27:389–395 Lemyre E, Letteboer T, Peltonen L, Ramesar RS, Ro- 158. Garnero P, Thompson E, Woodworth T, Smolen JS 2010 manengo M, Somer H, Steichen-Gersdorf E, Steinmann B, Rapid and sustained improvement in bone and cartilage Sullivan B, Superti-Furga A, Swoboda W, van den Boo- turnover markers with the anti-interleukin-6 receptor gaard MJ, Van Hul W, Vikkula M, Votruba M, Zabel B, inhibitor tocilizumab plus methotrexate in rheumatoid Garcia T, Baron R, Olsen BR, Warman ML 2001 LDL arthritis patients with an inadequate response to meth- receptor-related protein 5 (LRP5) affects bone accrual and otrexate: results from a substudy of the multicenter dou- eye development. Cell 107:513–523 ble-blind, placebo-controlled trial of tocilizumab in inade- 169. Van Wesenbeeck L, Cleiren E, Gram J, Beals RK, Be´nichou quate responders to methotrexate alone. Arthritis Rheum. O, Scopelliti D, Key L, Renton T, Bartels C, Gong Y, War- 62:33–43 man ML, De Vernejoul MC, Bollerslev J, Van Hul W 2003 56 Henriksen et al. Osteoclast Subtypes Endocrine Reviews, February 2011, 32(1):31–63

Six novel missense mutations in the LDL receptor-related ing the molecular basis of Paget’s disease of bone. J Clin protein 5 (LRP5) gene in different conditions with an in- Pathol 63:199–203 creased bone density. Am J Hum Genet 72:763–771 185. Roodman GD, Windle JJ 2005 Paget disease of bone. J Clin 170. Bollerslev J, Ueland T, Odgren PR 2003 Serum levels of Invest 115:200–208 TGF-␤ and fibronectin in autosomal dominant osteope- 186. Ralston SH, Langston AL, Reid IR 2008 Pathogenesis trosis in relation to underlying mutations and well-de- and management of Paget’s disease of bone. Lancet 372: scribed murine counterparts. Crit Rev Eukaryot Gene Expr 155–163 13:163–171 187. Neale SD, Smith R, Wass JA, Athanasou NA 2000 Oste- 171. Sarnsethsiri P, Hitt OK, Eyring EJ, Frost HM 1971 Tetra- oclast differentiation from circulating mononuclear pre- cycline-based study of bone dynamics in pycnodysostosis. cursors in Paget’s disease is hypersensitive to 1,25-dihy- Clin Orthop Relat Res 74:301–312 droxyvitamin D(3) and RANKL. Bone 27:409–416 172. Ho N, Punturieri A, Wilkin D, Szabo J, Johnson M, Wha- 188. Singer FR, Mills BG, Gruber HE, Windle JJ, Roodman GD ley J, Davis J, Clark A, Weiss S, Francomano C 1999 Mu- 2006 Ultrastructure of bone cells in Paget’s disease of bone. tations of CTSK result in pycnodysostosis via a reduction J Bone Miner Res 21(Suppl 2):P51–P54 in cathepsin K protein. J Bone Miner Res 14:1649–1653 189. Kurihara N, Hiruma Y, Zhou H, Subler MA, Dempster 173. Hou WS, Bro¨ mme D, Zhao Y, Mehler E, Dushey C, Wein- DW, Singer FR, Reddy SV, Gruber HE, Windle JJ, Rood- stein H, Miranda CS, Fraga C, Greig F, Carey J, Rimoin man GD 2007 Mutation of the sequestosome 1 (p62) gene DL, Desnick RJ, Gelb BD 1999 Characterization of novel increases osteoclastogenesis but does not induce Paget dis- cathepsin K mutations in the pro and mature polypep- ease. J Clin Invest 117:133–142 tide regions causing pycnodysostosis. J Clin Invest 103: 190. Hiruma Y, Kurihara N, Subler MA, Zhou H, Boykin CS, 731–738 Zhang H, Ishizuka S, Dempster DW, Roodman GD, 174. Chavassieux P, Asser Karsdal M, Segovia-Silvestre T, Windle JJ 2008 A SQSTM1/p62 mutation linked to Paget’s Neutzsky-Wulff AV, Chapurlat R, Boivin G, Delmas PD disease increases the osteoclastogenic potential of the bone 2008 Mechanisms of the anabolic effects of teriparatide on microenvironment. Hum Mol Genet 17:3708–3719 bone: insight from the treatment of a patient with pycno- 191. Gennari L, Merlotti D, Mossetti G, Rendina D, De Paola dysostosis. J Bone Miner Res 23:1076–1083 V, Martini G, Nuti R 2009 The use of intravenous ami- 175. Everts V, Aronson DC, Beertsen W 1985 Phagocytosis of nobisphosphonates for the treatment of Paget’s disease of bone collagen by osteoclasts in two cases of pycnodysos- bone. Mini Rev Med Chem 9:1052–1063 tosis. Calcif Tissue Int 37:25–31 192. Clines GA, Guise TA 2008 Molecular mechanisms and 176. Li CY, Jepsen KJ, Majeska RJ, Zhang J, Ni R, Gelb BD, treatment of bone metastasis. Expert Rev Mol Med 10:e7 Schaffler MB 2006 Mice lacking cathepsin K maintain 193. Guise TA, Mohammad KS, Clines G, Stebbins EG, Wong bone remodeling but develop bone fragility despite high DH, Higgins LS, Vessella R, Corey E, Padalecki S, Suva L, bone mass. J Bone Miner Res 21:865–875 Chirgwin JM 2006 Basic mechanisms responsible for os- 177. Fratzl-Zelman N, Valenta A, Roschger P, Nader A, Gelb teolytic and osteoblastic bone metastases. Clin Cancer Res BD, Fratzl P, Klaushofer K 2004 Decreased bone turnover 12:6213s–6216s and deterioration of bone structure in two cases of pycno- 194. Hu MI, Lu H, Gagel RF 2010 Cancer therapies and bone dysostosis. J Clin Endocrinol Metab 89:1538–1547 178. Bone HG, McClung MR, Roux C, Recker RR, Eisman JA, health. Curr Rheumatol Rep 12:177–185 Verbruggen N, Hustad CM, DaSilva C, Santora AC, Ince 195. Akhtari M, Mansuri J, Newman KA, Guise TM, Seth P BA 2010 Odanacatib, a cathepsin-K inhibitor for osteo- 2008 Biology of breast cancer bone metastasis. Cancer Biol porosis: a two-year study in postmenopausal women with Ther 7:3–9 low bone density. J Bone Miner Res 25:937–947 196. Lipton A 2006 Future treatment of bone metastases. Clin 179. Pennypacker B, Wesolowski G, Heo J, Duong LT 2009 Cancer Res 12:6305s–6308s Effects of odanacatib on central femur cortical bone in 197. Clezardin P, Teti A 2007 Bone metastasis: pathogenesis estrogen-deficient adult rhesus monkeys. J Bone Miner Res and therapeutic implications. Clin Exp Metastasis 24: 24(Suppl 1):1171 (Abstract) 599–608 180. Cusick T, Pennypacker B, Scott K, Duong LT, Kimmel D 198. Cicek M, Oursler MJ 2006 Breast cancer bone metastasis 2009 Effects of odanacatib on bone mass, turnover and and current small therapeutics. Cancer Metastasis Rev 25: strength in the femoral neck of estrogen deficient adult 635–644 rhesus monkeys. J Bone Miner Res 24(Suppl 1):FR0416 199. Leeming DJ, Koizumi M, Byrjalsen I, Li B, Qvist P, Tanko´ (Abstract) LB 2006 The relative use of eight collagenous and noncol- 181. Scott K, Cusick T, Duong LT, Pennypacker B, Kimmel D lagenous markers for diagnosis of skeletal metastases in 2009 Effects of odanacatib on bone turnover and osteo- breast, prostate, or lung cancer patients. Cancer Epidemiol clast morphology in the lumbar vertebra of ovariectomized Biomarkers Prev 15:32–38 adult rhesus monkeys. J Bone Miner Res 24(Suppl 1): 200. Leeming DJ, Delling G, Koizumi M, Henriksen K, Karsdal SU0227 (Abstract) MA, Li B, Qvist P, Tanko´ LB, Byrjalsen I 2006 ␣ CTX as 182. Helfrich M, Crockett JC, Hocking LJ, Coxon FP 2007 The a biomarker of skeletal invasion of breast cancer: immu- pathogenesis of osteoclast diseases: some knowns, but still nolocalization and the load dependency of urinary excre- many unknowns. BoneKEy-Osteovision 4:61–77 tion. Cancer Epidemiol Biomarkers Prev 15:1392–1395 183. Roodman GD 1996 Paget’s disease and osteoclast biology. 201. Koopmans N, de Jong IJ, Breeuwsma AJ, van der Veer E Bone 19:209–212 2007 Serum bone turnover markers (PINP and ICTP) for 184. Goode A, Layfield R 2010 Recent advances in understand- the early detection of bone metastases in patients with Endocrine Reviews, February 2011, 32(1):31–63 edrv.endojournals.org 57

prostate cancer: a longitudinal approach. J Urol 178:849– trials of biologics for rheumatoid arthritis: a Cochrane 853 overview. CMAJ 181:787–796 202. Winding B, NicAmhlaoibh R, Misander H, Høegh- 218. Licastro F, Chiappelli M, Ianni M, Porcellini E 2009 Tu- Andersen P, Andersen TL, Holst-Hansen C, Heegaard mor necrosis factor-␣ antagonists: differential clinical ef- AM, Foged NT, Bru¨ nner N, Delaisse´JM2002 Synthetic fects by different biotechnological molecules. Int J Immu- matrix metalloproteinase inhibitors inhibit growth of es- nopathol Pharmacol 22:567–572 tablished breast cancer osteolytic lesions and prolong sur- 219. Pettit AR, Ji H, von Stechow D, Mu¨ ller R, Goldring SR, vival in mice. Clin Cancer Res 8:1932–1939 Choi Y, Benoist C, Gravallese EM 2001 TRANCE/ 203. Le Gall C, Bonnelye E, Cle´zardin P 2008 Cathepsin K in- RANKL knockout mice are protected from bone erosion in hibitors as treatment of bone metastasis. Curr Opin Sup- a serum transfer model of arthritis. Am J Pathol 159:1689– port Palliat Care 2:218–222 1699 204. Le Gall C, Bellahce`ne A, Bonnelye E, Gasser JA, Cas- 220. Romas E, Sims NA, Hards DK, Lindsay M, Quinn JW, tronovo V, Green J, Zimmermann J, Cle´zardin P 2007 A Ryan PF, Dunstan CR, Martin TJ, Gillespie MT 2002 Os- cathepsin K inhibitor reduces breast cancer induced oste- teoprotegerin reduces osteoclast numbers and prevents olysis and skeletal tumor burden. Cancer Res 67:9894– bone erosion in collagen-induced arthritis. Am J Pathol 9902 161:1419–1427 205. Coussens LM, Fingleton B, Matrisian LM 2002 Matrix 221. Redlich K, Hayer S, Ricci R, David JP, Tohidast-Akrad M, metalloproteinase inhibitors and cancer: trials and tribu- Kollias G, Steiner G, Smolen JS, Wagner EF, Schett G 2002 lations. Science 295:2387–2392 Osteoclasts are essential for TNF-␣-mediated joint de- 206. Pavlaki M, Zucker S 2003 Matrix metalloproteinase in- struction. J Clin Invest 110:1419–1427 hibitors (MMPIs): the beginning of phase I or the termi- 222. Cohen SB, Dore RK, Lane NE, Ory PA, Peterfy CG, Sharp nation of phase III clinical trials. Cancer Metastasis Rev JT, van der Heijde D, Zhou L, Tsuji W, Newmark R 2008 22:177–203 Denosumab treatment effects on structural damage, bone 207. Coleman R, Gnant M 2009 New results from the use of mineral density, and bone turnover in rheumatoid arthri- bisphosphonates in cancer patients. Curr Opin Support tis: a twelve-month, multicenter, randomized, double- Palliat Care 3:213–218 blind, placebo-controlled, phase II clinical trial. Arthritis 208. Machado M, Cruz LS, Tannus G, Fonseca M 2009 Effi- Rheum 58:1299–1309 cacy of clodronate, pamidronate, and zoledronate in re- 223. Zwerina J, Redlich K, Polzer K, Joosten L, Kro¨ nke G, ducing morbidity and mortality in cancer patients with Distler J, Hess A, Pundt N, Pap T, Hoffmann O, Gasser J, bone metastasis: a meta-analysis of randomized clinical Scheinecker C, Smolen JS, van den Berg W, Schett G 2007 trials. Clin Ther 31:962–979 TNF-induced structural joint damage is mediated by IL-1. 209. Fizazi K, Lipton A, Mariette X, Body JJ, Rahim Y, Gralow Proc Natl Acad Sci USA 104:11742–11747 JR, Gao G, Wu L, Sohn W, Jun S 2009 Randomized phase 224. Jarrett SJ, Conaghan PG, Sloan VS, Papanastasiou P, Ort- II trial of denosumab in patients with bone metastases from mann CE, O’Connor PJ, Grainger AJ, Emery P 2006 Pre- prostate cancer, breast cancer, or other neoplasms after liminary evidence for a structural benefit of the new intravenous bisphosphonates. J Clin Oncol 27:1564–1571 bisphosphonate zoledronic acid in early rheumatoid ar- 210. Kearns AE, Khosla S, Kostenuik PJ 2008 Receptor activa- thritis. Arthritis Rheum 54:1410–1414 tor of nuclear factor ␬B ligand and osteoprotegerin regu- 225. Breuil V, Euller-Ziegler L 2006 Bisphosphonate therapy in lation of bone remodeling in health and disease. Endocr rheumatoid arthritis. Joint Bone Spine 73:349–354 Rev 29:155–192 226. Lems WF, Lodder MC, Lips P, Bijlsma JW, Geusens P, 211. Rachner TD, Singh SK, Schoppet M, Benad P, Bornha¨user Schrameijer N, van de Ven CM, Dijkmans BA 2006 Pos- M, Ellenrieder V, Ebert R, Jakob F, Hofbauer LC 2010 itive effect of alendronate on bone mineral density and Zoledronic acid induces apoptosis and changes the TRAIL/ markers of bone turnover in patients with rheumatoid ar- OPG ratio in breast cancer cells. Cancer Lett 287:109–116 thritis on chronic treatment with low-dose prednisone: a 212. Schett G, Teitelbaum SL 2009 Osteoclasts and arthritis. randomized, double-blind, placebo-controlled trial. Os- J Bone Miner Res 24:1142–1146 teoporos Int 17:716–723 213. Schett G 2007 Erosive arthritis. Arthritis Res Ther 9(Suppl 227. Reid DM, Devogelaer JP, Saag K, Roux C, Lau CS, Re- 1):S2 ginster JY, Papanastasiou P, Ferreira A, Hartl F, Fashola T, 214. Gravallese EM, Harada Y, Wang JT, Gorn AH, Thornhill Mesenbrink P, Sambrook PN 2009 Zoledronic acid and TS, Goldring SR 1998 Identification of cell types respon- risedronate in the prevention and treatment of glucocor- sible for bone resorption in rheumatoid arthritis and juve- ticoid-induced osteoporosis (HORIZON): a multicentre, nile rheumatoid arthritis. Am J Pathol 152:943–951 double-blind, double-dummy, randomised controlled 215. Feldmann M, Brennan FM, Maini RN 1996 Role of cy- trial. Lancet 373:1253–1263 tokines in rheumatoid arthritis. Annu Rev Immunol 14: 228. Sims NA, Green JR, Glatt M, Schlict S, Martin TJ, Gillespie 397–440 MT, Romas E 2004 Targeting osteoclasts with zoledronic 216. Keffer J, Probert L, Cazlaris H, Georgopoulos S, Kaslaris acid prevents bone destruction in collagen-induced arthri- E, Kioussis D, Kollias G 1991 Transgenic mice expressing tis. Arthritis Rheum 50:2338–2346 human tumour necrosis factor: a predictive genetic model 229. Herrak P, Go¨ rtz B, Hayer S, Redlich K, Reiter E, Gasser J, of arthritis. EMBO J 10:4025–4031 Bergmeister H, Kollias G, Smolen JS, Schett G 2004 217. Singh JA, Christensen R, Wells GA, Suarez-Almazor ME, Zoledronic acid protects against local and systemic bone Buchbinder R, Lopez-Olivo MA, Ghogomu ET, Tugwell P loss in tumor necrosis factor-mediated arthritis. Arthritis 2009 A network meta-analysis of randomized controlled Rheum 50:2327–2337 58 Henriksen et al. Osteoclast Subtypes Endocrine Reviews, February 2011, 32(1):31–63

230. Morko JP, So¨ derstro¨ m M, Sa¨a¨ma¨nen AM, Salminen HJ, modeling in patients with osteoporosis. J Clin Invest 100: Vuorio EI 2004 Up regulation of cathepsin K expression in 1475–1480 articular chondrocytes in a transgenic mouse model for 244. Recker RR, Weinstein RS, Chesnut 3rd CH, Schimmer RC, osteoarthritis. Ann Rheum Dis 63:649–655 Mahoney P, Hughes C, Bonvoisin B, Meunier PJ 2004 231. Morko J, Kiviranta R, Joronen K, Sa¨a¨ma¨nen AM, Vuorio Histomorphometric evaluation of daily and intermittent E, Salminen-Mankonen H 2005 Spontaneous develop- oral ibandronate in women with postmenopausal osteo- ment of synovitis and cartilage degeneration in transgenic porosis: results from the BONE study. Osteoporos Int 15: mice overexpressing cathepsin K. Arthritis Rheum 52: 231–237 3713–3717 245. Recker RR, Delmas PD, Halse J, Reid IR, Boonen S, 232. Schurigt U, Hummel KM, Petrow PK, Gajda M, Sto¨ ckigt García-Hernandez PA, Supronik J, Lewiecki EM, Ochoa R, Middel P, Zwerina J, Janik T, Bernhardt R, Schu¨ ler S, L, Miller P, Hu H, Mesenbrink P, Hartl F, Gasser J, Eriksen Scharnweber D, Beckmann F, Saftig P, Kollias G, Schett G, EF 2008 Effects of intravenous zoledronic acid once yearly Wiederanders B, Bra¨uer R 2008 Cathepsin K deficiency on bone remodeling and bone structure. J Bone Miner Res partially inhibits, but does not prevent, bone destruction in 23:6–16 human tumor necrosis factor-transgenic mice. Arthritis 246. Nenonen A, Cheng S, Ivaska KK, Alatalo SL, Lehtimaki T, Rheum 58:422–434 Schmidt-Gayk H, Uusi-Rasi K, Heinonen A, Kannus P, 233. Hou WS, Li Z, Gordon RE, Chan K, Klein MJ, Levy R, Sievanen H, Vuori I, Vaananen HK, Halleen JM 2005 Se- Keysser M, Keyszer G, Bro¨ mme D 2001 Cathepsin K is a rum TRACP 5b is a useful marker for monitoring alen- critical protease in synovial fibroblast-mediated collagen dronate treatment: comparison with other markers of bone degradation. Am J Pathol 159:2167–2177 turnover. J Bone Miner Res 20:1804–1812 234. Yasuda Y, Kaleta J, Bro¨ mme D 2005 The role of cathepsins 247. Hannon RA, Clowes JA, Eagleton AC, Al Hadari A, Eastell in osteoporosis and arthritis: rationale for the design of R, Blumsohn A 2004 Clinical performance of immunore- new therapeutics. Adv Drug Deliv Rev 57:973–993 active tartrate-resistant acid phosphatase isoform 5b as a 235. Ainola M, Valleala H, Nyka¨nen P, Risteli J, Hanemaaijer marker of bone resorption. Bone 34:187–194 R, Konttinen YT 2008 Erosive arthritis in a patient with 248. Mun˜ oz-Torres M, Reyes-García R, Mezquita-Raya P, Fer- pycnodysostosis: an experiment of nature. Arthritis na´ndez-García D, Alonso G, Luna Jde D, Ruiz-Requena Rheum 58:3394–3401 ME, Escobar-Jime´nez F 2009 Serum cathepsin K as a 236. Svelander L, Erlandsson-Harris H, Astner L, Grabowska marker of bone metabolism in postmenopausal women U, Klareskog L, Lindstrom E, Hewitt E 2009 Inhibition of treated with alendronate. Maturitas 64:188–192 cathepsin K reduces bone erosion, cartilage degradation 249. D’Amelio P, Grimaldi A, Di Bella S, Tamone C, Brianza and inflammation evoked by collagen-induced arthritis in SZ, Ravazzoli MG, Bernabei P, Cristofaro MA, Pescar- mice. Eur J Pharmacol 613:155–162 mona GP, Isaia G 2008 Risedronate reduces osteoclast 237. Salminen-Mankonen HJ, Morko J, Vuorio E 2007 Role of precursors and cytokine production in postmenopausal os- cathepsin K in normal joints and in the development of teoporotic women. J Bone Miner Res 23:373–379 arthritis. Curr Drug Targets 8:315–323 250. D’Amelio P, Grimaldi A, Cristofaro MA, Ravazzoli M, 238. Hakala M, Risteli J, Aman S, Kautiainen H, Korpela M, Molinatti PA, Pescarmona GP, Isaia GC 1 December 2009 Hannonen P, Leirisalo-Repo M, Laasonen L, Paimela L, Alendronate reduces osteoclast precursors in osteoporosis. Mo¨ tto¨ nen T 2008 Combination drug strategy in recent- Osteoporos Int 10.1007/s00198-009-1129-1 onset rheumatoid arthritis suppresses collagen I degrada- 251. Weinstein RS, Roberson PK, Manolagas SC 2009 Giant tion and is associated with retardation of radiological pro- osteoclast formation and long-term oral bisphosphonate gression. Scand J Rheumatol 37:90–93 therapy. N Engl J Med 360:53–62 239. Sassi ML, Aman S, Hakala M, Luukkainen R, Risteli J 252. Mori S, Harruff R, Ambrosius W, Burr DB 1997 Trabec- 2003 Assay for cross-linked carboxyterminal telopeptide ular bone volume and microdamage accumulation in the of type I collagen (ICTP) unlike CrossLaps assay reflects femoral heads of women with and without femoral neck increased pathological degradation of type I collagen in fractures. Bone 21:521–526 rheumatoid arthritis. Clin Chem Lab Med 41:1038–1044 253. Chapurlat RD, Arlot M, Burt-Pichat B, Chavassieux P, 240. Chopin F, Garnero P, le Henanff A, Debiais F, Daragon A, Roux JP, Portero-Muzy N, Delmas PD 2007 Microcrack Roux C, Sany J, Wendling D, Zarnitsky C, Ravaud P, frequency and bone remodeling in postmenopausal osteo- Thomas T 2008 Long-term effects of infliximab on bone porotic women on long-term bisphosphonates: a bone bi- and cartilage turnover markers in patients with rheuma- opsy study. J Bone Miner Res 22:1502–1509 toid arthritis. Ann Rheum Dis 67:353–357 254. Burr DB, Allen MR 2008 Low bone turnover and micro- 241. Kadono Y, Tanaka S, Nishino J, Nishimura K, Nakamura damage? How and where to assess it? J Bone Miner Res I, Miyazaki T, Takayanagi H, Nakamura K 2009 Rheu- 23:1150–1151; author reply 1152–1153 matoid arthritis associated with osteopetrosis. Mod Rheu- 255. Ravn P, Hosking D, Thompson D, Cizza G, Wasnich RD, matol 19:687–690 McClung M, Yates AJ, Bjarnason NH, Christiansen C 242. Russell RG, Watts NB, Ebetino FH, Rogers MJ 2008 1999 Monitoring of alendronate treatment and prediction Mechanisms of action of bisphosphonates: similarities and of effect on bone mass by biochemical markers in the early differences and their potential influence on clinical effi- postmenopausal intervention cohort study. J Clin Endo- cacy. Osteoporos Int 19:733–759 crinol Metab 84:2363–2368 243. Chavassieux PM, Arlot ME, Reda C, Wei L, Yates AJ, 256. Ravn P, Clemmesen B, Christiansen C 1999 Biochemical Meunier PJ 1997 Histomorphometric assessment of the markers can predict the response in bone mass during alen- long-term effects of alendronate on bone quality and re- dronate treatment in early postmenopausal women. Alen- Endocrine Reviews, February 2011, 32(1):31–63 edrv.endojournals.org 59

dronate Osteoporosis Prevention Study Group. Bone 24: and raloxifene decrease the formation of multinucleate 237–244 cells in human bone marrow cultures. Eur Cytokine Netw 257. Ravn P, Thompson DE, Ross PD, Christiansen C 2003 13:39–45 Biochemical markers for prediction of 4-year response in 271. Saintier D, Burde MA, Rey JM, Maudelonde T, de Ver- bone mass during bisphosphonate treatment for preven- nejoul MC, Cohen-Solal ME 2004 17␤-Estradiol down- tion of postmenopausal osteoporosis. Bone 33:150–158 regulates ␤3-integrin expression in differentiating and ma- 258. Eriksen EF, Melsen F, Sod E, Barton I, Chines A 2002 ture human osteoclasts. J Cell Physiol 198:269–276 Effects of long-term risedronate on bone quality and bone 272. Michael H, Ha¨rko¨ nen PL, Va¨a¨na¨nen HK, Hentunen TA turnover in women with postmenopausal osteoporosis. 2005 Estrogen and testosterone use different cellular path- Bone 31:620–625 ways to inhibit osteoclastogenesis and bone resorption. 259. Black DM, Schwartz AV, Ensrud KE, Cauley JA, Levis S, J Bone Miner Res 20:2224–2232 Quandt SA, Satterfield S, Wallace RB, Bauer DC, Palermo 273. Michael H, Ha¨rko¨ nen PL, Kangas L, Va¨a¨na¨nen HK, Hen- L, Wehren LE, Lombardi A, Santora AC, Cummings SR tunen TA 2007 Differential effects of selective oestrogen 2006 Effects of continuing or stopping alendronate after 5 receptor modulators (SERMs) tamoxifen, ospemifene and years of treatment: the Fracture Intervention Trial Long- raloxifene on human osteoclasts in vitro. Br J Pharmacol term Extension (FLEX): a randomized trial. JAMA 296: 151:384–395 2927–2938 274. Taranta A, Brama M, Teti A, De luca V, Scandurra R, 260. Durie BG, Katz M, Crowley J 2005 Osteonecrosis of the Spera G, Agnusdei D, Termine JD, Migliaccio S 2002 The jaw and bisphosphonates. N Engl J Med 353:99–102 selective estrogen receptor modulator raloxifene regulates 261. Watts NB, Diab DL 2010 Long-term use of bisphospho- osteoclast and osteoblast activity in vitro. Bone 30:368– nates in osteoporosis. J Clin Endocrinol Metab 95:1555– 376 1565 275. Oursler MJ, Osdoby P, Pyfferoen J, Riggs BL, Spelsberg 262. Khosla S, Burr D, Cauley J, Dempster DW, Ebeling PR, TC 1991 Avian osteoclasts as estrogen target cells. Proc Felsenberg D, Gagel RF, Gilsanz V, Guise T, Koka S, Natl Acad Sci USA 88:6613–6617 McCauley LK, McGowan J, McKee MD, Mohla S, Pendrys 276. Oursler MJ, Pederson L, Fitzpatrick L, Riggs BL, Spelsberg DG, Raisz LG, Ruggiero SL, Shafer DM, Shum L, Silverman T 1994 Human giant cell tumors of the bone (osteoclas- SL, Van Poznak CH, Watts N, Woo SB, Shane E 2007 tomas) are estrogen target cells. Proc Natl Acad Sci USA Bisphosphonate-associated osteonecrosis of the jaw: re- 91:5227–5231 port of a task force of the American Society for Bone and 277. Oursler MJ, Pederson L, Pyfferoen J, Osdoby P, Fitzpatrick Mineral Research. J Bone Miner Res 22:1479–1491 L, Spelsberg TC 1993 Estrogen modulation of avian osteo- 263. Cheng A, Daly CG, Logan RM, Stein B, Goss AN 2009 clast lysosomal gene expression. Endocrinology 132: Alveolar bone and the bisphosphonates. Aust Dent J 1373–1380 54(Suppl 1):S51–S61 278. Kremer M, Judd J, Rifkin B, Auszmann J, Oursler MJ 1995 264. Tran Van PT, Vignery A, Baron R 1982 Cellular kinetics Estrogen modulation of osteoclast lysosomal enzyme se- of the bone remodeling sequence in the rat. Anat Rec 202: cretion. J Cell Biochem 57:271–279 445–451 279. Parikka V, Lehenkari P, Sassi ML, Halleen J, Risteli J, 265. Tran Van P, Vignery A, Baron R 1982 An electron-micro- Ha¨rko¨ nen P, Va¨a¨na¨nen HK 2001 Estrogen reduces the scopic study of the bone-remodeling sequence in the rat. depth of resorption pits by disturbing the organic bone Cell Tissue Res 225:283–292 matrix degradation activity of mature osteoclasts. Endo- 266. Baron R, Neff L, Tran Van P, Nefussi JR, Vignery A 1986 crinology 142:5371–5378 Kinetic and cytochemical identification of osteoclast pre- 280. Ott SM, Oleksik A, Lu Y, Harper K, Lips P 2002 Bone cursors and their differentiation into multinucleated oste- histomorphometric and biochemical marker results of a oclasts. Am J Pathol 122:363–378 2-year placebo-controlled trial of raloxifene in postmeno- 267. Favia G, Pilolli GP, Maiorano E 2009 Histologic and his- pausal women. J Bone Miner Res 17:341–348 tomorphometric features of bisphosphonate-related osteo- 281. Steiniche T, Hasling C, Charles P, Eriksen EF, Mosekilde necrosis of the jaws: an analysis of 31 cases with confocal L, Melsen F 1989 A randomized study on the effects of laser scanning microscopy. Bone 45:406–413 estrogen/gestagen or high dose oral calcium on trabecular 268. Bedogni A, Blandamura S, Lokmic Z, Palumbo C, Ragazzo bone remodeling in postmenopausal osteoporosis. Bone M, Ferrari F, Tregnaghi A, Pietrogrande F, Procopio O, 10:313–320 Saia G, Ferretti M, Bedogni G, Chiarini L, Ferronato G, 282. Patel S, Pazianas M, Tobias J, Chambers TJ, Fox S, Chow Ninfo V, Lo Russo L, Lo Muzio L, Nocini PF 2008 J 1999 Early effects of hormone replacement therapy on Bisphosphonate-associated jawbone osteonecrosis: a cor- bone. Bone 24:245–248 relation between imaging techniques and histopathology. 283. Eriksen EF, Langdahl B, Vesterby A, Rungby J, Kassem M Oral Surg Oral Med Oral Pathol Oral Radiol Endod 105: 1999 Hormone replacement therapy prevents osteoclastic 358–364 hyperactivity: a histomorphometric study in early post- 269. Hansen T, Kunkel M, Weber A, James Kirkpatrick C 2006 menopausal women. J Bone Miner Res 14:1217–1221 Osteonecrosis of the jaws in patients treated with 284. Vedi S, Purdie DW, Ballard P, Bord S, Cooper AC, Comp- bisphosphonates—histomorphologic analysis in com- ston JE 1999 Bone remodeling and structure in postmeno- parison with infected osteoradionecrosis. J Oral Pathol pausal women treated with long-term, high-dose estrogen Med 35:155–160 therapy. Osteoporos Int 10:52–58 270. Ramalho AC, Couttet P, Baudoin C, Morieux C, Graulet 285. Ettinger B, Black DM, Mitlak BH, Knickerbocker RK, AM, de Vernejoul MC, Cohen-Solal ME 2002 Estradiol Nickelsen T, Genant HK, Christiansen C, Delmas PD, 60 Henriksen et al. Osteoclast Subtypes Endocrine Reviews, February 2011, 32(1):31–63

Zanchetta JR, Stakkestad J, Glu¨ er CC, Krueger K, Cohen 298. Jiang Y, Zhao J, Geusens P, Liao EY, Adriaensens P, Gelan FJ, Eckert S, Ensrud KE, Avioli LV, Lips P, Cummings SR J, Azria M, Boonen S, Caulin F, Lynch JA, Ouyang X, 1999 Reduction of vertebral fracture risk in postmeno- Genant HK 2005 Femoral neck trabecular microstructure pausal women with osteoporosis treated with raloxifene: in ovariectomized ewes treated with calcitonin: MRI mi- results from a 3-year randomized clinical trial. Multiple croscopic evaluation. J Bone Miner Res 20:125–130 Outcomes of Raloxifene Evaluation (MORE) Investiga- 299. Hoff AO, Catala-Lehnen P, Thomas PM, Priemel M, tors. JAMA 282:637–645 Rueger JM, Nasonkin I, Bradley A, Hughes MR, Ordonez 286. Meunier PJ, Vignot E, Garnero P, Confavreux E, Paris E, N, Cote GJ, Amling M, Gagel RF 2002 Increased bone Liu-Leage S, Sarkar S, Liu T, Wong M, Draper MW 1999 mass is an unexpected phenotype associated with deletion Treatment of postmenopausal women with osteoporosis of the calcitonin gene. J Clin Invest 110:1849–1857 or low bone density with raloxifene. Raloxifene Study 300. Dacquin R, Davey RA, Laplace C, Levasseur R, Morris Group. Osteoporos Int 10:330–336 HA, Goldring SR, Gebre-Medhin S, Galson DL, Zajac JD, 287. Lufkin EG, Whitaker MD, Nickelsen T, Argueta R, Caplan Karsenty G 2004 Amylin inhibits bone resorption while the RH, Knickerbocker RK, Riggs BL 1998 Treatment of es- calcitonin receptor controls bone formation in vivo. J Cell tablished postmenopausal osteoporosis with raloxifene: a Biol 164:509–514 randomized trial. J Bone Miner Res 13:1747–1754 301. Davey RA, Turner AG, McManus JF, Chiu WS, Tjahyono 288. Chambers TJ, Moore A 1983 The sensitivity of isolated F, Moore AJ, Atkins GJ, Anderson PH, Ma C, Glatt V, osteoclasts to morphological transformation by calcitonin. MacLean HE, Vincent C, Bouxsein M, Morris HA, Findlay J Clin Endocrinol Metab 57:819–824 DM, Zajac JD 2008 Calcitonin receptor plays a physio- 289. Suzuki H, Nakamura I, Takahashi N, Ikuhara T, Matsu- logical role to protect against hypercalcemia in mice. zaki K, Isogai Y, Hori M, Suda T 1996 Calcitonin-induced J Bone Miner Res 23:1182–1193 changes in the cytoskeleton are mediated by a signal path- 302. Turner C, Tjahyono A, Moore A, Findlay D, Morris H, way associated with protein kinase A in osteoclasts. En- Zajac J, Davey R 2009 The calcitonin receptor expressed docrinology 137:4685–4690 by osteoclasts plays a biological role to protect against 290. Shyu JF, Shih C, Tseng CY, Lin CH, Sun DT, Liu HT, induced hypercalcemia in mice. J Bone Miner Res 24(Suppl Tsung HC, Chen TH, Lu RB 2007 Calcitonin induces po- 1):1049 (Abstract) dosome disassembly and detachment of osteoclasts by 303. Karsdal MA, Byrjalsen I, Leeming DJ, Delmas PD, Chris- modulating Pyk2 and Src activities. Bone 40:1329–1342 tiansen C 2008 The effects of oral calcitonin on bone col- 291. Karsdal MA, Henriksen K, Bay-Jensen AC, Molloy B, Ar- lagen maturation: implications for bone turnover and nold M, John MR, Byrjalsen I, Azria M, Riis BJ, Qvist P, quality. Osteoporos Int 19:1355–1361 Christiansen C 2010 Lessons learned from the develop- 304. Holtrop ME, King GJ, Cox KA, Reit B 1979 Time-related ment of oral calcitonin: the first tablet formulation of a changes in the ultrastructure of osteoclasts after injection peptide in phase III clinical trials. J Clin Pharmacol of parathyroid hormone in young rats. Calcif Tissue Int 10.1177/0091270010372625 27:129–135 292. Karsdal MA, Byrjalsen I, Riis BJ, Christiansen C 2008 305. Ma YL, Cain RL, Halladay DL, Yang X, Zeng Q, Miles Investigation of the diurnal variation in bone resorption for RR, Chandrasekhar S, Martin TJ, Onyia JE 2001 Cata- optimal drug delivery and efficacy in osteoporosis with bolic effects of continuous human PTH (1–38) in vivo is oral calcitonin. BMC Clin Pharmacol 8:12 associated with sustained stimulation of RANKL and in- 293. Kung AW, Pasion EG, Sofiyan M, Lau EM, Tay BK, Lam hibition of osteoprotegerin and gene-associated bone for- KS, Wilawan K, Ongphiphadhanakul B, Thiebaud D 2006 mation. Endocrinology 142:4047–4054 A comparison of teriparatide and calcitonin therapy in 306. Hodsman AB, Kisiel M, Adachi JD, Fraher LJ, Watson PH postmenopausal Asian women with osteoporosis: a 2000 Histomorphometric evidence for increased bone 6-month study. Curr Med Res Opin 22:929–937 turnover without change in cortical thickness or porosity 294. Hwang JS, Tu ST, Yang TS, Chen JF, Wang CJ, Tsai KS after 2 years of cyclical hPTH(1–34) therapy in women 2006 Teriparatide vs. calcitonin in the treatment of Asian with severe osteoporosis. Bone 27:311–318 postmenopausal women with established osteoporosis. 307. Arlot M, Meunier PJ, Boivin G, Haddock L, Tamayo J, Osteoporos Int 17:373–378 Correa-Rotter R, Jasqui S, Donley DW, Dalsky GP, Martin 295. Trovas GP, Lyritis GP, Galanos A, Raptou P, Constante- JS, Eriksen EF 2005 Differential effects of teriparatide and lou E 2002 A randomized trial of nasal spray salmon cal- alendronate on bone remodeling in postmenopausal citonin in men with idiopathic osteoporosis: effects on women assessed by histomorphometric parameters. J Bone bone mineral density and bone markers. J Bone Miner Res Miner Res 20:1244–1253 17:521–527 308. Recker RR, Marin F, Ish-Shalom S, Mo¨ ricke R, Hawkins 296. Chesnut 3rd CH, Majumdar S, Newitt DC, Shields A, Van F, Kapetanos G, de la Pen˜ a MP, Kekow J, Farrerons J, Sanz Pelt J, Laschansky E, Azria M, Kriegman A, Olson M, B, Oertel H, Stepan J 2009 Comparative effects of teripa- Eriksen EF, Mindeholm L 2005 Effects of salmon calcito- ratide and strontium ranelate on bone biopsies and bio- nin on trabecular microarchitecture as determined by mag- chemical markers of bone turnover in postmenopausal netic resonance imaging: results from the QUEST study. women with osteoporosis. J Bone Miner Res 24:1358– J Bone Miner Res 20:1548–1561 1368 297. Ikegame M, Ejiri S, Ozawa H 2004 Calcitonin-induced 309. Dobnig H, Turner RT 1995 Evidence that intermittent change in serum calcium levels and its relationship to os- treatment with parathyroid hormone increases bone for- teoclast morphology and number of calcitonin receptors. mation in adult rats by activation of bone lining cells. En- Bone 35:27–33 docrinology 136:3632–3638 Endocrine Reviews, February 2011, 32(1):31–63 edrv.endojournals.org 61

310. Jilka RL, Weinstein RS, Bellido T, Roberson P, Parfitt AM, ric and microCT analysis of bone biopsies from postmeno- Manolagas SC 1999 Increased bone formation by preven- pausal osteoporotic women treated with strontium rane- tion of osteoblast apoptosis with parathyroid hormone. late. J Bone Miner Res 23:215–222 J Clin Invest 104:439–446 323. Meunier PJ, Slosman DO, Delmas PD, Sebert JL, Brandi 311. Luiz de Freitas PH, Li M, Ninomiya T, Nakamura M, ML, Albanese C, Lorenc R, Pors-Nielsen S, De Vernejoul Ubaidus S, Oda K, Udagawa N, Maeda T, Takagi R, Ami- MC, Roces A, Reginster JY 2002 Strontium ranelate: dose- zuka N 2009 Intermittent PTH administration stimulates dependent effects in established postmenopausal vertebral pre-osteoblastic proliferation without leading to enhanced osteoporosis—a 2-year randomized placebo controlled bone formation in osteoclast-less c-fos(Ϫ/Ϫ) mice. J Bone trial. J Clin Endocrinol Metab 87:2060–2066 Miner Res 24:1586–1597 324. Axmann R, Böhm C, Krönke G, Zwerina J, Smolen J, 312. Black DM, Greenspan SL, Ensrud KE, Palermo L, Schett G 2009 Inhibition of interleukin-6 receptor directly McGowan JA, Lang TF, Garnero P, Bouxsein ML, Bilez- blocks osteoclast formation in vitro and in vivo. Arthritis ikian JP, Rosen CJ 2003 The effects of parathyroid hor- Rheum 60:2747–2756 mone and alendronate alone or in combination in post- 325. Bruye`re O, Collette J, Rizzoli R, Decock C, Ortolani S, menopausal osteoporosis. N Engl J Med 349:1207–1215 Cormier C, Detilleux J, Reginster JY 2010 Relationship 313. Finkelstein JS, Hayes A, Hunzelman JL, Wyland JJ, Lee H, between 3-month changes in biochemical markers of bone Neer RM 2003 The effects of parathyroid hormone, alen- remodelling and changes in bone mineral density and frac- dronate, or both in men with osteoporosis. N Engl J Med ture incidence in patients treated with strontium ranelate 349:1216–1226 for 3 years. Osteoporos Int 21:1031–1036 314. Ettinger B, San Martin J, Crans G, Pavo I 2004 Differential 326. Bonnelye E, Chabadel A, Saltel F, Jurdic P 2008 Dual effect effects of teriparatide on BMD after treatment with ralox- of strontium ranelate: stimulation of osteoblast differen- ifene or alendronate. J Bone Miner Res 19:745–751 tiation and inhibition of osteoclast formation and resorp- 315. Johnston S, Andrews S, Shen V, Cosman F, Lindsay R, tion in vitro. Bone 42:129–138 Dempster DW, Iida-Klein A 2007 The effects of combina- 327. Canalis E, Hott M, Deloffre P, Tsouderos Y, Marie PJ 1996 tion of alendronate and human parathyroid hormone(1- The divalent strontium salt S12911 enhances bone cell rep- 34) on bone strength are synergistic in the lumbar vertebra lication and bone formation in vitro. Bone 18:517–523 and additive in the femur of C57BL/6J mice. Endocrinol- 328. Takahashi N, Sasaki T, Tsouderos Y, Suda T 2003 S ogy 148:4466–4474 12911–2 inhibits osteoclastic bone resorption in vitro. 316. Samadfam R, Xia Q, Goltzman D 2007 Co-treatment of J Bone Miner Res 18:1082–1087 PTH with osteoprotegerin or alendronate increases its an- 329. Hurtel-Lemaire AS, Mentaverri R, Caudrillier A, Courna- abolic effect on the skeleton of oophorectomized mice. rie F, Wattel A, Kamel S, Terwilliger EF, Brown EM, Bra- J Bone Miner Res 22:55–63 zier M 2009 The calcium-sensing receptor is involved in 317. Meunier PJ, Roux C, Seeman E, Ortolani S, Badurski JE, strontium ranelate-induced osteoclast apoptosis. New in- Spector TD, Cannata J, Balogh A, Lemmel EM, Pors- sights into the associated signaling pathways. J Biol Chem Nielsen S, Rizzoli R, Genant HK, Reginster JY 2004 The 284:575–584 effects of strontium ranelate on the risk of vertebral frac- 330. Brennan TC, Rybchyn MS, Green W, Atwa S, Conigrave ture in women with postmenopausal osteoporosis. N Engl AD, Mason RS 2009 Osteoblasts play key roles in the J Med 350:459–468 mechanisms of action of strontium ranelate. Br J Pharma- 318. Seeman E, Vellas B, Benhamou C, Aquino JP, Semler J, col 157:1291–1300 Kaufman JM, Hoszowski K, Varela AR, Fiore C, Brixen K, 331. Engvall IL, Svensson B, Tengstrand B, Brismar K, Haf- Reginster JY, Boonen S 2006 Strontium ranelate reduces stro¨m I 2008 Impact of low-dose prednisolone on bone the risk of vertebral and nonvertebral fractures in women synthesis and resorption in early rheumatoid arthritis: ex- eighty years of age and older. J Bone Miner Res 21:1113– periences from a two-year randomized study. Arthritis Res 1120 Ther 10:R128 319. Reginster JY, Seeman E, De Vernejoul MC, Adami S, 332. Caplan L, Saag KG 2009 Glucocorticoids and the risk of Compston J, Phenekos C, Devogelaer JP, Curiel MD, osteoporosis. Expert Opin Drug Saf 8:33–47 Sawicki A, Goemaere S, Sorensen OH, Felsenberg D, 333. Canalis E, Mazziotti G, Giustina A, Bilezikian JP 2007 Meunier PJ 2005 Strontium ranelate reduces the risk of Glucocorticoid-induced osteoporosis: pathophysiology nonvertebral fractures in postmenopausal women with os- and therapy. Osteoporos Int 18:1319–1328 teoporosis: Treatment of Peripheral Osteoporosis (TRO- 334. van Brussel MS, Bultink IE, Lems WF 2009 Prevention of POS) study. J Clin Endocrinol Metab 90:2816–2822 glucocorticoid-induced osteoporosis. Expert Opin Phar- 320. Seeman E, Boonen S, Borgstro¨ m F, Vellas B, Aquino JP, macother 10:997–1005 Semler J, Benhamou CL, Kaufman JM, Reginster JY 2010 335. Weinstein RS, Jilka RL, Parfitt AM, Manolagas SC 1998 Five years treatment with strontium ranelate reduces ver- Inhibition of osteoblastogenesis and promotion of apopto- tebral and nonvertebral fractures and increases the number sis of osteoblasts and osteocytes by glucocorticoids. Po- and quality of remaining life-years in women over 80 years tential mechanisms of their deleterious effects on bone. of age. Bone 46:1038–1042 J Clin Invest 102:274–282 321. Ferrari S 2009 Continuous broad protection against os- 336. Hofbauer LC, Zeitz U, Schoppet M, Skalicky M, Schu¨ ler teoporotic fractures with strontium ranelate. Rheumatol- C, Stolina M, Kostenuik PJ, Erben RG 2009 Prevention of ogy (Oxford) 48(Suppl 4):iv20–iv24 glucocorticoid-induced bone loss in mice by inhibition of 322. Arlot ME, Jiang Y, Genant HK, Zhao J, Burt-Pichat B, RANKL. Arthritis Rheum 60:1427–1437 Roux JP, Delmas PD, Meunier PJ 2008 Histomorphomet- 337. Jia D, O’Brien CA, Stewart SA, Manolagas SC, Weinstein 62 Henriksen et al. Osteoclast Subtypes Endocrine Reviews, February 2011, 32(1):31–63

RS 2006 Glucocorticoids act directly on osteoclasts to in- Rosen CJ, Beamer W, Majumdar S, Halloran BP 2002 crease their life span and reduce bone density. Endocrinol- Insulin-like growth factor I is required for the anabolic ogy 147:5592–5599 actions of parathyroid hormone on mouse bone. J Bone 338. Yao W, Cheng Z, Busse C, Pham A, Nakamura MC, Lane Miner Res 17:1570–1578 NE 2008 Glucocorticoid excess in mice results in early 351. Cummings SR, San Martin J, McClung MR, Siris ES, Eas- activation of osteoclastogenesis and adipogenesis and pro- tell R, Reid IR, Delmas P, Zoog HB, Austin M, Wang A, longed suppression of osteogenesis: a longitudinal study of Kutilek S, Adami S, Zanchetta J, Libanati C, Siddhanti S, gene expression in bone tissue from glucocorticoid-treated Christiansen C 2009 Denosumab for prevention of frac- mice. Arthritis Rheum 58:1674–1686 tures in postmenopausal women with osteoporosis. N Engl 339. Weinstein RS, Chen JR, Powers CC, Stewart SA, Landes J Med 361:756–765 RD, Bellido T, Jilka RL, Parfitt AM, Manolagas SC 2002 352. Kostenuik PJ, Nguyen HQ, McCabe J, Warmington KS, Promotion of osteoclast survival and antagonism of Kurahara C, Sun N, Chen C, Li L, Cattley RC, Van G, bisphosphonate-induced osteoclast apoptosis by glucocor- Scully S, Elliott R, Grisanti M, Morony S, Tan HL, Asun- ticoids. J Clin Invest 109:1041–1048 cion F, Li X, Ominsky MS, Stolina M, Dwyer D, Dougall 340. Kim HJ, Zhao H, Kitaura H, Bhattacharyya S, Brewer JA, WC, Hawkins N, Boyle WJ, Simonet WS, Sullivan JK 2009 Muglia LJ, Ross FP, Teitelbaum SL 2006 Glucocorticoids Denosumab, a fully human monoclonal antibody to suppress bone formation via the osteoclast. J Clin Invest RANKL, inhibits bone resorption and increases BMD in 116:2152–2160 knock-in mice that express chimeric (murine/human) 341. Sivagurunathan S, Muir MM, Brennan TC, Seale JP, RANKL. J Bone Miner Res 24:182–195 Mason RS 2005 Influence of glucocorticoids on human 353. Bone HG, Bolognese MA, Yuen CK, Kendler DL, Wang H, osteoclast generation and activity. J Bone Miner Res Liu Y, San Martin J 2008 Effects of denosumab on bone 20:390–398 mineral density and bone turnover in postmenopausal 342. Hofbauer LC, Gori F, Riggs BL, Lacey DL, Dunstan CR, women. J Clin Endocrinol Metab 93:2149–2157 Spelsberg TC, Khosla S 1999 Stimulation of osteoprote- 354. McClung MR, Lewiecki EM, Cohen SB, Bolognese MA, gerin ligand and inhibition of osteoprotegerin production Woodson GC, Moffett AH, Peacock M, Miller PD, Led- by glucocorticoids in human osteoblastic lineage cells: po- erman SN, Chesnut CH, Lain D, Kivitz AJ, Holloway DL, tential paracrine mechanisms of glucocorticoid-induced Zhang C, Peterson MC, Bekker PJ 2006 Denosumab in osteoporosis. Endocrinology 140:4382–4389 postmenopausal women with low bone mineral density. 343. Soares-Schanoski A, Go´ mez-Pin˜ a V, del Fresno C, Ro- N Engl J Med 354:821–831 dríguez-Rojas A, García F, Glaría A, Sa´nchez M, Vallejo- 355. Ominsky MS, Schroeder J, Jolette J, Smith SY, Farrell DJ, Cremades MT, Baos R, Fuentes-Prior P, Arnalich F, Atkinson JE, Kostenuik PJ 2007 Decreased bone turnover Lo´ pez-Collazo E 2007 6-Methylprednisolone down-regu- and porosity are associated with improved bone strength in lates IRAK-M in human and murine osteoclasts and boosts ovariectomized (OVX) cynomolgus monkeys treated with bone-resorbing activity: a putative mechanism for corti- denosumab, a fully human RANKL antibody. J Bone coid-induced osteoporosis. J Leukoc Biol 82:700–709 Miner Res 22:S126 (Abstract) 344. Søe K, Delaisse´JM30 April 2010 Glucocorticoids main- 356. Sassi ML, Eriksen H, Risteli L, Niemi S, Mansell J, Gowen tain human osteoclasts in the active mode of their resorp- M, Risteli J 2000 Immunochemical characterization of as- tion cycle. J Bone Miner Res 10.1002/jbmr.113 say for carboxyterminal telopeptide of human type I col- 345. Dovio A, Perazzolo L, Osella G, Ventura M, Termine A, lagen: loss of antigenicity by treatment with cathepsin K. Milano E, Bertolotto A, Angeli A 2004 Immediate fall of Bone 26:367–373 bone formation and transient increase of bone resorption 357. Fuller K, Lawrence KM, Ross JL, Grabowska UB, Shiroo in the course of high-dose, short-term glucocorticoid ther- M, Samuelsson B, Chambers TJ 2008 Cathepsin K inhib- apy in young patients with multiple sclerosis. J Clin En- itors prevent matrix-derived growth factor degradation by docrinol Metab 89:4923–4928 human osteoclasts. Bone 42:200–211 346. Minisola S, Del Fiacco R, Piemonte S, Iorio M, Mascia ML, 358. Chappard D, Libouban H, Mindeholm L, Basle´ MF, Leg- Fidanza F, Cipriani C, Raso I, Porfiri ML, Francucci CM, rand E, Audran M 2010 The cathepsin K inhibitor D’Erasmo E, Romagnoli E 2008 Biochemical markers in AAE581 induces morphological changes in osteoclasts of glucocorticoid-induced osteoporosis. J Endocrinol Invest treated patients. Microsc Res Tech 73:726–732 31:28–32 359. Adams CS, Shapiro IM 2003 Mechanisms by which ex- 347. Dalle Carbonare L, Bertoldo F, Valenti MT, Zenari S, tracellular matrix components induce osteoblast apopto- Zanatta M, Sella S, Giannini S, Cascio VL 2005 Histo- sis. Connect Tissue Res 44(Suppl 1):230–239 morphometric analysis of glucocorticoid-induced osteo- 360. Walker EC, McGregor NE, Poulton IJ, Pompolo S, Allan porosis. Micron 36:645–652 EH, Quinn JM, Gillespie MT, Martin TJ, Sims NA 2008 348. Stellon AJ, Webb A, Compston JE 1988 Bone histomor- Cardiotrophin-1 is an osteoclast-derived stimulus of bone phometry and structure in corticosteroid treated chronic formation required for normal bone remodeling. J Bone active hepatitis. Gut 29:378–384 Miner Res 23:2025–2032 349. Stoch SA, Saag KG, Greenwald M, Sebba AI, Cohen S, 361. Gottschalck IB, Jeppesen PB, Hartmann B, Holst JJ, Hen- Verbruggen N, Giezek H, West J, Schnitzer TJ 2009 Once- riksen DB 2008 Effects of treatment with glucagon-like weekly oral alendronate 70 mg in patients with glucocor- peptide-2 on bone resorption in colectomized patients with ticoid-induced bone loss: a 12-month randomized, place- distal ileostomy or jejunostomy and short-bowel syn- bo-controlled clinical trial. J Rheumatol 36:1705–1714 drome. Scand J Gastroenterol 43:1304–1310 350. Bikle DD, Sakata T, Leary C, Elalieh H, Ginzinger D, 362. Gottschalck IB, Jeppesen PB, Holst JJ, Henriksen DB 2008 Endocrine Reviews, February 2011, 32(1):31–63 edrv.endojournals.org 63

Reduction in bone resorption by exogenous glucagon-like in number and size of osteoclasts in response to calcium peptide-2 administration requires an intact gastrointesti- or phosphorus deficiency in the rat. Endocrinology 97: nal tract. Scand J Gastroenterol 43:929–937 283–289 363. Sørensen MG, Henriksen K, Neutzsky-Wulff AV, Dziegiel 372. Howard GA, Bottemiller BL, Turner RT, Rader JI, Bay- MH, Karsdal MA 2007 Diphyllin, a novel and naturally link DJ 1981 Parathyroid hormone stimulates bone for- potent V-ATPase inhibitor, abrogates acidification of the mation and resorption in organ culture: evidence for a cou- osteoclastic resorption lacunae and bone resorption. pling mechanism. Proc Natl Acad Sci USA 78:3204–3208 J Bone Miner Res 22:1640–1648 373. Lazowski DA, Fraher LJ, Hodsman A, Steer B, Modrowski 364. Schaller S, Henriksen K, Sveigaard C, Heegaard AM, He´lix D, Han VK 1994 Regional variation of insulin-like growth N, Stahlhut M, Ovejero MC, Johansen JV, Solberg H, factor-I gene expression in mature rat bone and cartilage. Andersen TL, Hougaard D, Berryman M, Shiødt CB, Sø- Bone 15:563–576 rensen BH, Lichtenberg J, Christophersen P, Foged NT, 374. Robinson JA, Riggs BL, Spelsberg TC, Oursler MJ 1996 Delaisse´ JM, Engsig MT, Karsdal MA 2004 The chloride Osteoclasts and transforming growth factor-␤: estrogen- channel inhibitor n53736 prevents bone resorption in mediated isoform-specific regulation of production. En- ovariectomized rats without changing bone formation. docrinology 137:615–621 J Bone Miner Res 19:1144–1153 375. Karsdal MA, Fjording MS, Foged NT, Delaisse´ JM, 365. Visentin L, Dodds RA, Valente M, Misiano P, Bradbeer Lochter A 2001 Transforming growth factor-␤-induced JN, Oneta S, Liang X, Gowen M, Farina C 2000 A selective osteoblast elongation regulates osteoclastic bone resorp- inhibitor of the osteoclastic V-H(ϩ)-ATPase prevents bone tion through a p38 mitogen-activated protein kinase- and loss in both thyroparathyroidectomized and ovariecto- matrix metalloproteinase-dependent pathway. J Biol mized rats. J Clin Invest 106:309–318 Chem 276:39350–39358 366. Rzeszutek K, Sarraf F, Davies JE 2003 Proton pump in- 376. Mundy GR, Bonewald LF 1990 Role of TGF ␤ in bone hibitors control osteoclastic resorption of calcium phos- phate implants and stimulate increased local reparative remodeling. Ann NY Acad Sci 593:91–97 bone growth. J Craniofac Surg 14:301–307 377. Baylink DJ, Finkelman RD, Mohan S 1993 Growth factors 367. Schaller S, Henriksen K, Sørensen MG, Karsdal MA 2005 to stimulate bone formation. J Bone Miner Res 8(Suppl The role of chloride channels in osteoclasts: ClC-7 as a 2):S565–S572 target for osteoporosis treatment. Drug News Perspect 18: 378. Hayden JM, Mohan S, Baylink DJ 1995 The insulin-like 489–495 growth factor system and the coupling of formation to 368. Brown EM 2007 The calcium-sensing receptor: physiol- resorption. Bone 17:93S–98S ogy, pathophysiology and CaR-based therapeutics. Sub- 379. Janssens K, ten Dijke P, Janssens S, Van Hul W 2005 Trans- cell Biochem 45:139–167 forming growth factor-␤1 to the bone. Endocr Rev 26: 369. Martin TJ 2005 Osteoblast-derived PTHrP is a physiolog- 743–774 ical regulator of bone formation. J Clin Invest 115:2322– 380. Zhang M, Xuan S, Bouxsein ML, von Stechow D, Akeno 2324 N, Faugere MC, Malluche H, Zhao G, Rosen CJ, Efstra- 370. Kramer I, Keller H, Leupin O, Kneissel M 2010 Does os- tiadis A, Clemens TL 2002 Osteoblast-specific knockout teocytic SOST suppression mediate PTH bone anabolism? of the insulin-like growth factor (IGF) receptor gene reveals Trends Endocrinol Metab 21:237–244 an essential role of IGF signaling in bone matrix mineral- 371. Thompson ER, Baylink DJ, Wergedal JE 1975 Increases ization. J Biol Chem 277:44005–44012 Rev Endocr Metab Disord (2010) 11:219–227 DOI 10.1007/s11154-010-9153-1

Cellular mechanisms of bone remodeling

Erik Fink Eriksen

Published online: 29 December 2010 # The Author(s) 2010. This article is published with open access at Springerlink.com

Abstract Bone remodeling is a tightly regulated process (RUNX, Osterix) are involved in osteoblast differentiation. securing repair of microdamage (targeted remodeling) and Both pathways are modulated by calcitropic hormones. replacement of old bone with new bone through sequential osteoclastic resorption and osteoblastic bone formation. The Keywords Osteoblasts . Osteoclasts . Lining cells . Growth rate of remodeling is regulated by a wide variety of factors . Cytokines . Bone remodeling . Osteoporosis . Bone calcitropic hormones (PTH, thyroid hormone, sex steroids remodeling compartment etc.). In recent years we have come to appreciate that bone remodeling proceeds in a specialized vascular structure,— the Bone Remodeling Compartment (BRC). The outer 1 Introduction lining of this compartment is made up of flattened cells, displaying all the characteristics of lining cells in bone Bone histomorphometry has given us great insights into including expression of OPG and RANKL. Reduced bone bone physiology and bone remodeling in particular. turnover leads to a decrease in the number of BRCs, while Histomorphometric indices obtained using tetracycline increased turnover causes an increase in the number of double labeling techniques are unique, because they BRCs. The secretion of regulatory factors inside a confined contrary to DXA and bone markers reflect cellular activity space separated from the bone marrow would facilitate of osteoclasts and osteoblasts using the incorporation of a local regulation of the remodeling process without interfer- time marker, namely spaced administration of an agent ence from growth factors secreted by blood cells in the (tetracycline) reflecting ongoing active bone formation. The marrow space. The BRC also creates an environment where study of bone remodeling originated with the classical cells inside the structure are exposed to denuded bone, works of Harold Frost 40 years ago [1] and our ever which may enable direct cellular interactions with integrins expanding understanding of this process is the basis for the and other matrix factors known to regulate osteoclast/ development of highly effective treatments for osteoporo- osteoblast activity. However, the denuded bone surface sis, that we have seen over the last 20 years. inside the BRC also constitutes an ideal environment for the seeding of bone metastases, known to have high affinity for bone matrix. Circulating osteoclast- and osteoblast 2 The bone remodeling cycle precursor cells have been demonstrated in peripheral blood. The dominant pathway regulating osteoclast recruitment is Although macroscopically the skeleton seems to be a static the RANKL/OPG system, while many different factors organ, it is an extremely dynamic tissue at the microscopic level. The ability of bone to sustain the tremendous loads placed on it in everyday life depends on, constant repair of E. F. Eriksen (*) mechanical microdamage that develops both in cancellous Department Of Clinical Endocrinology, Oslo University Hospital, bone—the “spongy” bone present in the vertebrae, pelvis, and Aker, ends (metaphyses) of long bones—and in cortical bone—the Trondheimsveien 235, 0514 Oslo, Norway compact bone present in the shafts (diaphyses) of the long e-mail: [email protected] bones and surrounding cancellous bone as a thin layer in the 220 Rev Endocr Metab Disord (2010) 11:219–227 vertebrae and pelvis. Bone remodeling is based on the [5]. In cortical bone remodeling proceeds in tunnels with concerted action of resorptive and formative cell populations osteoclasts forming “cutting cones” removing damaged in order to replace old bone with new bone and thus secure the bone followed by refilling by osteoblasts in the “closing integrity of the skeleton. This sequence has to be tightly cone” occurring behind the osteoclasts [6]. In normal bone regulated by both local and systemic factors, because the duration of the remodeling cycle in cortical is shorter significant deviations from a neutral balance between resorp- than in cancellous bone with a median of 120 days [6]. The tion and formation would mean severe accelerated bone loss total surface of cancellous bone is completely remodeled or bone gains with possible disastrous consequences in terms over a period of 2 years. of increased fracture risk or compression syndromes. Contrary to remodeling sites in cancellous bone, Bone remodeling takes place in what Frost termed the whichareclosetoredmarrow,knowntocontain Basic Multicellular Unit (BMU), which comprises the osteoprogenitor cells [7], remodeling sites in cortical bone osteoclasts, osteoblasts, and osteocytes within the bone- are distant from red marrow. Therefore, it was assumed remodeling cavity (Fig. 1). In cancellous bone remodeling that the mechanisms of bone remodeling were different in occurs on the surface of trabeculae and lasts about 200 days cancellous versus cortical bone, i.e. that the cells needed in normal bone. The remodeling cycle can be as short as for bone remodeling in cancellous bone traveled directly 100 days in thyrotoxicosis and primary hyperparathyroid- from the red marrow to bone surfaces in cancellous bone, ism and exceed 1,000 days in low turnover states like while cells reached cortical remodeling sites bone via the Myxedema and after bisphosphonate treatment [2]. Remod- vasculature [8]. eling is initiated by osteoclastic resorption, which erodes a resorption lacuna, the depth of which varies between 60 in 2.1 Osteoblast differentiation young individuals and 40 μm in older individuals. The resorption period has a median duration of 30–40 days and Osteoblasts are mesenchymal cells derived from mesoder- is followed by bone formation over a period of 150 days mal and neural crest progenitor cells and their formation (Fig. 1)[3, 4]. In normal bone the result of the remodeling entails differentiation from progenitors into proliferating cycle is complete refilling of the resorption lacuna with new preosteoblasts, bone matrix-producing osteoblasts, and bone. In disease states like osteoporosis, the main defect is eventually into osteocytes or a bone-lining cells. The that the osteoblast is unable to refill the resorption lacuna earliest osteoblastic marker, Runt-related transcription leading to a net loss of bone with each remodeling event factor 2 (Runx2) is necessary for progenitor cell differen- tiation along the osteoblast lineage [9]. During this sequence of cellular proliferation Runx2 regulates expres- Resorption Formation sion of genes encoding osteocalcin, VEGF, RANKL, Marrow capillary sclerostin, and dentin matrix protein 1 [DMP1] [10]. Cancellous bone Osterix is another transcription factor essential for osteo- 50 µm blast differentiation [11]. A large number of paracrine, autocrine, and endocrine factors affect osteoblast develop- ment and maturation like: bone morphogenetic proteins 80 µm (BMPs), growth factors like FGF and IGF, angiogenic factors like endothelin-1, hormones like PTH and prosta- Cortical bone Central vessel glandin agonists, all modulate osteoblast differentiation Haversian system [12]. The action of PTH and BMPs is closely associated with activation of Wnt signalling pathways [13]. 0 100 200 The fully differentiated osteoblast is characterized by Time -days coexpression of alkaline phosphatase and type I collagen, Fig. 1 Schematic representation of Bone Multicellular Units (BMUs) both important for synthesis of bone matrix and in cancellous and cortical bone. Broken lines denote the outer limit of subsequent mineralization thereof [14]. Mature osteo- Bone Remodeling Compartment associated with the resorptive and blasts also produce regulators of matrix mineralization formative sites of the BMU. The mean thickness of the structure in like osteocalcin, osteopontin and ostenectin, RANKL cancellous bone is 50 μm and 80 μm in cortical bone equivalent to a mean Haversian system diameter of 160 μm. The Blood supply for the which is necessary for osteoclast differentiation as well BRCs is provided by capillaries either coming from the marrow space as the receptor for PTH (PTHR1). At the end of their as is the case for cancellous BMUs or from the central vessel of lifespan osteoblasts transform into either osteocytes which Haversian systems in cortical bone. The duration of the remodeling become embedded in the mineralized matrix or lining sequence is somewhat longer in cancellous than in cortical bone. The position of marrow cappillaries is hypothetical, as the exact cells, which cover all surfaces of bone. Specific molecules distribution is poorly elucidated expressed by osteocytes include DMP1, FGF 23 and Rev Endocr Metab Disord (2010) 11:219–227 221 sclerostin, which control bone formation and phosphate reduced estrogen levels favoring increased resorption. In metabolism [15]. many instances [26]. PTH given as daily injections favors bone anabolism, reduces RANKL and increases OPG 2.1.1 Wnts and osteoblast differentiation levels. In cases with chronic elevation of circulating PTH levels as seen in primary hyperparathyroidism the opposite Wnts are secreted glycoproteins crucial for the development pattern is seen with elevated RANKL and reduced OPG and renewal of many tissues, including bone. Wnt levels. A humanized monoclonal antibody against RANKL signalling dominate osteoblast differentiation pathways has been shown to elicit even more pronounced reduction and act via binding to a receptor complex consisting of in osteoclast numbers [27] than bisphosphonates and has LDL receptor-related protein 5 (LRP5) orLRP6 and one of demonstrated excellent reduction of fracture risk in post- ten Frizzled molecules [13]. The so called canonical Wnt menopausal osteoporosis [28]. signaling pathway is active in all cells of the osteoblastic lineage, and involves the stabilization of β-catenin and 2.3 Coupling between resorption and formation regulation of multiple transcription factors [16]. Wnt/β- catenin signaling is also important for mechanotransduc- During normal bone remodeling, the amount of resorbed tion, fracture healing and osteoclast maturation [17–19]. bone is completely replaced in location and amount by new The activation of canonical Wnt-signaling promotes bone. This is secured through tight coupling of bone osteoblast differentiation from mesenchymal progenitors at resorption to bone formation. The mechanisms underlying the expense of adipogenesis, which leads to improved bone the coupling process still remains largely elusive, although strength, while suppression causes bone loss [20]. Canon- the last 15 years has increased our knowledge significantly. ical Wnt signaling in osteoblast differentiation is modulated The dominating hypothesis years ago was that liberation by Runx2 and osterix [21]. of growth factors like IGF 1 and 2 and cytokines embedded Wnt signaling is a prime target for bone active drugs and in bone matrix during bone resorption secured the balance the approaches include inhibition of Wnt antagonist like between resorption and formation during bone remodeling Dkk1, sclerostin, and Sfrp1 with neutralizing antibodies [29]. Later work showing that osteoblastic bone formation and inhibition of glycogen synthase kinase 3 β (GSK3 β), proceeds unperturbed despite lack of bone resorption in the which promotes phosphorylation and degradation of β- presence of defective osteoclasts lacking for example catenin. One of the most promising approaches so far has chloride channels or important factors for ruffled border been inhibition of the osteocyte protein sclerostin, which formation in osteoclasts like c-Src [30] has supplemented exerts tonic inhibition of osteoblast activity [22]. Sclerostin this hypothesis. The important role that osteoclasts play in is the product of the SOST gene, which is mutated and the regulation of bone formation is also corroborated by downregulated in patients with sclerosteosis and van studies on mice lacking c-fos or M-CSF, which display Buchem disease and sclerosteosis [23], which are diseases absence of osteoclasts and defective bone formation [31]. characterized by high bone density. Expression levels of Other system involved in coupling of bone resorption to sclerostin are repressed in response to mechanical loading and bone formation are the transmembrane proteins, ephrinB2, intermittent PTH treatment [24]. Preliminary studies with a which are expressed on osteoblasts and EPH receptor B4 humanized monoclonal antibody against sclerostin have (EphB4), which are expressed on osteoclasts [32]. Also the shown bone anabolism in animals as well as humans [25]. osteoclastic factor sphingosine 1-phosphate (S1P) [33] seems to play a significant role. The interaction of Ephrin 2.2 Osteoclast differentiation and EPH by cell to cell contact promotes osteoblast differentiation and represses osteoclast differentiation. The dominating pathway regulating osteoclast differentia- Secretion of S1P by osteoclast seems to recruit osteoblast tion is the RANKL/RANK/OPG pathway. This pathway is progenitor cells to sites of bone resorption and stimulate based on osteoblasts promoting osteoclast differentiation differentiation of these progenitor cells by stimulating through membrane presentation of RANKL and binding of EphB4 signaling, This causes a shut down of bone this factor to the membrane receptor RANK on mononu- resorption and initiate the formative phase of bone clear osteoclast precursors. Osteoclast differentiation is also remodeling in the so called transition phase. modulated by M-CSF [26]. The promotion of osteoclast differentiation by RANKL is inhibited by the decoy receptor osteoprotegerin (OPG), which is also produced 3 Targeted and non-targeted remodeling by osteoblasts [26] (74). Estrogens increase OPG and decrease RANKL expression in osteblasts, thus favoring Through its constant removal and renewal of damaged bone formation. Postmenopausal bone loss is linked to bone, bone remodeling secures skeletal integrity throughout 222 Rev Endocr Metab Disord (2010) 11:219–227 life. It has become customary to distinguish between Table 1 Osteoblastic and endothelial markers detected on cells lining targeted and non-targeted (stochastic) remodeling. Non- the Bone remodeling Compartment (BRC) vs. vascular endothelial cells as assessed by immuno- and enzyme histochemical staining targeted remodeling denotes regulation remodeling by hormones like PTH, thyroxine, growth hormone and Antigen BRC Vascular endothelium estrogen, but also antiresorptive drugs like bisphospho- – nates may affect non-targeted remodeling. It seems that VEGF + – the main pathway is via modulation of osteoclasts, which Von Willebrand Factor + – then via the coupling between resorption and formation CD 34 + – subsequently affects osteoblast activity. Targeted remod- Alkaline Phosphatase* + – eling secures removal of damaged bone through targeted Osteocalcin + – resorption. Osteocytes are the most abundant cells in Osteonectin + bone, and their death by microdamage has been suggested to IGF 1,2 + – be the major event leading in the initiation of osteoclastic bone TGF β 1,2,3 + – resorption. In normal bone [34]. Resorption lacunae are 3 bFGF + – times more frequent in association with microcracks, indicating OPG + – that remodeling is associated with repair of such microdamage RANKL + – [35]. Damaged osteocytes promote differentiation of osteo- clast precursors driven by secretion of M-CSF and RANKL [36]. In cortical bone there is evidence to suggest, that microdamage not only activates new BMUs, but may also EG may direct the movement of existing BMUs as they tunnel through the cortex. It also seems that the degree of damage to the osteocyte network determines osteocyte metabolic responses to loading and influences targeted remodeling [37]. Analysis of the relationship of between mean microcrack length and BMU resorption space density in cortical bone EG Ob indicates that BMUs have an effective area about 40 times CV greater than their actual cross-section, which suggests that osteoclasts in the cutting cone of cortical BMUs are able to OC sense and steer toward microdamage [38]. The relation OC between microdamage and initiation of bone remodeling is further corroborated by the fact that osteoclastic resorption is augmented in old bone [39].

4 The bone remodeling compartment

The work by Hauge et al. [40] demonstrated that the cells in the BMU, even in cancellous bone, were not directly contiguous to the bone marrow, but rather they were covered by a “canopy” of cells forming the outer lining of a specialized vascular structure with the denuded bone surface as the other delineation. The cells of this canopy display all classicial markers of the osteoblastic phenotype (Table 1), and are therefore most probably bone-lining cells, which seem to be connected to bone-lining cells on the upper quiescent bone surface. The structure has been demonstrat- Fig. 2 Different representations of BRC structures in cortical ( panel) and trabecular bone (lower panel). In cortical bone the BRC ed in cortical as well as cortical bone (Fig. 2). In turn, these (outer demarcation by the broken line) is filled with erythrocyte ghosts bone-lining cells on the quiescent bone surface are in (EG) and is located at the closing cone of the Haversian system communication with osteocytes embedded within the bone situated over osteoblasts (OB). A few osteoclasts (OC) are also seen. matrix. Penetrating the canopy of bone-lining cells, and CV denotes the central vessel of the Haversian system. In trabecular bone (lower panel) the outer lining of the BRC is clearly discernible, presumably serving as a conduit for the cells needed in the demarcating a vascular structure on top of osteoblasts (OB). Picture in BMU, are capillaries. upper panel courtesy of Pierre Delmas, Lyon, France Rev Endocr Metab Disord (2010) 11:219–227 223

Angiogenesis is closely associated with bone resorption high concentrations inhibiting bone resorption [51]. Simi- and bone and angiogenic factors like VEGF and endothelin larly, osteoblastic growth and differentiation are inhibited regulate osteoclast and osteoblast activity [41]. In addition by high concentrations of NO, while lower concentrations blood vessels serve as a way of transporting circulating may play a role in regulating normal osteoblast growth and osteoblast [42] and osteoclast precursors [43]tosites in mediating the effects of estrogens on bone formation, undergoing active remodeling. The involvement of vascular mechanotransduction and bone anabolic responses [51]. cells during the initiation of bone resorption is still The dominating isoform of nitrogen oxide synthase (eNOS) unresolved. Is the very first step adhesion of a blood vessel is expressed in osteocytes and lining cells, but not in to bone lining cells at a site where targeted repair is cuboidal osteoblasts [52]. Acidosis and hypoxia generally needed? Conceivably, osteocyte apoptosis and possible increase bone resorption [53–56] and inhibit bone forma- release of osteotropic growth factors and cytokines could tion [57]. As hypoxia may cause acidosis through increased be attractants for blood vessels, which would then subse- anaerobic metabolism, the two factors may act synergisti- quently initiate the formation of a resorptive BRC. But, as cally at the tissue level [56]. Hypoxia and acidosis also outlined above, the framework for signaling within the affect secretion of pro-angiogenic factors like VEGF as osteocyte-lining cell-BRC network could also be a way by outlined below. which remodeling events on bony surfaces are triggered from damage accumulation or changes in mechanical strain within bone. 5 Key functions of the bone remodeling compartment There is increasing evidence for a common lineage and close interaction between vascular endothelial cells and 1. The BRC provides a closed microenvironment permit- bone cells. Endothelial cells drive differentiation of marrow ting tight regulation of bone remodeling. Current stromal cell towards the osteoblastic phenotype [44] concepts regarding local regulation of bone remodeling Endothelin and VEGF are also involved in signaling generally assumes that the local growth factors, between vasculature and bone [45], and VEGF as well as cytokines and even NO come either from cells in the other angiogenic factors are expressed during intramem- marrow space or vascular cells having free access to the branous osteogenesis. Osteoblastic cells, as well as osteo- remodeling site without barriers, or are produced by clasts, possess receptors for VEGF and also produce VEGF osteoclasts and osteoblasts at the remodeling site. The [46]. Expression of VEGF is closely associated with the BRC concept implies that the all factors liberated from early phases of bone modeling and remodeling events [47] the cells or vessels in the marrow space exert their and it induces osteoblast chemotaxis and differentiation regulatory role either through diffusion through the [48] and differentiation. outer layer of the BRC, transport via the bloodstream to Cells may enter the remodeling space either via the interior of the BRC or indirectly via modulation of diapedesis through the lining cell dome covering the BRC cell activity in the outer wall of the BRC. The presence or via the circulation. It is still debatable whether all cells of a specific compartment in which remodeling can involved in remodeling arrive via the circulation, but while proceed without interference from local factors liberat- circulating osteoclast precursors were demonstrated more ed in the marrow space seems to be logical. If the than a decade ago, there is now increasing evidence that access to the marrow space was open, the very high osteoblast lineage cells are also present in the circulation levels of growth factors in the marrow microenviron- strengthening the involvement of circulating precursor cells ment might offset eventual localized regulatory effects in the process [42, 49, 50]. by local growth factors, crucial to osteoclast and While the systemic hormonal regulation of the remodel- osteoblast differentiation and the remodeling process. ing process has to occur via factors arriving at individual 2. The BRC is the structure translating microdamage into remodeling sites via the bloodstream, the way by which targeted remodeling by which mechanosensory signals local regulatory factors exert their action on individual cell from the osteocyte network are translated into changes populations involved is still obscure. Over the last decades, in osteoclast and osteoblast activity on trabecular however, we have increased our knowledge about the surfaces. Lining cells are connected to the osteocyte different growth factors and cytokines involved in local network via gap junctions between lining cells on regulation of bone remodeling tremendously (Fig. 3). Apart quiescent surfaces and osteocyte cannaliculi [58] from growth factors and cytokines, simple molecules like (Fig. 4). Signals from lining cells indicating damage nitrogen oxide (NO), as well as hypoxia and acidosis have or stress could be transmitted to the outer lining cell been shown to exert pronounced effects on bone remodel- layer of the BRC and trigger osteoclast recruitment. By ing balance and activity. NO exerts biphasic effects on analogy with remodeling in cortical bone, which is osteoclast activity with low concentrations potentiating and clearly associated with growth of a blood vessel into 224 Rev Endocr Metab Disord (2010) 11:219–227

MONOCYTES VEGF, FGF, ENDOTHELIN,N Marrow capillary/sinusoids IL1,6,7,11, TNFα O, H+, HYPOXIA E2, PTH, 1,25D,T3

IGF-2, IGF-1, TGFβ,

-- OC OB

IL,M-CSF. C-fos TNF, Osteoblastic Vascular endothelium Lining cells EphrinB2 RANKL EphB4 EphrinB2

Osteoclast precursor EphB4 RANK reseptor

IGFs, S1P Osteoclast Osteoblast Monocyte Osteoclast

Fig. 3 Depiction of some of the main local regulatory factors endothelial cells (vascular endothelial growth factor (VEGF), endo- operating at remodeling sites with osteoclasts (OC) and osteoblasts thelin, nitrogen oxide (NO)) may diffuse to receptors on osteoclasts or (OB). Interleukins (IL), tumor necrosis factors (TNF), transforming osteoblasts. The cellular responses in the BMU are then further growth factors (TGF), colony stimulating factors (CSF), Insulin like modulated by systemic hormones in the circulation (estrogen (E2), growth factors (IGF), fibroblast growth factors (FGF), platelet derived parathyroid hormone (PTH), active vitamin D (1,25D), thyroid growth factors (PDGF), bone morphogenetic proteins (BMP)) are hormone (T3)). Left lower insert depicts in detail osteoblast- formed by both monocytic cells in the marrow space or circulation, as osteoclast interactions inside the BRC and right lower insert depict well as bone cells in the BMU. NFκB- or RANK- ligand (RANKL) an alternativ, still hypohetical, version of that interaction based on and osteoprotegerin (OPG) are formed specifically by osteoblasts. lining cells acting as the osteoblastic component in thata interaction Factors from the marrow space as well as factors liberated by

the remodeling site (5), the presumed ingrowth of a MARROW CAPILLARY capillary into the BRC provides the vascular supply for the cells in the BMU of cancellous bone and might also provide the necessary osteoclasts and, subsequently, the OC osteoblasts that are needed for bone remodeling in both OB cancellous and cortical bone The BRC would also be a site where hormonal modulation (e.g. ERT) of the mechanosensory input could take place [59]. 3. The BRC is the most probable structure at which coupling between osteoclasts and osteoblasts occurs. OSTEOCYTES The RANKL/OPG pathway involves presentation of osteoblastic, membrane bound RANKL to the RANK receptor on osteoclast precursors by cell to cell contact. Fig. 4 Connections between the osteocyte network, lining cells and the Due to the timing and sequence of bone resorption and BRC. All cells in this network are connected with gap junctions, which bone formation, however, resorption and formation are mayprovideapathway(block arrows), by which signals generated deep generally separated in time and space, which makes the within bone may reach the surface and elicit remodeling events by needed cell to cell contact between osteoclast precur- osteoclasts (OC) and osteoblasts (OB) in response to mechanical stimuli. The response may be modulated by factors liberated from the sors and active osteoblasts highly unlikely on a broader vascular endothelium or marrow capillaries/sinusoids and paracrine basis, and even if soluble RANKL played a major role factors (broken arrow) liberated from lining cells may also play a role it had no RANKL on precursor cells within the BRC to Rev Endocr Metab Disord (2010) 11:219–227 225

bind to. A more likely cell, which could present RANKL 6 Conclusion to RANK on osteoclast precursors, would be the lining cell. As demonstrated in animals [60] and in humans Bone remodeling involves tight coupling and regulation of [61], lining cells exhibit positive immunoreactivity for osteoclasts and osteoblasts and is modulated by a wide variety OPG and RANKL, and might therefore be responsible of hormones and osteocyte products secreted in response to for the cell to cell contact with osteoclast precursors. mechanical stimulation and microdamage. Bone remodeling 4. The BRC also obviates the need for a “postal code” proceeds in a specialized vascular entity the “Bone Remodel- system ensuring that resorptive and formative cells ing Compartment” (BRC), which provides the structural basis adhere to areas on the bone surface, where they are for coupling and regulation of cellular activity. Increased needed. Bone surfaces are generally covered by lining knowledge about the interplay between different factors and cells, which would prevent direct contact between bone cells surrounding the BRC will most likely result in even cells and integrins or other adhesion molecules known better treatment options for skeletal disease. to modulate cell activity. The BRC would be the only place where these cells (circulating osteoclasts as well as circulating osteoblast precursors) would be exposed Open Access This article is distributed under the terms of the to these matrix constituents, because the formation of Creative Commons Attribution Noncommercial License which per- the BRC involves detachment of lining cells from the mits any noncommercial use, distribution, and reproduction in any bone surface. medium, provided the original author(s) and source are credited. 5. The BRC may play a crucial role in the spread of bone metastases. It is well established that apart from References entering bone via local ingrowth, tumor cells reach bony surfaces via the circulation. The growth of 1. Frost HM. Tetracycline-based histological analysis of bone metastatic cells in bone is enhanced by the so called – “ ” remodeling. Calcif Tissue Res. 1969;3:211 37. vicious cycle , where PTHrp produced by tumor cells 2. Eriksen EF. Normal and pathological remodeling of human (e.g. breast cancer cells) induces increased local bone trabecular bone: three dimensional reconstruction of the remodel- resorption and subsequent liberation of TGFβ from the ing sequence in normals and in . Endocr – bone matrix [62]. The local effects of TGFβ in the Rev. 1986;7:379 408. 3. Eriksen EF, Gundersen HJ, Melsen F, Mosekilde L. Reconstruc- bone microenvironment are two-fold: 1) it enhances the tion of the formative site in iliac trabecular bone in 20 normal growth of bone metastases and 2) increases PTHrp individuals employing a kinetic model for matrix and mineral formation from tumor cells further [62], thus maintain- apposition. Metab Bone Dis Relat Res. 1984;5:243–52. ing the vicious cycle. As shown above, one of the key 4. Eriksen EF, Melsen F, Mosekilde L. Reconstruction of the β resorptive site in iliac trabecular bone: a kinetic model for bone components of the vicious cycle, TGF , is produced by resorption in 20 normal individuals. Metab Bone Dis Relat Res. the cells lining the BRC. Other key promoters of bone 1984;5:235–42. metastases like IL-1 and IL-6 are also produced by the 5. Eriksen EF, Hodgson SF, Eastell R, Cedel SL, O’Fallon WM, lining cell layer covering BRCs. It is therefore probable Riggs BL. Cancellous bone remodeling in type I (postmenopaus- al) osteoporosis: quantitative assessment of rates of formation, that the microenvironment in the BRC is highly resorption, and bone loss at tissue and cellular levels. J Bone conducive to metastatic seeding and the formation of Miner Res. 1990;5:311–9. the vicious cycle, further enabling growth of the bone 6. Agerbaek MO, Eriksen EF, Kragstrup J, Mosekilde L, Melsen F. metastasis. Moreover, the existence of a closed com- A reconstruction of the remodelling cycle in normal human cortical iliac bone. Bone Miner. 1991;12:101–12. partment would make vicious cycle formation easier 7. Parfitt AM. The mechanism of coupling: a role for the vasculature. due to absence of interference with cytokine and Bone. 2000;26:319–23. growth factors from the marrow space. Several large 8. Burkhardt R. In: Arlet J, Ficat RP, Hungerford DS, editors. The scale studies have established that bisphosphonates structural relationship of bone forming and endothelial cells of the bone marrow. Baltimore: Williams & Wilkins; 1984. p. 2–14. reduce the number of skeletal events in breast cancer, 9. Komori T, Yagi H, Nomura S, Yamaguchi A, Sasaki K, Deguchi K, prostate cancer and myelomatosis, and iv bisphospho- et al. Targeted disruption of Cbfa1 results in a complete lack of bone nates are now used routinely in advanced cancer [63]. formation owing to maturational arrest of osteoblasts. Cell. – There is still debate as to how much of the beneficial 1997;89:755 64. 10. Lian JB, Stein GS, Javed A, van Wijnen AJ, Stein JL, effects of bisphosphonates in advanced cancer are due Montecino M, et al. Networks and hubs for the transcriptional to inhibition of angiogenesis or to other, direct anti- control of osteoblastogenesis. Rev Endocr Metab Disord. tumor effects. Bisphosphonate, however, could exert 2006;7:1–16. their inhibitory effects on bone metastases simply by 11. Nakashima K, Zhou X, Kunkel G, Zhang Z, Deng JM, Behringer RR, et al. The novel zinc finger-containing transcription factor reducing the number of BRCs and thereby the surface osterix is required for osteoblast differentiation and bone formation. of denuded bone available for metastatic seeding. Cell. 2002;108:17–29. 226 Rev Endocr Metab Disord (2010) 11:219–227

12. Qin L, Qiu P, Wang L, Li X, Swarthout JT, Soteropoulos P, et abnormal osteoblast behavior in developing bone. J Bone Miner al. Gene expression profiles and transcription factors involved Res. 2004;19:1441–51. in parathyroid hormone signaling in osteoblasts revealed by 32. Zhao C, Irie N, Takada Y, Shimoda K, Miyamoto T, Nishiwaki T, microarray and bioinformatics. J Biol Chem. 2003;278:19723– et al. Bidirectional ephrinB2-EphB4 signaling controls bone 31. homeostasis. Cell Metab. 2006;4:111–21. 13. Westendorf JJ, Kahler RA, Schroeder TM. Wnt signaling in 33. Ryu J, Kim HJ, Chang EJ, Huang H, Banno Y, Kim HH. osteoblasts and bone diseases. Gene. 2004;341:19–39. Sphingosine 1-phosphate as a regulator of osteoclast differentia- 14. Murshed M, Harmey D, Millan JL, McKee MD, Karsenty G. tion and osteoclast-osteoblast coupling. EMBO J. 2006;25:5840– Unique coexpression in osteoblasts of broadly expressed genes 51. accounts for the spatial restriction of ECM mineralization to bone. 34. Cardoso L, Herman BC, Verborgt O, Laudier D, Majeska RJ, Genes Dev. 2005;19:1093–104. Schaffler MB. Osteocyte apoptosis controls activation of intra- 15. Bonewald L. Osteocytes as multifunctional cells. J Musculoskelet cortical resorption in response to bone fatigue. J Bone Miner Res. Neuronal Interact. 2006;6:331–3. 2009;24:597–605. 16. Hens JR, Wilson KM, Dann P, Chen X, Horowitz MC, 35. Li J, Mashiba T, Burr DB. Bisphosphonate treatment suppresses Wysolmerski JJ. TOPGAL mice show that the canonical Wnt not only stochastic remodeling but also the targeted repair of signaling pathway is active during bone development and growth microdamage. Calcif Tissue Int. 2001;69:281–6. and is activated by mechanical loading in vitro. J Bone Miner Res. 36. Kurata K, Heino TJ, Higaki H, Vaananen HK. Bone marrow 2005;20:1103–13. cell differentiation induced by mechanically damaged osteo- 17. Robinson JA, Chatterjee-Kishore M, Yaworsky PJ, Cullen DM, cytes in 3D gel-embedded culture. J Bone Miner Res. Zhao W, Li C, et al. Wnt/beta-catenin signaling is a normal 2006;21:616–25. physiological response to mechanical loading in bone. J Biol 37. Colopy SA, Benz-Dean J, Barrett JG, Sample SJ, Lu Y, Danova Chem. 2006;281:31720–8. NA, et al. Response of the osteocyte syncytium adjacent to and 18. Chen Y, Whetstone HC, Lin AC, Nadesan P, Wei Q, Poon R, et al. distant from linear microcracks during adaptation to cyclic fatigue Beta-catenin signaling plays a disparate role in different phases of loading. Bone. 2004;35:881–91. fracture repair: implications for therapy to improve bone healing. 38. Burger EH, Klein-Nulend J, Smit TH. Strain-derived canalicular PLoS Med. 2007;4:e249. fluid flow regulates osteoclast activity in a remodelling osteon—a 19. Spencer GJ, Utting JC, Etheridge SL, Arnett TR, Genever PG. proposal. J Biomech. 2003;36:1453–9. Wnt signalling in osteoblasts regulates expression of the receptor 39. Henriksen K, Leeming DJ, Byrjalsen I, Nielsen RH, Sorensen activator of NFkappaB ligand and inhibits osteoclastogenesis in MG, Dziegiel MH, et al. Osteoclasts prefer aged bone. Osteoporos vitro. J Cell Sci. 2006;119:1283–96. Int. 2007;18:751–9. 20. Bodine PV, Komm BS. Wnt signaling and osteoblastogenesis. 40. Hauge EM, Qvesel D, Eriksen EF, Mosekilde L, Melsen F. Rev Endocr Metab Disord. 2006;7:33–9. Cancellous bone remodeling occurs in specialized compartments 21. Hill TP, Spater D, Taketo MM, Birchmeier W, Hartmann C. lined by cells expressing osteoblastic markers. J Bone Miner Res. Canonical Wnt/beta-catenin signaling prevents osteoblasts from 2001;16:1575–82. differentiating into chondrocytes. Dev Cell. 2005;8:727–38. 41. Brandi ML, Collin-Osdoby P. Vascular biology and the skeleton. J 22. van Bezooijen RL, Svensson JP, Eefting D, Visser A, van der Bone Miner Res. 2006;21:183–92. Horst G, Karperien M, et al. Wnt but not BMP signaling is 42. Eghbali-Fatourechi GZ, Lamsam J, Fraser D, Nagel D, Riggs BL, involved in the inhibitory action of sclerostin on BMP-stimulated Khosla S. Circulating osteoblast-lineage cells in humans.[see bone formation. J Bone Miner Res. 2007;22:19–28. comment]. N Engl J Med. 2005;352:1959–66. 23. Balemans W, Ebeling M, Patel N, Van HE, Olson P, Dioszegi M, 43. Kassem M, Risteli L, Mosekilde L, Melsen F, Eriksen EF. et al. Increased bone density in sclerosteosis is due to the Formation of osteoblast-like cells from human mononuclear bone deficiency of a novel secreted protein (SOST). Hum Mol Genet. marrow cultures. APMIS. 1991;99:269–74. 2001;10:537–43. 44. Kaigler D, Krebsbach PH, Polverini PJ, Mooney DJ. Role of 24. Keller H, Kneissel M. SOST is a target gene for PTH in bone. vascular endothelial growth factor in bone marrow stromal cell Bone. 2005;37:148–58. modulation of endothelial cells. Tissue Eng. 2003;9:95–103. 25. Padhi D, Jang G, Stouch B, Fang L, Posvar E. Single-dose, 45. Veillette CJ, von Schroeder HP. Endothelin-1 down-regulates the placebo-controlled, randomized study of AMG 785, a sclerostin expression of vascular endothelial growth factor-A associated with monoclonal antibody. J Bone Miner Res. 2010. osteoprogenitor proliferation and differentiation. Bone. 2004;34:288– 26. Khosla S. Minireview: the OPG/RANKL/RANK system. Endo- 96. crinology. 2001;142:5050–5. 46. Tombran-Tink J, Barnstable CJ. Osteoblasts and osteoclasts 27. Reid IR, Miller PD, Brown JP, Kendler DL, Fahrleitner-Pammer express PEDF, VEGF-A isoforms, and VEGF receptors: possible A, Valter I, et al. Effects of denosumab on bone histomorphom- mediators of angiogenesis and matrix remodeling in the bone. etry: the FREEDOM and STAND studies. J Bone Miner Res. Biochem Biophys Res Commun. 2004;316:573–9. 2010;25:2256–65. 47. Xiong H, Rabie AB. Neovascularization and mandibular condylar 28. Cummings SR, San MJ, McClung MR, Siris ES, Eastell R, Reid bone remodeling in adult rats under mechanical strain. Front IR, et al. Denosumab for prevention of fractures in postmeno- Biosci. 2005;10:74–82. pausal women with osteoporosis. N Engl J Med. 2009;361:756– 48. Li G, Cui Y, McIlmurray L, Allen WE, Wang H. rhBMP-2, 65. rhVEGF(165), rhPTN and thrombin-related peptide, TP508 29. Mohan S, Baylink DJ. Insulin-like growth factor system compo- induce chemotaxis of human osteoblasts and microvascular nents and the coupling of bone formation to resorption. Horm Res. endothelial cells. J Orthop Res. 2005;23:680–5. 1996;45 Suppl 1:59–62. 49. Zvaifler NJ, Marinova-Mutafchieva L, Adams G, Edwards CJ, 30. Karsdal MA, Martin TJ, Bollerslev J, Christiansen C, Henriksen Moss J, Burger JA, et al. Mesenchymal precursor cells in the K. Are nonresorbing osteoclasts sources of bone anabolic activity? blood of normal individuals. Arthritis Res. 2000;2:477–88. J Bone Miner Res. 2007;22:487–94. 50. Kuznetsov SA, Mankani MH, Gronthos S, Satomura K, Bianco P, 31. Dai XM, Zong XH, Akhter MP, Stanley ER. Osteoclast deficiency Robey PG. Circulating skeletal stem cells. J Cell Biol. results in disorganized matrix, reduced mineralization, and 2001;153:1133–40. Rev Endocr Metab Disord (2010) 11:219–227 227

51. Ralston SH, Ho LP, Helfrich MH, Grabowski PS, Johnston PW, 58. Marotti G, Ferretti M, Muglia MA, Palumbo C, Palazzini S. A Benjamin N. Nitric oxide: a cytokine-induced regulator of bone quantitative evaluation of osteoblast-osteocyte relationships on resorption. J Bone Miner Res. 1995;10:1040–9. growing endosteal surface of rabbit tibiae. Bone. 1992;13:363–8. 52. Fox SW, Chow JW. Nitric oxide synthase expression in bone 59. Bonewald LF. Osteocyte biology: its implications for osteoporo- cells. Bone. 1998;23:1–6. sis. J Musculoskelet Neuronal Interact. 2004;4:101–4. 53. Bushinsky DA. Acid-base imbalance and the skeleton [Review] 60. Silvestrini G, Ballanti P, Patacchioli F, Leopizzi M, Gualteri N, [67 refs]. Eur J Nutr. 2001;40:238–44. Monnazzi P, et al. Detection of osteoprotegerin (OPG) and its 54. Goldhaber P, Rabadjija L. H+ stimulation of cell-mediated bone ligand (RANKL) mRNA and protein in femur and tibia of the rat. resorption in tissue culture. Am J Physiol. 1987;253:E90–8. J Mol Histol. 2005;36:59–67. 55. Arnett TR, Dempster DW. Effect of pH on bone resorption by rat 61. Eriksen EF, Qvesel D, Hauge EM, Melsen F. Further evidence that osteoclasts in vitro. Endocrinology. 1986;119:119–24. vascular remodeling spaces are lined by cells of osteogenic origin: 56. Arnett TR, Gibbons DC, Utting JC, Orriss IR, Hoebertz A, characterization of a possible coupling structure. J Bone Miner Rosendaal M, et al. Hypoxia is a major stimulator of osteoclast Res. 2005;15:S371. formation and bone resorption. J Cell Physiol. 2003;196:2–8. 62. Mundy GR. Mechanisms of bone metastasis [Review] [99 refs]. 57. Utting JC, Robins SP, Brandao-Burch A, Oriss IR, Behar J, Cancer. 1997;80:1546–56. Arnett TR. Hypoxia inhibits the growth, differentiation and 63. Polascik TJ. Bisphosphonates in oncology: evidence for the bone forming capacity of rat osteoblasts. Exp Cell Res. prevention of skeletal events in patients with bone metastases. 2006;312:1694–702. Drug Des Devel Ther. 2009;3:27–40.