Sunteți pe pagina 1din 8

Archives of Biochemistry and Biophysics 473 (2008) 139–146

Contents lists available at ScienceDirect

Archives of Biochemistry and Biophysics


journal homepage: www.elsevier.com/locate/yabbi

Review

Functions of RANKL/RANK/OPG in bone modeling and remodeling


Brendan F. Boyce *, Lianping Xing
Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, 601 Elmwood Avenue, Box 626, Rochester, NY 14642, USA

a r t i c l e i n f o a b s t r a c t

Article history: The discovery of the RANKL/RANK/OPG system in the mid 1990s for the regulation of bone resorption has
Received 18 January 2008 led to major advances in our understanding of how bone modeling and remodeling are regulated. It had
and in revised form 18 March 2008 been known for many years before this discovery that osteoblastic stromal cells regulated osteoclast for-
Available online 25 March 2008
mation, but it had not been anticipated that they would do this through expression of members of the
TNF superfamily: receptor activator of NF-jB ligand (RANKL) and osteoprotegerin (OPG), or that these
Keywords: cytokines and signaling through receptor activator of NF-jB (RANK) would have extensive functions
Bone resorption
beyond regulation of bone remodeling. RANKL/RANK signaling regulates osteoclast formation, activation
Osteoclasts
RANKL
and survival in normal bone modeling and remodeling and in a variety of pathologic conditions charac-
RANK terized by increased bone turnover. OPG protects bone from excessive resorption by binding to RANKL
OPG and preventing it from binding to RANK. Thus, the relative concentration of RANKL and OPG in bone is
a major determinant of bone mass and strength. Here, we review our current understanding of the role
of the RANKL/RANK/OPG system in bone modeling and remodeling.
Ó 2008 Elsevier Inc. All rights reserved.

Normal bone modeling only a few trabeculae to comprise the spongy bone of the secondary
spongiosa. This secondary spongiosa comprises the metaphyseal
With the exception of the bones of the calvaria, all bones in the bone between the epiphysis and the diaphysis of the bones. Osteo-
mammalian skeleton are preformed in cartilage moulds from mes- blasts lay down new bone on parts of the surfaces of these surviving
enchymal progenitors, which under appropriate stimuli also have trabeculae where there has been osteoclastic resorption and much of
the potential to differentiate into a variety of tissue types, includ- this new bone is subsequently resorbed by osteoclasts in a remodel-
ing fibrous tissue, fat and muscle. Chondrocytes proliferate near ing process that ensures that the volume of bone in the medullary
the ends of the cartilage moulds to drive their longitudinal growth, cavity is limited and does not fill the space. This osteoclastic resorp-
while others in the centers of undergo hypertrophic differentiation. tion of new bone provides space for the proliferation of hematopoi-
The hypertrophic chondrocytes at the periphery of the centers of etic cells, which like OCPs come from circulating precursors in the
these moulds are invaded by blood vessels and undergo apoptosis. spleen and liver, and form the bone marrow. Osteoclastic resorption
Some of this hypertrophic cartilage survives as thin islands of car- thus prevents the development of osteopetrosis, a congenital defect
tilage in the centers of ossification in growing bones. Osteoblasts, of endochondral ossification, which occurs if osteoclasts fail to form
which differentiate from progenitors in a collar of connective tissue or have impaired activity [2,3].
around the middle of the bones where vascular invasion takes As the cartilaginous centers of the growing bones are removed
place, follow the endothelial cells and lay down bone matrix on and replaced by bone and marrow, condensations of proliferating
the surfaces of these islands of cartilage to form bone struts or tra- and prehypertrophic chondrocytes form close to the ends of long
beculae [1]. Osteoclast precursors (OCPs)1, derived from progeni- bones where along with a layer of hypertrophic chondrocytes they
tors in the spleen and liver are attracted from blood in the constitute the epiphyseal growth plates. This process, called endo-
invading blood vessels close to newly formed bone trabeculae. These chondral ossification, requires expression by mesenchymal osteo-
osteoclast precursors fuse with one another to form multinucleated blast precursor cells of Runx2, the master transcription factor
osteoclasts, which resorb most of the newly formed bone leaving that regulates bone formation [4,5] because in the absence of
Runx2 vascular invasion of the cartilage moulds does not take
place and no bone is formed [6,7]. Runx2 also mediates the osteo-
* Corresponding author. Fax: +1 585 273 3637. blast differentiating effects of BMP2 [8], which regulates chondro-
E-mail address: brendan_boyce@urmc.rochester.edu (B.F. Boyce). cyte differentiation as well as the formation of bone during
1
Abbreviations used: OCPs, Osteoclast precursors; OPG, osteoprotegerin; OPGL, OPG endochondral ossification [9] in an NF-jB-dependent manner [10].
ligand; ODF, osteoclast differentiation factor; HSCs, hematopoietic stem cells; TRAFs,
TNF receptor-associated factors; JNK, c-Jun N-terminal kinase; AP-1, activator
Hypertrophic and to a lesser extent prehypertrophic chondro-
protein-1; ITAM, immunoreceptor tyrosine-based activation motif; IKKs, inhibitory cytes express RANKL, OPG and RANK. Mice deficient in RANKL,
kappa kinases. RANK and NF-jB p50 and p52 develop osteopetrosis because they

0003-9861/$ - see front matter Ó 2008 Elsevier Inc. All rights reserved.
doi:10.1016/j.abb.2008.03.018
140 B.F. Boyce, L. Xing / Archives of Biochemistry and Biophysics 473 (2008) 139–146

do not form osteoclasts (see below) and have thickened hypertro- cavity in bones, bone formation matches resorption during normal
phic cartilage zones in their growth plates. This defect is rectified bone remodeling.
spontaneously between 2 and 3 weeks of age in the RANKL / The processes that drive bone remodeling are still not fully ex-
and RANK / mice and 2–3 weeks later in the NF-jB p50 and plained, but they include damage to parts of bones in response to
p52 double knockout mice (our unpublished observations). The normal wear and tear, changes in mechanical forces following
precise role of RANKL/RANK/NF-jB signaling in chondrocytes dur- alterations in body shape or weight or exercise, and local release
ing endochondral ossification remains poorly understood, but of cytokines or growth factors due to alterations in levels of sys-
these findings suggest that it may regulate the lifespan of hypertro- temic hormones. It is a tightly regulated process in that formation
phic chondrocytes through NF-jB p50 and p52-regulated genes at follows resorption in a site-specific manner and there are more
least temporarily, similar to MMP-9 [11], which is regulated by than 1 million of these microscopic foci of remodeling at any time
NF-jB [12]. All of these knockout mice are dwarfed, suggesting that in the adult skeleton.
RANKL/RANK/NF-jB signaling during the first 2–3 weeks of life is Recent studies suggest that at least in response to mechanical
essential for attainment of full skeletal growth, but it is not known forces, osteocytes regulate the recruitment of osteoclasts to sites
if this requirement is in cells of the osteoclast, osteoblast, chondro- of bone resorption by inducing the expression of RANKL by osteo-
cyte, or endothelium lineages. blastic cells in the local micro-environment [20]. Despite this ad-
Recent studies suggest that hypertrophic chondrocytes are vance, the precise molecular mechanisms that control the
involved in the differentiation of osteoclast precursors at growth initiation, progression and cessation of remodeling at any given
plates. Their expression of RANKL is regulated positively by Vita- site remain poorly understood. Osteocytes are terminally differen-
min D3 [13] and BMP2 [14] in a dose-dependent manner in vitro. tiated osteoblasts, which became embedded in the osteoid matrix
BMP2 is expressed by prehypertrophic chondrocytes in vivo and they were laying down. They communicate with one another and
it induces higher expression of RANKL from chick upper sternal with osteoblastic cells on the surface of overlying bone and
chondrocytes than from lower sternal chondrocytes [14], which through them with osteoblastic cells in the bone marrow cavity
consist predominantly of prehypertrophic cells. Upper sternal and in this way are thought to regulate RANKL expression by these
chondrocytes, in contrast are predominantly hypertrophic cells latter cells, although they do not appear to express RANKL
and these, like osteoblasts, also express OPG. However, these chon- themselves.
drocytes do not express OPG to the same degree as RANKL in The rate of bone remodeling and the number of remodeling
response to BMP2 and thus the RANKL/OPG ratio changes in favor sites are increased in a variety of pathologic conditions affecting
of osteoclast differentiation in response to BMP2. Chondrocytes the skeleton, including postmenopausal osteoporosis, hyperpara-
express RANKL constitutively in vitro and surprisingly can support thyroidism, and rheumatoid arthritis, in which local and/or sys-
osteoclast differentiation from precursors when both cell types are temic alterations in the levels of hormones or pro-inflammatory
cultured in a transwell assay system [14]. Bone growth during cytokines stimulate bone resorption [19]. Most of these factors in-
embryonic development and in the first 2 weeks after birth occurs duce bone resorption predominantly by an indirect mechanism
at a very high rate. Thus, it is essential that osteoclast formation is that involves upregulation of the expression of M-CSF and RANKL
induced at the growth plate to mediate the rapid removal of calci- by osteoblastic and other cells [21]. Our understanding of the
fied matrix and the formation of the marrow cavity. Hypertrophic molecular mechanisms that regulate the formation and activity
chondrocytes are ideally situated in growth plates close to the of osteoclasts has advanced rapidly following the identification in
blood vessels that bring osteoclast precursors from the circulation the mid-to-late 1990s of the RANKL/RANK/OPG signaling system,
into developing bones to mediate this process. They secrete VEGF coupled with study of bones from genetically altered mice and
[11] and other factors that regulate vascular in-growth and could from animal models of bone diseases. More recently, it has become
also promote egression of OCPs from these newly-formed blood increasingly clear that osteoclasts are not simply bone resorbing
vessels through RANKL expression. RANKL has a chemo-attractant cells, but that they also regulate osteoblast functions positively
effect on peripheral blood monocytes [15,16]. Therefore, local re- and negatively [22,23], mediate the egression of hematopoietic
lease of RANKL by chondrocytes and induction of its expression stems from the marrow into the blood [24], and function as immu-
by BMP2 could mediate this OCP egression. Interestingly, osteo- nomodulators in pathologic states [25].
clasts are not required for the removal of the cartilage that forms
during endochondral ossification since cartilage is resorbed and Regulation of osteoclast formation and activation by OPG,
bone forms in the absence of osteoclasts, such as in RANKL and RANKL and RANK
RANK knockout mice [17,18]. Presumably the cartilage is removed
by chondroclasts, but these remain poorly defined cells, which may Osteoclasts are derived from mononuclear precursors in the
be in the monocyte lineage [11]. myeloid lineage of hematopoietic cells that also give rise to macro-
phages. Understanding of the molecular mechanisms that regulate
Bone remodeling osteoclast formation and activation has advanced rapidly in the
last 12 years since the discovery of the RANKL/RANK signaling sys-
Bone has multiple functions in vertebrates, including protection tem. M-CSF expression by osteoblastic stromal cells is required for
of vital organs and hematopoietic marrow, structural support for progenitor cells to differentiate into osteoclasts, but M-CSF on its
muscles, and storage and release of vital ions, such as calcium, own is unable to complete this process. This requirement for
and of growth factors stored in the matrix. Bone in the adult skel- M-CSF was discovered by the observation that op/op mice, which
eton is renewed continuously in response to a variety of stimuli by do not express functional M-CSF, have osteopetrosis because they
the process of bone remodeling. This involves removal of trenches lack osteoclasts [26]. Completion of OCP differentiation requires
or tunnels of bone from the surfaces of trabecular and cortical expression of RANKL by osteoblastic stromal cells and of RANK
bone, respectively, by osteoclasts [19]. Osteoblasts subsequently by OCPs.
fill in these trenches by laying down new bone matrix in them. This The requirement of osteoblastic stromal cell expression of a fac-
sequence is similar to that which occurs in the trabeculae formed tor(s) that mediated OCP differentiation had been recognized for
during endochondral ossification, but unlike in the latter where many years before the discovery of RANKL, its decoy receptor,
resorption exceeds formation to allow formation of the medullary OPG and its receptor, RANK. Many groups had attempted unsuc-
B.F. Boyce, L. Xing / Archives of Biochemistry and Biophysics 473 (2008) 139–146 141

cessfully to purify what turned out to be RANKL from bone cells for and beta integrin-mediated signaling from bone matrix activate
many years before its discovery by four groups working indepen- osteoclasts [41]. Osteoclast precursors fuse with one another and
dently and using different approaches. Researchers at Amgen become multinucleated under the influence of RANKL. This fusion
unexpectedly discovered the naturally-occurring inhibitor of requires expression by OCPs of DC-STAMP [42] and of Atp6v0d2, a
RANKL, a molecule they named osteoprotegerin (OPG) because it subunit of v-ATPase, a component of the V-type H+ ATP6i proton
protected against bone loss. They were making transgenic mice pump complex that secretes H+ from osteoclasts [23]. RANKL also
over-expressing various TNF receptor-related cDNAs in attempts induces expression of tartrate-resistant acid phosphatase and
to find molecules that could interfere with TNF signaling. They cathepsin K through NFATc1 [35].
observed that mice over-expressing a particular cDNA developed
osteopetrosis due to a lack of osteoclasts [27]. Researchers at the
Snow Brand Milk Products Co. in Japan discovered an identical RANKL
molecule [28] by purifying a factor from ST-2 osteoblastic cells that
inhibited osteoclast formation. Both groups used expression clon- RANKL exists as a homotrimeric protein and is typically
ing and OPG as a probe and quickly identified its ligand, which they membrane-bound on osteoblastic and activated T cells or is
called OPG ligand (OPGL) and osteoclast differentiation factor secreted by some cells, such as activated T cells [43–45]. The
(ODF), respectively [29,30]. This protein turned out to be identical secreted protein is derived from the membrane form as a result
to a member of the TNF super-family, which had been identified of either proteolytic cleavage or alternative splicing [46]. The
the year before and called receptor activator of nuclear factor-jB proteolytic cleavage of RANKL is carried out by matrix metallo-
ligand (RANKL) [31] and TNF-related activation induced cytokine proteases (MMP3 or 7) [47] or ADAM (a disintegrin and metallo-
(TRANCE) [32]. Researchers at Immunex had already discovered protease domain) [48]. Most of the factors known to stimulate
RANK while they were sequencing cDNAs from a human bone- osteoclast formation and activity induce RANKL expression by
marrow-derived myeloid dendritic-cell cDNA library [18] and it osteoblastic stromal cells. However, RANKL is also highly ex-
was soon identified as the receptor for OPGL/ODF. They had found pressed in lymph nodes, thymus, mammary glands and lung
that RANK was involved in the survival of dendritic cells and iso- and at low levels in a variety of other tissues, including spleen
lated RANKL by direct expression screening. They found that and bone marrow [43]. It is expressed by synovial cells and acti-
RANKL was expressed by T cells and that it increased proliferation vated T cells in joints of patients with inflammatory arthritis to
and survival of dendritic cells. RANKL has come to be the generally contribute at least in part to the joint destruction seen in pa-
accepted acronym for this cytokine following the recommendation tients with rheumatoid arthritis.
of a nomenclature committee of the American Society for Bone and This joint destruction in RA is also mediated by TNF by a
Mineral Research [33]. RANKL/RANK/NF-jB signaling is also re- number of mechanisms: it increases the proliferation of OCPs
quired for lymph node development and B cell maturation in the bone marrow and the number of them circulating system-
[18,34]. These discoveries that RANKL/RANK/ NF-jB signaling is in- ically by promoting their egression from the bone marrow [49];
volved in osteoclast formation and immune responses and that T it also promotes the egression of OCPs from the blood to in-
and B cells express RANKL have spawned the growing field of flamed joints where along with RANKL and IL-1 it promotes fu-
osteoimmunology. sion of these cells into osteoclasts [49]. RANKL also stimulates
OCP differentiation is regulated by a number of transcription the release of OCPs into the circulation; and recent studies using
factors and signaling pathways that are activated by RANKL/RANK PTPe-knockout mice suggest that osteoclasts themselves regulate
interaction. The completion of OCP differentiation by RANKL the egression of hematopoietic stem cells (HSCs) from niches
requires the sequential expression of NF-jB, c-Fos and NFATc1 within the marrow under the control of RANKL. Osteoclasts from
[35–37]. Interestingly, TNF, which like RANKL can induce osteo- PTPe-knockout mice have defective adhesion to bone and im-
clast differentiation directly from wt OCPs, also induces the paired resorption and RANKL did not induce HSC mobilization
sequential expression of NF-jB, c-Fos and NFATc1 [37]. Impor- in these mice [24]. Thus, RANKL-induced osteoclast activation
tantly, TNF can also induce osteoclast formation from OCPs from appears to regulate HSC mobilization as part of homeostasis
RANKL / and RANK / mice in vitro [38] and thus could and host defense mechanisms, linking bone remodeling with
augment the osteoclast formation induced indirectly by itself and the regulation of hematopoiesis.
other factors through induction of osteoblastic cell expression of RANKL is also expressed in epithelial cells in mammary gland
RANKL. c-Fos or NFATc1 can substitute for NF-jB in OCPs in this lobules during pregnancy and is required for hyperplasia of these
sequential activation program by RANKL and TNF [37], and when cell during lactation and thus milk production in mice [50]. It is
either of these transcription factors is over-expressed in OCPs IL- expressed by some malignant tumor cells which also express
1 can induce osteoclast formation directly from them also [39]. RANK, and thus RANKL signaling may regulate tumor cell prolif-
RANKL and TNF induce c-Fos expression in OCPs [37,39]. This eration [51] by either an autocrine or a paracrine mechanism
may be an important role for these cytokines at sites of inflamma- when it is produced by accessory cells.
tion in bone where IL-1 concentrations are increased because this More recent studies have identified a role for RANKL signaling
could facilitate direct induction of osteoclast formation by IL-1 and in tumor cell migration and bone metastasis [52]. T cell production
thus augment the effects of these cytokines on osteoclastogenesis. of RANKL also induces expression of INFb by activated osteoclasts
To resorb bone effectively, osteoclasts must attach themselves through c-Fos signaling to negatively regulate their formation [53],
firmly to the bone surface using specialized actin-rich podosomes. a mechanism that can be enhanced by T cell produced INFc which
By means of these podosomes, they form tight seals with the degrades TRAF6, an essential adapter protein recruited to RANK to
underlying bone matrix in roughly circular extensions of their mediate RANK signaling (vide infra) [54]. IFN-c is a cytokine se-
cytoplasm and within these sealed zones they form ruffled border creted primarily by activated T cells and NK cells and was origi-
membranes. This ruffling of the cytoplasmic membrane increases nally characterized as a powerful macrophage activator that
the area of the cell surface for secretion of the proteolytic enzyme, upregulated nitric oxide production and MHC class II expression
cathepsin K, and hydrochloric acid onto the bone surface [40]. By in macrophages [55]. It has been reported to be a strong suppressor
this sealing and secretory mechanism, they simultaneously of osteoclastogenesis in vitro through inhibition of RANKL signal-
degrade the matrix and dissolve the mineral of bone, while pro- ing [54,56]. However, its effects on osteoclast formation are con-
tecting neighboring cells from the harmful effects of HCl. RANKL troversial. IFN-c also enhances osteoclast generation in cultures
142 B.F. Boyce, L. Xing / Archives of Biochemistry and Biophysics 473 (2008) 139–146

of peripheral blood from osteopetrotic patients, in part by normal- ity and osteolysis seen in some patients with familial Paget’s dis-
izing superoxide production [57] and has been shown to be effica- ease, confirming the importance of this system in humans [64]. A
cious in the treatment of osteoporosis in humans [58]. A recent potential role for RANK in tumor cell proliferation [51] is being
study analyzed the in vivo effects of IFN-c in 3 mouse models of investigated and if proven could be a future target for anti-tumor
bone loss, including ovariectomy, LPS injection, and inflammation therapy.
induced by silencing TGF-b signaling in T cells. It demonstrated
that the net effect of IFN-c in these conditions was bone loss due OPG
to increased osteoclast formation through the following mecha-
nism: IFN-c increased the antigen presenting function of bone mar- OPG is secreted by many cell types in addition to osteoblasts,
row cells by up-regulating their expression of MHC class II including those in the heart, kidney, liver, and spleen. A recent
molecules and thus stimulating T lymphocytes to produce RANKL, study reports that B cells may be responsible for 64% of total
TNF and IFN-c [59]. It is likely that the effects of IFN-c on osteoclast bone marrow OPG production and B cell-deficient mice are con-
formation are complex and will depend on specific conditions in sistently osteoporotic, consistent with B cells being a major
the bone microenvironment and relative concentrations of other source of OPG in the bone marrow of normal mice [65]. Most
cytokines that can affect the differentiation of OCPs. c-Fos/ of the factors that induce RANKL expression by osteoblasts also
NFATc1-induced OCP differentiation can also be inhibited by re- regulate OPG expression [66]. Although there some are contra-
verse signaling through the OCP-expressed ligand, ephrin B2 dictory data, in general when RANKL expression is up-regulated,
[22]. Interaction between ephrin B2 and its receptor, Eph 4 on OPG expression is down-regulated or not induced to the same
osteoblast precursors prevents c-Fos activation of NFATc1 to inhi- degree as RANKL, such that the RANKL/OPG ratio changes in fa-
bit OCP differentiation. Intriguingly, forward signaling through Eph vor of osteoclastogenesis [43,67]. Osteoclast numbers and activ-
4 in osteoblast precursors promotes their differentiation [22], pro- ity can increase if there is a change in the RANKL/OPG ratio due
viding further evidence that osteoclastic cells have functions in to either an increase in the former or a decrease in the latter or
bone other than bone resorption. It is likely that other mechanisms a change in both that leads to a change in the ratio in favor of
that limit osteoclast formation and activation through RANKL or RANKL.
TNF signaling will be identified. OPG’s osteo-protective role in humans has been supported by
Failure of osteoclast formation or activation leads to the devel- homozygous partial deletions of opg in patients with juvenile
opment of osteopetrosis in mice, humans and other mammals [2]. Paget’s disease, an autosomal recessive disorder in which affected
To date, most cases of osteopetrosis in humans have been attrib- individuals have increased bone remodeling, osteopenia, and frac-
uted to mutations in genes, such as ClCN7, which encodes the chlo- tures [68]; in addition, some patients with idiopathic hyperphos-
ride channel through which Cl flows from osteoclasts, OSTM1, phatasia, an autosomal recessive bone disease in which affected
which has a closely related function, TCIRG1, which encodes the children have increased bone turnover associated with deformities
a3 subunit of the H+ATPase of the proton pump, carbonic anhy- of long bones, kyphosis and acetabular protrusion have an inacti-
drase II, which catalyzes the hydration of CO2 to H2CO3 to provide vating deletion in exon 3 of OPG [69].
a source of H+, cathepsin K, which degrades the collagenous matrix, OPG expression is regulated in osteoblasts not only by a
and Plekhm1, which encodes for a vesicle-associated protein variety of cytokines, hormones and growth factors [67], but also
linked to small GTPase signaling [2,3,60]. Recently, the first report by Wnt/b-catenin [70,71]. The Wnt/b-catenin pathway also reg-
of a mutation in the RANKL gene was described in a kindred of First ulates osteoblastic bone formation and the commitment of mes-
Nations individuals in Canada [61]. The affected individuals had enchymal cells to the osteoblast lineage [72]. Jagged1/Notch1
osteopetrosis, but they did not have obvious defect in immunologic signaling negatively regulates osteoclast formation both directly
parameters such as the number of B and T lymphocytes and in osteoclast precursors and indirectly by affecting the OPG/
frequency of infection. It is also not known if they have some of RANKL expression ratio in stromal cells [73]. Thus, bone mass
the other phenotypic features of RANKL / mice, such as absence is determined by many influences on osteoblasts and osteo-
of lymph nodes or failure of lactational hyperplasia. Apparent clasts and is regulated by osteoblasts through three major sig-
absence of anticipated immunologic features of RANKL deficiency naling pathways: RANKL/RANK, Wnt/b-catenin and Jagged1/
in affected patients could reflect species-specific differences, or Notch1.
the possibility that these individuals make a mutant RANKL Based on the presence of renal and aortic calcification in
proteins that has some biological activity that mediates immune OPG / mice, OPG appears to protect large blood vessels from
function, or that these individuals have immune cell deficiencies medial calcification [74]. Calcification commonly complicates
that have not yet been detected. long-standing atherosclerosis, and the absence of OPG in OPG/
apoE double knockout mice accelerates calcific atherosclerosis
RANK that develops in apoE / mice [75]. Other findings suggest that
OPG could also limit calcification of atherosclerotic plaques [76].
RANK is a homotrimeric transmembrane protein member of the However, whether OPG or RANKL play important roles in cardio-
TNF receptor superfamily. It appears to be expressed in fewer vascular disease remains to be determined and is controversial
tissues than RANKL at the protein level, but in addition to OCPs, [77]. For example, OPG serum levels are high in patients with
mature osteoclasts and dendritic cells, it is expressed in mammary high blood pressure and heart failure [78] and in patients with
glands [50] and some cancer cells, including breast and prostate chronic renal failure, and cardiovascular disease is more common
cancers [51,62], two tumors with high bone metastatic potential. in osteoporotic patients [79]. However, OPG does not appear to
No humans with osteopetrosis have been identified to date with protect the skeleton against the increased bone resorption in pa-
mutations in rank. However, a deletion mutation that occurred tients with renal failure and associated secondary hyperparathy-
spontaneously in rank was reported in transgenic mice. These mice roidism mediated by PTH. Further studies will be required to
had all of the features of mice with targeted deletion of RANK, con- determine the significance of high serum OPG levels in such pa-
firming the importance of RANK for osteoclastogenesis [63]. In tients because they raise questions about the importance of the
contrast, activating mutations in exon 1 of rank have been reported RANKL/OPG ratio in serum samples being predictive of bone
in humans to account for the increased osteoclast formation, activ- mass and bone resorption in these settings [80].
B.F. Boyce, L. Xing / Archives of Biochemistry and Biophysics 473 (2008) 139–146 143

Transcription factor activation by RANKL/RANK in osteoclasts tivity of osteoclast differentiation to RANKL signaling. RGS10 /
and OCPs mice exhibit severe osteopetrosis and impaired osteoclast differen-
tiation. Deficiency of RGS10 resulted in the absence of [Ca2+]i oscil-
A key preliminary step in downstream signaling after RANKL lations and loss of NFATc1, which can be rescued by over-
ligation to RANK is the binding of TNF receptor-associated factors expression of NFATc1. RGS10 competitively interacts with Ca2+/
(TRAFs) to specific sites in the cytoplasmic domain of RANK calmodulin and PIP3 in a [Ca2+]i-dependent manner to mediate
[32,81]. RANK is a transmembrane protein, which like other TNF PLCc activation and [Ca2+]i oscillations in osteoclasts [94]. These
family receptors has no intrinsic protein kinase activating activity studies indicate that RGS10 specifically regulates RANKL-evoked
to mediate signaling. TRAFs 2, 5 and 6 all bind to RANK [81], but RGS10/calmodulin–[Ca2+]i oscillation–calcineurin–NFATc1 signal-
only TRAF6 appears to have essential functions in OCPs and osteo- ing in osteoclast differentiation and may be a potential therapeutic
clasts, since deletion of only TRAF6 and no other TRAFs results in target for the treatment of bone diseases in which bone resorption
osteopetrosis [82,83]. TRAF6-deficient mice produced by two inde- is increased.
pendent groups of investigators developed osteopetrosis [82,83].
Surprisingly, however, one set of mice has normal numbers of
osteoclasts, but they are inactive [82], while the other has no Immunoreceptors, osteoimmunology and RANKL
osteoclasts [83]. How inactivation of TRAF6 resulted in two differ-
ent osteoclast phenotypes remains unexplained. Several signaling Activation of calcium signaling during osteoclast formation
pathways are activated by RANK/TRAF-mediated protein kinase appears to involve the Fc receptor common c subunit (FcRc)
signaling; 4 directly mediate osteoclasts formation (inhibitor of immunoreceptor expressed by osteoclasts and the adapter pro-
NF-jB kinase (IKK)/NF- jB, c-Jun N-terminal kinase (JNK)/activator tein, DNAX-activating protein 12 (DAP12), which associates with
protein-1 (AP-1), c-myc, and calcineurin/NFATc1) and 3 mediate an immunoreceptor tyrosine-based activation motif (ITAM) [95].
osteoclast activation (Src and MKK6/p38/MITF) and survival (Src DAP12/FcRc double knockout mice are severely osteopetrotic due
and extracellular signal-regulated kinase) [84]. to impaired RANKL-induced NFATc1 activation and they do not
Several adapter molecules bind to the intracytoplasmic domain form osteoclasts, while mice deficient in either gene have only
of RANK along with TRAFs to mediate signaling. These include mildly impaired osteoclast formation [96]. Despite the essential
Grb-2-associated binder (Gab) protein 2, a member of a family of functions of DAP12/FcRc, this receptor-mediated signaling path-
proteins that are phosphorylated at tyrosine residues and recruit way cannot induce osteoclast formation on its own. Like M-
signaling molecules that contain Src homology-2 domains. Gab2- CSF, it is necessary, but not sufficient for osteoclastogenesis.
deficient mice have reduced RANKL-induced osteoclast differentia- FcRc-associating receptors include osteoclast-associated receptor
tion, decreased bone resorption and mild osteopetrosis [85]. The (OSCAR) whose expression in OCPs is regulated by NFATc1 [97].
development of mild, rather than marked osteopetrosis in these In inflammatory bone disorders, signaling through RANKL/
mice suggests that Gab2 plays a significant, but not essential role RANK and these immunoreceptors provides an autocrine
in RANKL-induced osteoclast formation. NFATc1-mediated self-amplifying mechanism to potentially in-
Essential roles for NF-jB, AP-1 and NFATc1 signaling in osteo- crease osteoclast formation beyond that of RANKL alone. This
clast formation was discovered after the generation of mice with mechanism for enhanced osteoclast formation in inflammatory
targeted deletion of the genes encoding the precursor molecules bone diseases can also be augmented by TNF and other cyto-
of both p50 and p52 sub-units of NF-jB [34,86], c-Fos [87], and kines in a variety of ways. For example, TNF induces expression
NFATc1 [36]. Over-expression of c-Fos rescues the defect in osteo- of c-fms, the receptor for M-CSF, by OCPs and increases OCP pro-
clast formation in M-CSF-treated NF-jB p50/p52 double knockout liferation and survival [98]. It also enhances OCP egression from
osteoclast precursors in the absence of RANKL [37], and expression the bone marrow into the bloodstream from where they can
of NFATc1 in Fos / OCPs rescues their defect in differentiation alight in increased numbers at sites of inflammation [49]. These
[35]. Activation of c-Fos by RANKL signaling requires expression induced OCPs also increase their production of TNF in response
of NF-jB p50 and p52, indicating that c-Fos and NFATc1 [37] are to RANKL and TNF (our unpublished observation). As mentioned
downstream from NF-jB (Fig. 1). On the basis of all these studies, earlier, TNF also induces c-Fos expression in OCPs through NF-jB
NFATc1 has been described as the master regulator of osteoclast to induce osteoclast formation directly and to facilitate direct
formation [36]. It is activated transiently in OCPs within 60 min induction of osteoclastogenesis by IL-1 (Fig. 1) [39,99]. By com-
of treatment with RANKL when it interacts with NF-jB p65 [88] parative microarray analysis of RNA from OCPs isolated from
and again during the fusion phase around 50–60 h later by cal- the blood of TNF transgenic (TNF-Tg) mice with established
cium-dependent calcineurin dephosphorylation. Several additional inflammatory erosive joint disease and from wild type litter-
factors that are activated by RANKL and also participate in NFATc1 mates, we found that vascular endothelial growth factor
up-regulation include c-Fos and RNA polymerase II [89]. Cyclospor- (VEGF)-C expression is significantly increased in OCPs from the
ine A, a calcineurin inhibitor, inhibits NFATc1 activation; yet treat- TNF-Tg mice [100]. Interestingly, RANKL significantly increases
ment of patients with this immunosuppressant is associated with the expression VEGF-C in osteoclasts and OCPs, and VEGF-C
bone loss [90]. NFATc1 also positively regulates expression of stimulates osteoclast bone resorption. Furthermore, blockade of
osterix, a transcription factor that regulates osteoblast differentia- the VEGF-C receptor signaling reduces RANKL-induced osteoclast
tion and function [91]. Thus the likely reason why cyclosporine A bone resorption [101]. Thus, VEGF-C is another RANKL target
induces bone loss in vivo is that it has a greater inhibitory effect gene and affects osteoclast function through an autocrine
on osteoblasts than on osteoclasts [92]. mechanism.
Recently, signaling molecules other than the transcription fac- By all of these mechanisms, cytokines can induce self-ampli-
tors mentioned above have been implicated in mediating the effect fying autocrine and paracrine cycles in OCPs and osteoclasts to
of RANKL/RANK in osteoclasts. Differential screening of a human induce increased osteoclast formation and activation and aggres-
osteoclastoma cDNA library demonstrated that the regulator of sive bone loss. It is likely that osteoclasts and their precursors
G-protein signaling 10 (RGS10) is specifically expressed in osteo- secrete many more factors that interact with immune and other
clasts [93]. The expression of RGS10 is induced by RANKL in OCPs, cells to affect bone volume and turnover in a variety of bone
and ectopic expression of RGS10 dramatically increases the sensi- disorders.
144 B.F. Boyce, L. Xing / Archives of Biochemistry and Biophysics 473 (2008) 139–146

Accessory cells
Activated T cells, Synovial cells
Endothelial cells, Cancer cells

TNF
RANKL IL-1

TNFR

IL-1R
TRAF2
My
NK

D8 Auto-amplification
6 8
RA

AF
TR
IKKs JNK
p38/Erk

NF-κB

c-Fos

NFATc1
Osteoclast
precursor
Differentiation of osteoclast precursor
Fig. 1. Signaling pathways involved osteoclastogenesis in disease states leading to activation of NFATc1. In inflammatory conditions, such as rheumatoid arthritis, the
numbers of immune and accessory cells are increased in affected joints. Some of these cells produce RANKL in response to locally elevated levels of pro-inflammatory
cytokines and other inflammatory mediators. RANKL binds to RANK on the surface of osteoclast precursors and recruits the adapter protein, TRAP6, leading to activation of
NF-jB through phosphorylation and inactivation of inhibitory kappa kinases (IKKs) and NF-jB inhibitory kinase (not shown here). This induces activation of c-Fos. NF-jB and
c-Fos interact with the NFATc1 promoter to trigger NFATc1 auto-amplification of NFATc1 and the transcription of genes, which mediate completion of the differentiation
process. In addition to RANKL expression, these cells as well as macrophage/monocytes and osteoclasts themselves produce large amounts of TNF. TNF binds to the TNF
receptor and this also activates c-Fos through both the NF-jB and JNK pathways in osteoclast precursors. TNF also stimulates its own expression and that of IL-1 by OCPs
(orange lines). IL-1 does not activate c-Fos, but in OCPs in which c-Fos has been activated, for example by RANKL or TNF, IL-1 can induce osteoclastogenesis directly. This leads
to more osteoclast formation using the same NFATc1 activated mechanism as RANKL. In this model, RANKL/RANK signaling is essential for OCP differentiation under both
physiologic (through osteoblastic cells) and pathologic conditions (through accessory cells and OCP themselves), while TNF signaling appears to play a major role in
inflammatory bone diseases.

Pharmacologic inhibition of RANKL/RANK signaling multiple myeloma and metastatic bone disease [43,109]. To date,
treatment of patients in these studies has resulted in significant
Numerous preclinical in vivo studies using inhibitors of RANKL/ inhibition of bone resorption without any obvious significant ad-
RANK signaling have confirmed the important roles of this system verse effects.
in rodents and non-human primates. For example, OPG and
RANK:Fc inhibited bone loss in models of sex-steroid deficiency Summary
and glucocorticoid-induced osteoporosis, rheumatoid arthritis,
multiple myeloma, and metastatic bone disease [102–105]. These Discovery of the RANKL/RANK/OPG system has been one of the
studies were followed by phase 1 clinical trials of two forms of most important advances in bone biology in the last decade. This
OPG: Fc-OPG and OPG-Fc, Single injections of these OPG constructs signaling system is essential for skeletal homeostasis, and disrup-
into normal volunteers resulted in prolonged dose-dependent tion of it leads to inhibition of bone resorption in vitro and in ani-
reductions in biochemical markers of bone resorption [106,107]. mal models of most bone diseases characterized by increased
Neither of these proteins was developed for further clinical trials, resorption. RANKL/RANK signaling plays important roles in tissues
perhaps because of concerns about the possibility of immune re- other than bone. Elucidation of the specific roles of RANKL/RANK in
sponses to them and consequent unwanted adverse effects on these various types of cells will likely link bone remodeling in nor-
the immune system. They were replaced by Denosumab, a fully hu- mal and disease states with regulation of the function of other or-
man monoclonal antibody developed by injecting mice with hu- gan systems in health and disease. The discovery of RANKL, RANK
man RANKL, which binds to and inactivates RANKL, similar to and OPG has led to the development of specific inhibitors of
the action of OPG. Compared to OPG:Fc, Denosumab has a signifi- RANKL, some of which, such as OPG and a monoclonal antibody
cantly longer circulating half-life and a more prolonged effect to to RANKL, have been tested in humans in clinical trials with suc-
reduce serum levels of markers of bone resorption and formation cessful inhibition of bone resorption. Thus, it will be important to
[108]. There are several phase 2 and 3 clinical trails investigating determine if long-term inhibition of RANKL has any unwanted ad-
the efficacy of Denosumab in patients with a variety of bone disor- verse effects in these tissues and on immune responses in
ders, including postmenopausal osteoporosis, rheumatoid arthritis, particular.
B.F. Boyce, L. Xing / Archives of Biochemistry and Biophysics 473 (2008) 139–146 145

References [42] M. Yagi, T. Miyamoto, Y. Sawatani, K. Iwamoto, N. Hosogane, N. Fujita, K.


Morita, K. Ninomiya, T. Suzuki, K. Miyamoto, et al., J. Exp. Med. 202 (2005)
345–351.
[1] G. Karsenty, E.F. Wagner, Dev. Cell 2 (2002) 389–406.
[43] A.E. Kearns, S. Khosla, P. Kostenuik, Endocr. Rev. (2007).
[2] A. Del Fattore, A. Cappariello, A. Teti, Genetics, pathogenesis and
[44] T. Wada, T. Nakashima, N. Hiroshi, J.M. Penninger, Trends Mol. Med. 12 (2006)
complications of osteopetrosis, Bone 42 (2008) 19–29.
17–25.
[3] J. Tolar, S.L. Teitelbaum, P.J. Orchard, N. Engl. J. Med. 351 (2004) 2839–2849.
[45] H. Takayanagi, Nat. Rev. Immunol. 7 (2007) 292–304.
[4] J.B. Lian, G.S. Stein, Curr. Pharm. Des. 9 (2003) 2677–2685.
[46] T. Ikeda, M. Kasai, M. Utsuyama, K. Hirokawa, Endocrinology 142 (2001)
[5] G. Karsenty, Semin. Cell Dev. Biol. 11 (2000) 343–346.
1419–1426.
[6] P. Ducy, R. Zhang, V. Geoffroy, A.L. Ridall, G. Karsenty, Cell 89 (1997) 747–754.
[47] C.C. Lynch, A. Hikosaka, H.B. Acuff, M.D. Martin, N. Kawai, R.K. Singh, T.C.
[7] Y. Komori, H. Yagi, S. Nomura, A. Yamaguchi, K. Sasaki, K. Deguchi, Y. Shimizu,
Vargo-Gogola, J.L. Begtrup, T.E. Peterson, B. Fingleton, et al., Cancer Cell 7
R.T. Bronson, Y.H. Gao, M. Inada, et al., Cell 89 (1997) 755–764.
(2005) 485–496.
[8] M.Q. Hassan, R.S. Tare, S.H. Lee, M. Mandeville, M.I. Morasso, A. Javed, A.J. van
[48] A. Hikita, I. Yana, H. Wakeyama, M. Nakamura, Y. Kadono, Y. Oshima, K.
Wijnen, J.L. Stein, G.S. Stein, J.B. Lian, J. Biol. Chem. 281 (2006) 40515–40526.
Nakamura, M. Seiki, S. Tanaka, J. Biol. Chem. (2006).
[9] M.H. Drissi, X. Li, T.J. Sheu, M.J. Zuscik, E.M. Schwarz, J.E. Puzas, R.N. Rosier, R.J.
[49] P. Li, E.M. Schwarz, R.J. O’Keefe, L. Ma, R.J. Looney, C.T. Ritchlin, B.F. Boyce, L.
O’Keefe, J. Cell. Biochem. 90 (2003) 1287–1298.
Xing, Arthritis Rheum. 50 (2004) 265–276.
[10] J.Q. Feng, L. Xing, J.H. Zhang, M. Zhao, D. Horn, J. Chan, B.F. Boyce, S.E. Harris,
[50] J.E. Fata, Y.-Y. Kong, J. Li, T. Sasaki, J. Irie-Sasaki, R.A. Moorehead, R. Elliott, S.
G.R. Mundy, D. Chen, J. Biol. Chem. 278 (2003) 29130–29135.
Scully, E.B. Voura, D.L. Lacey, Cell 103 (2000) 41–50.
[11] T.H. Vu, J.M. Shipley, G. Bergers, J.E. Berger, J.A. Helms, D. Hanahan, S.D.
[51] N.S. Kim, H.J. Kim, B.K. Koo, M.C. Kwon, Y.W. Kim, Y. Cho, Y. Yokota, J.M.
Shapiro, R.M. Senior, Z. Werb, Cell 93 (1998) 411–422.
Penninger, Y.Y. Kong, Mol. Cell. Biol. 26 (2006) 1002–1013.
[12] J.W. Rhee, K.W. Lee, W.J. Sohn, Y. Lee, O.H. Jeon, H.J. Kwon, D.S. Kim, Mol.
[52] D.H. Jones, T. Nakashima, O.H. Sanchez, I. Kozieradzki, S.V. Komarova, I.
Immunol. 44 (2007) 1393–1400.
Sarosi, S. Morony, E. Rubin, R. Sarao, C.V. Hojilla, et al., Nature 440 (2006)
[13] R. Masuyama, I. Stockmans, S. Torrekens, R. Van Looveren, C. Maes, P.
692–696.
Carmeliet, R. Bouillon, G. Carmeliet, J. Clin. Invest. 116 (2006) 3150–3159.
[53] H. Takayanagi, S. Kim, K. Matsuo, H. Suzuki, T. Suzuki, K. Sato, T. Yokochi, H.
[14] M. Usui, L. Xing, H. Drissi, M. Zuscik, R. O’Keefe, D. Chen, B.F. Boyce, J. Bone
Oda, K. Nakamura, N. Ida, et al., Nature 416 (2002) 744–749.
Miner. Res. (2007).
[54] H. Takayanagi, K. Ogasawara, S. Hida, T. Chiba, S. Murata, K. Sato, A. Takaoka,
[15] V. Breuil, H. Schmid-Antomarchi, A. Schmid-Alliana, R. Rezzonico, L. Euller-
T. Yokochi, H. Oda, K. Tanaka, et al., Nature 408 (2000) 600–605.
Ziegler, B. Rossi, FASEB J. 17 (2003) 1751–1753.
[55] H. Chen, C.A. Gilbert, J.A. Hudson, S.C. Bolick, K.L. Wright, J.F. Piskurich, Mol.
[16] B.A. Mosheimer, N.C. Kaneider, C. Feistritzer, D.H. Sturn, C.J. Wiedermann,
Immunol. 44 (2007) 1461–1470.
Arthritis Rheum. 50 (2004) 2309–2316.
[56] H. Takayanagi, S. Kim, T. Taniguchi, Signaling crosstalk between RANKL and
[17] Y.Y. Kong, H. Yoshida, I. Sarosi, H.L. Tan, E. Timms, C. Capparelli, S.
interferons in osteoclast differentiation, Arthritis Res. 4 (Suppl 3) (2002)
Morony, A.J. Oliveira-dos-Santos, G. Van, A. Itie, et al., Nature 397 (1999)
S227–S232.
315–323.
[57] P.R. Madyastha, S. Yang, W.L. Ries, L.L. Key Jr., J. Interferon Cytokine Res. 20
[18] W.C. Dougall, M. Glaccum, K. Charrier, K. Rohrbach, K. Brasel, T. De Smedt, E.
(2000) 645–652.
Daro, J. Smith, M.E. Tometsko, C.R. Maliszewski, et al., Genes Dev. 13 (1999)
[58] L.L. Key Jr., R.M. Rodriguiz, S.M. Willi, N.M. Wright, H.C. Hatcher, D.R. Eyre, J.K.
2412–2424.
Cure, P.P. Griffin, W.L. Ries, N. Engl. J. Med. 332 (1995) 1594–1599.
[19] B.F. Boyce, L. Xing, W. Shakespeare, Y. Wang, D. Dalgarno, J. Iuliucci, T.
[59] Y. Gao, F. Grassi, M.R. Ryan, M. Terauchi, K. Page, X. Yang, M.N. Weitzmann, R.
Sawyer, Kidney Int. Suppl. (2003) 2–5.
Pacifici, J. Clin. Invest. 117 (2007) 122–132.
[20] S. Tatsumi, K. Ishii, N. Amizuka, M. Li, T. Kobayashi, K. Kohno, M. Ito, S.
[60] L. Van Wesenbeeck, P.R. Odgren, F.P. Coxon, A. Frattini, P. Moens, B. Perdu,
Takeshita, K. Ikeda, Cell Metab. 5 (2007) 464–475.
C.A. MacKay, E. Van Hul, J.P. Timmermans, F. Vanhoenacker, et al., J. Clin.
[21] B.F. Boyce, L. Xing, Arthritis Res. Ther. 9 (1) (2007) S1.
Invest. 117 (2007) 919–930.
[22] C. Zhao, N. Irie, Y. Takada, K. Shimoda, T. Miyamoto, T. Nishiwaki, T. Suda, K.
[61] C. Sobacchi, A. Frattini, M.M. Guerrini, M. Abinun, A. Pangrazio, L. Susani, R.
Matsuo, Cell Metab. 4 (2006) 111–121.
Bredius, G. Mancini, A. Cant, N. Bishop, et al., Nat. Genet. 39 (2007) 960–962.
[23] S.H. Lee, J. Rho, D. Jeong, J.Y. Sul, T. Kim, N. Kim, J.S. Kang, T. Miyamoto, T.
[62] G. Chen, K. Sircar, A. Aprikian, A. Potti, D. Goltzman, S.A. Rabbani, Cancer 107
Suda, S.K. Lee, et al., Nat. Med. 12 (2006) 1403–1409.
(2006) 289–298.
[24] O. Kollet, A. Dar, S. Shivtiel, A. Kalinkovich, K. Lapid, Y. Sztainberg, M. Tesio,
[63] R.P. Kapur, Z. Yao, M.H. Iida, C.M. Clarke, B. Doggett, L. Xing, B.F. Boyce, J. Bone
R.M. Samstein, P. Goichberg, A. Spiegel, et al., Nat. Med. 12 (2006) 657–664.
Miner. Res. 19 (2004) 1689–1697.
[25] L. Xing, E.M. Schwarz, B.F. Boyce, Immunol. Rev. 208 (2005) 19–29.
[64] A.E. Hughes, S.H. Ralston, J. Marken, C. Bell, H. MacPherson, R.G. Wallace, W.
[26] H. Yoshida, S. Hayashi, T. Kunisada, M. Ogawa, S. Nishikawa, H. Okamura, T.
van Hul, M.P. Whyte, K. Nakatsuka, L. Hovy, et al., Nat. Genet. 24 (2000) 45–
Sudo, L.D. Shultz, Nature 345 (1990) 442–444.
48.
[27] W.S. Simonet, D.L. Lacey, C.R. Dunstan, M. Kelley, M.S. Chang, R. Luthy, H.Q.
[65] Y. Li, G. Toraldo, A. Li, X. Yang, H. Zhang, W.P. Qian, M.N. Weitzmann, Blood
Nguyen, S. Wooden, L. Bennett, T. Boone, et al., Cell 89 (1997) 309–319.
109 (2007) 3839–3848.
[28] H. Yasuda, N. Shima, N. Nakagawa, S.I. Mochizuki, K. Yano, N. Fujise, Y. Sato,
[66] L.C. Hofbauer, M. Schoppet, JAMA 292 (2004) 490–495.
M. Goto, K. Yamaguchi, M. Kuriyama, et al., Endocrinology 139 (1998) 1329–
[67] S. Theoleyre, Y. Wittrant, S.K. Tat, Y. Fortun, F. Redini, D. Heymann, Cytokine
1337.
Growth Factor Rev. 15 (2004) 457–475.
[29] H. Yasuda, N. Shima, N. Nakagawa, K. Yamaguchi, M. Kinosaki, S. Mochizuki,
[68] M.P. Whyte, S.E. Obrecht, P.M. Finnegan, J.L. Jones, M.N. Podgornik, W.H.
A. Tomoyasu, K. Yano, M. Goto, A. Murakami, et al., Proc. Natl. Acad. Sci. USA
McAlister, S. Mumm, N. Engl. J. Med. 347 (2002) 175–184.
95 (1998) 3597–3602.
[69] T. Cundy, M. Hegde, D. Naot, B. Chong, A. King, R. Wallace, J. Mulley, D.R. Love,
[30] D.L. Lacey, E. Timms, H.L. Tan, M.J. Kelley, C.R. Dunstan, T. Burgess, R. Elliott, A.
J. Seidel, M. Fawkner, et al., Hum. Mol. Genet. 11 (2002) 2119–2127.
Colombero, G. Elliott, S. Scully, et al., Cell 93 (1998) 165–176.
[70] D.A. Glass 2nd, P. Bialek, J.D. Ahn, M. Starbuck, M.S. Patel, H. Clevers, M.M.
[31] D.M. Anderson, E. Maraskovsky, W.L. Billingsley, W.C. Dougall, M.E.
Taketo, F. Long, A.P. McMahon, R.A. Lang, et al., Dev. Cell 8 (2005) 751–764.
Tometsko, E.R. Roux, M.C. Teepe, R.F. DuBose, D. Cosman, L. Galibert, Nature
[71] M. Kieslinger, S. Folberth, G. Dobreva, T. Dorn, L. Croci, R. Erben, G.G.Consalez,
390 (1997) 175–179.
R. Grosschedl, EBF2 regulates osteoblast-dependent differentiation of
[32] B.R. Wong, J. Rho, J. Arron, E. Robinson, J. Orlinick, M. Chao, S. Kalachikov, E.
osteoclasts, Cell Metab., 2005.
Cayani, F.S. Bartlett III, W.N. Frankel, et al., J. Biol. Chem. 272 (1997) 25190–
[72] T.P. Hill, D. Spater, M.M. Taketo, W. Birchmeier, C. Hartmann, Dev. Cell 8
25194.
(2005) 727–738.
[33] B.R. Wong, J. Rho, J. Arron, E. Robinson, J. Orlinick, M. Chao, S. Kalachikov, E.
[73] S. Bai, R. Kopan, W. Zou, M.J. Hilton, C.T. Ong, F. Long, F.P. Ross, S.L.
Cayani, F.S. Bartlett III, W.N. Frankel, et al., J. Bone Miner. Res. 15 (2000)
Teitelbaum, J. Biol. Chem. (2007).
2293–2296.
[74] N. Bucay, I. Sarosi, C.R. Dunstan, S. Morony, J. Tarpley, C. Capparelli, S. Scully,
[34] G. Franzoso, L. Carlson, L. Xing, L. Poljak, E.W. Shores, K.D. Brown, A. Leonardi,
H.L. Tan, W. Xu, D.L. Lacey, et al., Genes Dev. 12 (1998) 1260–1268.
T. Tran, B.F. Boyce, U. Siebenlist, Genes Dev. 11 (1997) 3482–3496.
[75] B.J. Bennett, M. Scatena, E.A. Kirk, M. Rattazzi, R.M. Varon, M. Averill, S.M.
[35] K. Matsuo, D.L. Galson, C. Zhao, L. Peng, C. Laplace, K.Z. Wang, M.A. Bachler, H.
Schwartz, C.M. Giachelli, M.E. Rosenfeld, Arterioscler. Thromb. Vasc. Biol. 26
Amano, H. Aburatani, H. Ishikawa, et al., J. Biol. Chem. 279 (2004) 26475–
(2006) 2117–2124.
26480.
[76] S. Morony, Y. Tintut, Z. Zhang, R.C. Cattley, G. Van, D. Dwyer, M. Stolina, P.J.
[36] H. Takayanagi, S. Kim, T. Koga, H. Nishina, M. Isshiki, H. Yoshida, A. Saiura, M.
Kostenuik, L.L. Demer, Circulation 117 (2008) 411–420.
Isobe, T. Yokochi, J. Inoue, et al., Dev. Cell 3 (2002) 889–901.
[77] P. Collin-Osdoby, Circ. Res. 95 (2004) 1046–1057.
[37] T. Yamashita, Z. Yao, F. Li, Q. Zhang, I.R. Badell, E.M. Schwarz, S. Takeshita, E.F.
[78] T. Ueland, A. Yndestad, E. Oie, G. Florholmen, B. Halvorsen, S.S. Froland, S.
Wagner, M. Noda, K. Matsuo, et al., J. Biol. Chem. 282 (2007) 18245–18253.
Simonsen, G. Christensen, L. Gullestad, P. Aukrust, Circulation 111 (2005)
[38] N. Kim, Y. Kadono, M. Takami, J. Lee, S.H. Lee, F. Okada, J.H. Kim, T. Kobayashi,
2461–2468.
P.R. Odgren, H. Nakano, et al., J. Exp. Med. 202 (2005) 589–595.
[79] A. Sammartino, D. Cirillo, V.D. Mandato, C. Di Carlo, C. Nappi, J. Endocrinol.
[39] Z. Yao, L. Xing, C. Qin, E.M. Schwarz, B.F. Boyce, Osteoclast precursor
Invest. 28 (2005) 80–84.
interaction with bone matrix induces osteoclast formation directly by an
[80] A. Rogers, R. Eastell, J. Clin. Endocrinol. Metab. 90 (2005) 6323–6331.
IL-1-mediated autocrine mechanism, J. Biol. Chem. 283 (15) (2008)
[81] H.H. Kim, D.E. Lee, J.N. Shin, Y.S. Lee, Y.M. Jeon, C.H. Chung, J. Ni, B.S. Kwon,
9917–9924.
Z.H. Lee, FEBS Lett. 443 (1999) 297–302.
[40] S.L. Teitelbaum, F.P. Ross, Nat. Rev. Genet. 4 (2003) 638–649.
[82] M.A. Lomaga, W.C. Yeh, I. Sarosi, G.S. Duncan, C. Furlonger, A. Ho, S. Morony,
[41] S.L. Teitelbaum, Ann. NY Acad. Sci. 1068 (2006) 95–99.
C. Capparelli, G. Van, S. Kaufman, et al., Genes Dev. 13 (1999) 1015–1024.
146 B.F. Boyce, L. Xing / Archives of Biochemistry and Biophysics 473 (2008) 139–146

[83] A. Naito, S. Azuma, S. Tanaka, T. Miyazaki, S. Takaki, K. Takatsu, K. Nakao, K. [98] Z. Yao, P. Li, Q. Zhang, E.M. Schwarz, P. Keng, A. Arbini, B.F. Boyce, L. Xing, J.
Nakamura, M. Katsuki, T. Yamamoto, et al., Genes Cells 4 (1999) 353–362. Biol. Chem. 281 (2006) 11846–11855.
[84] W.J. Boyle, W.S. Simonet, D.L. Lacey, Nature 423 (2003) 337–342. [99] Z. Yao, L. Xing, E.M. Schwarz, B.F. Boyce, JBMR 21 (2006) S262.
[85] T. Wada, T. Nakashima, A.J. Oliveira-dos-Santos, J. Gasser, H. Hara, G. Schett, [100] Q. Zhang, Y. Lu, S. Proulx, R. Guo, Z. Yao, E.M. Schwarz, B.F. Boyce, L. Xing,
J.M. Penninger, Nat. Med. 11 (2005) 394–399. Arthritis Res. Ther. 9 (2007) R118.
[86] V. Iotsova, J. Caamano, J. Loy, A. Lewin, R. Bravo, Nat. Med. 3 (1997) 1285–1289. [101] R. Guo, Q. Zhang, Y. Lu, E.M. Schwarz, Z. Jin, B.F. Boyce, L. Xing, JBMR 22
[87] Z.-Q. Wang, C. OVitt, A.E. Grigoriadis, U. Mohle-Steinlein, U. Ruther, E.F. (2007) S30.
Wagner, Nature 360 (1992) 741–745. [102] G. Schett, S. Hayer, J. Zwerina, K. Redlich, J.S. Smolen, Nat. Clin. Pract.
[88] M. Asagiri, K. Sato, T. Usami, S. Ochi, H. Nishina, H. Yoshida, I. Morita, E.F. Rheumatol. 1 (2005) 47–54.
Wagner, T.W. Mak, E. Serfling, et al., J. Exp. Med. 202 (2005) 1261–1269. [103] E.L. Humphrey, J.H. Williams, M.W. Davie, M.J. Marshall, Bone 38 (2006) 652–
[89] J.A. Fretz, N.K. Shevde, S. Singh, B.G. Darnay, J.W. Pike, Mol. Endocrinol. 661.
(2007). [104] J.M. Blair, H. Zhou, M.J. Seibel, C.R. Dunstan, Nat. Clin. Pract. Oncol. 3 (2006)
[90] D. Thiebaud, M.A. Krieg, D. Gillard-Berguer, A.F. Jacquet, J.J. Goy, P. 41–49.
Burckhardt, Eur. J. Clin. Invest. 26 (1996) 549–555. [105] W.C. Dougall, M. Chaisson, Cancer Metastasis Rev. 25 (2006) 541–549.
[91] K. Nakashima, X. Zhou, G. Kunkel, Z. Zhang, J.M. Deng, R.R. Behringer, B. de [106] P.J. Bekker, D. Holloway, A. Nakanishi, M. Arrighi, P.T. Leese, C.R. Dunstan, J.
Crombrugghe, Cell 108 (2002) 17–29. Bone Miner. Res. 16 (2001) 348–360.
[92] T. Koga, Y. Matsui, M. Asagiri, T. Kodama, B. de Crombrugghe, K. Nakashima, [107] J.J. Body, P. Greipp, R.E. Coleman, T. Facon, F. Geurs, J.P. Fermand, J.L.
H. Takayanagi, Nat. Med. 11 (2005) 880–885. Harousseau, A. Lipton, X. Mariette, C.D. Williams, et al., Cancer 97 (2003)
[93] S. Yang, W. Chen, P. Stashenko, Y.P. Li, J. Cell Sci. 120 (2007) 3362–3371. 887–892.
[94] S. Yang, Y.P. Li, Genes Dev. 21 (2007) 1803–1816. [108] P.J. Bekker, D.L. Holloway, A.S. Rasmussen, R. Murphy, S.W. Martin, P.T. Leese,
[95] T. Koga, M. Inui, K. Inoue, S. Kim, A. Suematsu, E. Kobayashi, T. Iwata, H. G.B. Holmes, C.R. Dunstan, A.M. DePaoli, J. Bone Miner. Res. 19 (2004) 1059–
Ohnishi, T. Matozaki, T. Kodama, et al., Nature 428 (2004) 758–763. 1066.
[96] H. Takayanagi, J. Mol. Med. 83 (2005) 170–179. [109] E.M. Lewiecki, P.D. Miller, M.R. McClung, S.B. Cohen, M.A. Bolognese, Y.
[97] Y. Kim, K. Sato, M. Asagiri, I. Morita, K. Soma, H. Takayanagi, J. Biol. Chem. 280 Liu, A. Wang, S. Siddhanti, L.A. Fitzpatrick, J. Bone Miner. Res. 22 (2007)
(2005) 32905–32913. 1832–1841.

S-ar putea să vă placă și