You are on page 1of 13

REVIEWS

Osteoimmunology: shared
mechanisms and crosstalk between
the immune and bone systems
Hiroshi Takayanagi
Abstract | Osteoimmunology is an interdisciplinary research field focused on the molecular
understanding of the interplay between the immune and skeletal systems. Although
osteoimmunology started with the study of the immune regulation of osteoclasts, its scope
has been extended to encompass a wide range of molecular and cellular interactions,
including those between osteoblasts and osteoclasts, lymphocytes and osteoclasts, and
osteoblasts and haematopoietic cells. Therefore, the two systems should be understood to
be integrated and operating in the context of the ‘osteoimmune’ system, a heuristic concept
that provides not only a framework for obtaining new insights by basic research, but also a
scientific basis for the discovery of novel treatments for diseases related to both systems.

Osteoclastogenesis The close relationship between the immune and immune cells and bone cells are derived from the same
A process whereby skeletal systems has long been appreciated, since the origin13. RANKL is not only expressed by the bone-
haematopoietic stem cells pioneering works in the early 1970s (REFS 1–4). Soluble forming osteoblasts that support osteoclastogenesis in
differentiate into
factors secreted from antigen-stimulated immune bone tissue, but also by activated T cells, indicating
multinucleated osteoclasts
with bone-resorbing activity. cells were known as osteoclast-activating factors 1, that osteoclastic bone resorption is influenced by the
one of which was revealed to be interleukin-1 (IL-1)3. immune system12,14. As the analyses of osteoclasts and
Since then, accumulating evidence has indicated that RANKL have been at the forefront of recent progress in
the immune and skeletal systems share a number of osteoimmunology, this Review first focuses on the sig-
regulatory molecules including cytokines, recep- nalling mechanism of RANKL and the role of immune
tors, signalling molecules and transcription factors5,6. mediators in osteoclastic signal transduction.
Furthermore, immune cells are formed and haema- However, M-CSF and RANKL are not the only fac-
topoietic stem cells (HSCs) are maintained in the bone tors linking the immune and skeletal systems. Targeted
marrow where they interact with bone cells. Therefore, disruption of various immunomodulatory molecules
the evidence that the physiology and pathology of one has resulted in unexpected phenotypes in the skel-
system might affect the other is compelling (FIG. 1) and etal system (TABLE 1; Supplementary information S1
Department of Cell Signaling, the term osteoimmunology was coined to cover these (table)). It is worth noting that extracellular matrix
Graduate School, Tokyo overlapping scientific fields7. components, such as osteopontin, also have crucial
Medical and Dental
University, Yushima 1-5-45,
The most typical example of the interaction between roles in both systems. Moreover, recent studies have
Bunkyo-ku, Tokyo 113-8549; the skeletal and immune systems is seen in the abnor- indicated that osteoblasts and even osteoclasts are
COE Program for Frontier mal and/or prolonged activation of the immune system involved in the maintenance and regulation of HSCs,
Research on Molecular in autoimmune diseases, such as rheumatoid arthritis, indicating the crucial role of skeletal cells in both the
Destruction and
that lead to bone destruction caused by bone-resorbing haematopoietic and immune systems15–18. Clinically, the
Reconstruction of Tooth and
Bone, Tokyo Medical and osteoclasts8,9. Osteoclast differentiation is regulated effectiveness of TNF-specific antibody therapy against
Dental University; and by macrophage colony-stimulating factor (M-CSF)10 bone destruction in patients with rheumatoid arthri-
SORST, Japan Science and and receptor activator of nuclear factor-κB (NF-κB) tis also highlights the importance of the relationship
Technology Agency, Honcho ligand (RANKL; also known as TNFSF11), a tumour- between the immune and skeletal systems19. Therefore,
4-1-8, Kawaguchi, Saitama
332-0012, Japan.
necrosis factor (TNF)-family cytokine11,12. A mutation osteoimmunology is becoming increasingly important
e-mail: taka.csi@tmd.ac.jp in the M-CSF gene causes a defect in both macrophage for understanding the pathogenesis of, and developing
doi:10.1038/nri2062 and osteoclast formation, pointing to the notion that new therapeutic strategies for, diseases affecting both

292 | APRIL 2007 | VOLUME 7 www.nature.com/reviews/immunol


© 2007 Nature Publishing Group
REVIEWS

systems. Here I introduce the key cells and molecules Inputs


involved in the interactions between the bone and • Calcium intake
immune systems and summarize recent progress in • Mechanostress
• Ageing
this field. • Stress
• Tumour
Overview of the cells in the skeletal system • Infection
• Inflammation
The bony skeleton, the essential component of the
skeletal system, enables locomotive activity, the storage
of calcium and the harbouring of HSCs from which
immune cells are derived. This multifunctional organ
is characterized by calcified hard tissue composed of Controllers
• Immune system
type I collagen and highly organized deposits of calcium • Other regulatory
phosphate (hydroxyapatite)20. Although it seems to be systems
metabolically inert, bone is restructured at such a high
speed that approximately 10% of the total bone content is
replaced per year in adult vertebrates. This process, called
bone remodelling, is dependent on the dynamic balance
of bone formation and resorption, which are mediated Bone
by osteoblasts and osteoclasts, respectively. A delicate Bone marrow
regulation of this process is a prerequisite for normal
Haematopoietic cells
bone homeostasis (TABLE 1; Supplementary information
S1 (table)), and an imbalance is often linked to meta-
bolic bone diseases in humans, such as osteoporosis and
inflammatory bone loss21. Osteoblast Osteoclast
Osteoblasts are cells of mesenchymal origin that
secrete bone-matrix proteins and promote mineral-
ization20,21. The proliferation and differentiation of Osteocyte
osteoblasts are under the control of a number of solu-
ble factors and transcription factors such as RUNX2
(runt-related transcription factor 2) and OSTERIX
(also known as SP7) 22,23. Differentiated osteoblasts
Outputs
embedded in the bone matrix are called osteocytes, • Calcium metabolism
and might have a specific but as-yet unclear role in • Bone mass/quality
mechanotransduction20. (locomotion and
body support)
Osteoclasts are cells of haematopoietic origin that • Haematopoiesis
decalcify and degrade the bone matrix by acid decal-
cification and proteolytic degradation, respectively24. Figure 1 | The osteoimmune system. The skeletal
They are large, multinucleated cells formed by the system is involved in the regulation of three main
fusion of precursor cells of the monocyte–macrophage outputs that are related to calcium reserves,
locomotion and haematopoiesis. To maintain
lineage. In vitro osteoclast differentiation is supported
homeostasis while responding to various inputs
by mesenchymal cells (bone-marrow stromal cells or (such as nutrition, mechanical stress, ageing, and
Osteoporosis osteoblasts) through cell–cell contact25, although there inflammation), the cells in the bone marrow are
A metabolic or ageing-related has been little in vivo information about osteoclasto- controlled by the immune systems in concert with
(often occurring in post- genesis-supporting cells. Osteoclastogenic signals are other regulatory systems, such as the endocrine and
menopausal women) disease in
mediated by RANKL and its co-stimulatory signals, neural systems. As depicted, the bone system and the
which low bone mineral density
causes bone fragility. in addition to M-CSF 5,11,12. The congenital lack of immune system in the bone-marrow microenvironment
osteoclasts leads to osteopetrosis, which is character- are regulated as if they were integrated in the context
Mechanotransduction ized by a high bone mass and a defect in bone-marrow of the osteoimmune system.
A process by which the formation. Naturally occurring mutant mice or geneti-
mechanical stress (such as
gravity, loading and tension)
cally modified mice with osteopetrosis have provided
is converted to biological insights into the molecular mechanism of osteoclast M-CSF
responses, which in this case differentiation and function26 (TABLE 1; Supplementary M-CSF was first identified as an essential factor for
control bone remodelling. information S1 (table)): M-CSF and the transcription osteoclastogenesis. M-CSF transmits its signal to the cell
factor PU.1 are crucial for the proliferation and survival through the specific receptor cFMS, which is a member
Osteopetrosis
A rare congenital disease of osteoclast precursor cells; transcription factors such of the receptor tyrosine kinase superfamily. M-CSF is cru-
with extremely high bone mass as cFOS, microphthalmia-associated transcription fac- cial for the proliferation and survival of precursor cells
and low strength (typically, tor (MITF) and NF-κB have been shown to be essential of osteoclasts as well as macrophages, mainly by activat-
no bone-marrow formation for osteoclast differentiation; and factors such as cSRC, ing ERK (extracellular-signal-regulated kinase) through
and no tooth eruption),
which results from impaired
VAV3, β3-integrin, chloride-channel family member GRB2 (growth-factor-receptor-bound protein 2) and
differentiation or function of ClC7, vacuolar ATPase and cathepsin K are crucial for AKT through PI3K (phosphoinositide 3-kinase) 10.
osteoclasts. osteoclast function26. M-CSF also stimulates the expression of RANK in

NATURE REVIEWS | IMMUNOLOGY VOLUME 7 | APRIL 2007 | 293


© 2007 Nature Publishing Group
REVIEWS

Familial expansile osteolysis monocyte–macrophage precursor cells, thereby rendering now called RANKL, or TNF-related activation-induced
(FEO). A rare autosomal- them able to efficiently respond to RANKL. PU.1, a mem- cytokine (TRANCE), according to the nomenclature
dominant disorder resembling ber of the ETS family of transcription factors, regulates the of the immune system. RANKL, a type II membrane
Paget’s disease of bone (PDB), development of macrophages and osteoclasts by control- protein, belongs to the TNF superfamily and contains a
characterized by the erosion of
long bones by progressive
ling the expression of cFMS27. MITF, activated by M-CSF, C-terminal receptor-binding domain and a transmem-
osteoclastic resorption (the induces BCL-2 (B-cell lymphoma 2), which has a pivotal brane domain. Its receptor, RANK, is a type I membrane
constitutive active mutation role in cell survival28. The transgenic expression of BCL-2 protein sharing high homology with CD40, and the
of RANK has been reported). rescues the osteopetrosis found in op/op mice (which are binding of RANKL to RANK is inhibited by the decoy
deficient in M-CSF), indicating the main role of M-CSF as receptor osteoprotegerin (OPG)34,35. In bone, RANKL is
Paget’s disease of the bone
(PDB). A metabolic bone
a survival factor of osteoclast precursor cells29. expressed by osteoclastogenesis-supporting cells, includ-
disorder in which focal ing osteoblasts, in response to osteoclastogenic factors,
abnormalities of increased RANKL such as 1,25-dihydroxyvitamin D 3 (1,25(OH) 2 D 3 ),
bone turnover (excessive The crucial role of RANKL in bone. It was unex- prostaglandin E2 (PGE2) and parathyroid hormone, and
osteoclastic bone resorption
and irregular bone formation)
pected both for bone biologists and for immunolo- is a crucial determinant of the level of bone resorption
lead to bone pain and gists that the long-sought osteoclast-differentiation in vivo25. A series of genetically modified mice has clearly
deformity. factor expressed by osteoblasts would be the same shown that RANKL and its receptor RANK are indispen-
molecule expressed by T cells to stimulate dendritic sable for osteoclastogenesis12,36, and human mutations
cells (DCs)30–33. This osteoclast-differentiation factor is in RANK and OPG cause familial expansile osteolysis and

Table 1 | Bone phenotypes in mice deficient in osteoimmunoregulatory molecules


Disrupted osteoimmunomodulatory Osteoclast-mediated bone resorption Osteoblast-mediated Bone volume References
molecule bone formation
Differentiation Function
Cytokine or secreted protein
RANKL ↓ ND ND ↑ 12
Receptors, channels and membrane factors
DAP12/FcRγ ↓ ↓ ↓ ↑ 99
DC-STAMP ↓ ↓ ND ↑ 94
Plexin-A1 ↓ ↓ ↔ ↑ 97
IFNα/βR1 ↑ ND ↔ ↓ 72
IFNγR1 ↑* ND ND ND 73
EPHB4‡ ↓ ↓ ↑ ↑ 96
Adaptor and signalling molecules
TRAF6 ↓ ND ND ↑ 49
GAB2 ↓ ND ↔ ↑ 56
STAT1 ↑ ND ↑ ↑ 88
SOCS1/3 ↓* ND ND ND 90
Kinases and phosphatases
NIK ↓* ND ND ↔ 76
IKKβ ↓ ND ↓ ↑ 75
v
SHP1 (me /me ) v §
↑ ND ↓ ↓ 102
SHIP1 ↑ ↑ ND ↓ 104
Transcription factors
NF-κB p50/p52 ↓ ND ↔ ↑ 53
NFATc1 ↓ ND ↓ ↑ 63
NFATc2 ↔ ↔ ↓ ↓ 81
SHN3 ↔ ↔ ↑ ↑ 91
*Analysed in models of inflammatory bone loss. ‡Data in transgenic mice. §Naturally occurring mutant in brackets. See the full list of mutant mice in Supplementary
information S1 (table). ↑, increased; ↓, decreased; ↔, unchanged; DC-STAMP, dendritic-cell-specific transmembrane protein; EPHB4, ephrin receptor B4;
FcRγ, Fc receptor common γ-subunit; GAB2, growth-factor-receptor-bound-protein-2-associated binding protein 2; IFN, interferon; IKK-β, inhibitor of nuclear
factor-κB (IκB) kinase-β; ND, not determined; NFAT, nuclear factor of activated T cells; NIK, NF-κB-inducing kinase; RANKL, receptor activator of NF-κB ligand;
SHN3, Schnurri-3; SHIP1, SH2-domain-containing inositol-5-phosphatase; SHP1, SH2-domain-containing protein tyrosine phosphatase 1; SOCS, suppressor of
cytokine signalling; STAT1, signal transducer and activator of transcription 1; TRAF6, tumour-necrosis factor-receptor-associated factor 6.

294 | APRIL 2007 | VOLUME 7 www.nature.com/reviews/immunol


© 2007 Nature Publishing Group
REVIEWS

a Generation of osteoclast precursor b Early response to RANKL c Initial induction of NFATc1


cells by M-CSF

Unknown
ligand
M-CSF RANKL
Ig-like
cFMS RANK receptor

DAP12 Phosphorylation P
or FcRγ TRAF6 of ITAM P SYK PLCγ Ca2+
• Proliferation NFATc2
• Survival NFATc2 NFATc2
Co-stimulatory
signals NF-κB
Cytosol

Nucleus NFATc1

d Autoamplification of NFATc1 e Induction of osteoclast-specific genes

P P
SYK PLCγ Ca2+ SYK PLCγ Ca2+

cFOS CREB CaMKIV

Calcineurin
Calcineurin
AP1
Auto-amplification

MITF PU.1 Induction of


AP1 osteoclast-
NFATc1 NFATc1 CREB NFATc1 specific genes

Figure 2 | Spatiotemporal control of signal transduction during osteoclastogenesis. Osteoclast differentiation


is induced by M-CSF (macrophage colony-stimulating factor), RANKL (receptor activator of nuclear factor-κB
(NF-κB) ligand) and its co-stimulatory factor, immunoglobulin (Ig)-like receptor. a | Precursor-cell stage.
The binding of M-CSF to its receptor, cFMS, activates the proliferation and survival of osteoclast precursor cells
of the monocyte–macrophage lineage that express RANK. The co-stimulatory receptors might be stimulated from
early stages, although ligands of co-stimulatory receptors have yet to be identified. b | Proximal RANK signals.
RANKL binding to RANK results in the recruitment of TRAF6 (tumour-necrosis-factor-receptor-associated factor
6). At the same time, RANK activation results in the phosphorylation of the ITAM (immunoreceptor tyrosine-based
activation motif) in DAP12 and FcRγ (Fc-receptor common γ-subunit), both of which are adaptor proteins
associating with distinct Ig-like receptors. Ig-like receptor signals are called co-stimulatory signals for RANK.
c | Initial induction of NFATc1 (nuclear factor of activated T cells, cytoplasmic 1). NFATc1, a key transcription factor
for osteoclastogenesis, is initially induced by TRAF6-activated NF-κB and NFATc2 that is present in the cell before
RANKL stimulation. Phosphorylation of the ITAM in DAP12 (or FcRγ) results in the recruitment of spleen tyrosine
kinases (SYKs) that activate calcium signalling through phospholipase Cγ (PLCγ). d | Auto-amplification of NFATc1.
Calcium-signal-mediated persistent activation of NFATc1, as well as cooperation with activator protein 1 (AP1), is
a prerequisite for the robust induction of NFATc1. AP1 activation is mediated by the induction and activation of
cFOS by CaMKIV (calcium/calmodulin-dependent protein kinase type IV)-stimulated CREB (cyclic AMP
responsive-element-binding protein) and cFMS. The NFATc1 promoter is epigenetically activated through histone
acetylation and NFATc1 binds to an NFAT-binding site of its own promoter. e | In the nucleus, NFATc1 works
together with other transcription factors, such as AP1, PU.1, MITF (microphthalmia-associated transcription
factor) and CREB, to induce various osteoclast-specific genes, including TRAP (tartrate-resistant acid
phosphatase), cathepsin K and calcitonin receptor.

NATURE REVIEWS | IMMUNOLOGY VOLUME 7 | APRIL 2007 | 295


© 2007 Nature Publishing Group
REVIEWS

juvenile Paget’s disease of the bone, respectively37. T cells (MAPKs), including JUN N-terminal kinase (JNK)
not only express RANKL on their membrane, but also and p38 (REF. 52). The essential role of NF-κB in osteo-
release it as a soluble form, although the in vivo function clastogenesis has been shown genetically53,54, but the
of the soluble form remains to be clarified12,38. in vivo function of each MAPK remains to be elucidated.
The lysine-63-linked polyubiquitylation mediated by
Immunological functions of RANKL. Interestingly, the RING (really interesting new gene)-finger motif
RANKL–/– mice have a defect in the development of of TRAF6 has been shown to be important for NF-κB
secondary lymphoid organs36 (a lack of lymph nodes activation in other cell types55, but deletion analysis has
and abnormality in the spleen) and mammary glands39. indicated that the RING-finger domain of TRAF6 is
RANKL might regulate lymphotoxin expression in dispensable for the formation of osteoclasts52. Therefore,
lymph nodes, but the intact formation of Peyer’s the importance of the ubiquitin-ligase activity of TRAF6
patches in RANKL–/– mice indicates a more complicated in osteoclastogenesis is yet to be established.
mechanism that needs to be clarified12. Although RANK activation is linked to the phos-
T cells that express RANKL have a positive role in DC phorylation of DAP12 (also known as TYROBP and
activation and survival in vitro30,40,41, indicating that the KARAP) and Fc-receptor common γ-subunit (FcRγ),
RANKL–RANK system regulates DC function through as mentioned below, it is still not clear how RANK
the T cell–DC interaction. However, DC development alone among the TRAF6-binding receptors is able to
and function are normal in RANKL–/– mice12 and the stimulate osteoclastogenesis so strongly. Additional
function of RANKL in the eradication of pathogens RANK-specific adaptor molecule(s) might exist and
such as viruses has not been clearly observed in vivo42. enhance TRAF6 signalling50. For example, the molecular
The inhibition of RANKL greatly decreases antiviral scaffold GRB2-associated binding protein 2 (GAB2) has
responses in mice lacking CD40L, possibly because the been shown to be associated with RANK and to have
CD40L–CD40 system has a redundant role 42. On an important role in its signal transduction56. Signalling
the basis of this functional redundancy, the crucial role crosstalk between RANKL and M-CSF, another essential
of RANKL in the immune system might not be obvious cytokine for osteoclast differentiation, might be crucially
under physiological conditions. important for osteoclastogenesis10, but it is only partially
RANKL was shown to have a pathological role in a characterized (FIG. 3). Similar to RANK, Toll-like recep-
model of inflammatory bowel disease, indicating that tors (TLRs) also use TRAF6 as a crucial component of
RANKL stimulates DC activation in vivo and acceler- their signalling cascade57, but how such receptor signal-
ates immune reactions under certain autoimmune con- ling is able to bring about completely distinct outputs is
ditions43. On the other hand, RANKL is involved in the poorly understood.
generation of regulatory T cells in a diabetic model44. RANK also activates the transcription-factor
In addition, RANKL expressed by keratinocytes in complex, activator protein 1 (AP1), through induc-
the skin has a crucial role in the control of regulatory tion of its component cFOS58, which has recently been
T cells by modulating the function of Langerhans cells shown to be dependent on the activation of calcium/
and it suppresses autoimmune reactions induced by calmodulin-dependent protein kinase type IV (CaMKIV)
CD40L45. These reports clearly indicate that RANKL and cyclic-AMP-responsive-element-binding protein
has an immunosuppressive role in vivo. In addition, a (CREB)59. Importantly, RANKL specifically and strongly
recent report indicated that RANKL is a chemotactic induces nuclear factor of activated T cells, cytoplasmic 1
factor for RANK-expressing tumour cells as well as (NFATc1), the master regulator of osteoclast differen-
osteoclasts, indicating that RANKL also functions tiation60, and this induction is dependent on both the
as a chemokine46. Therefore, although the function of TRAF6–NF-κB and the cFOS pathways. The NFAT fam-
RANKL in the immune system needs to be elucidated ily of transcription factors was originally discovered in
in more detail, it is clear that RANKL is one of the T cells, but they are involved in the regulation of various
most important molecules that explicitly link the bone biological systems61,62. Activation of NFAT is mediated
and immune systems. by a specific phosphatase, calcineurin, which is activ-
ated by calcium–calmodulin signalling. The essential
Cellular response to RANKL. The intracellular signal- and sufficient role of the NFATc1 gene in osteoclas-
ling pathways of RANKL have been extensively studied togenesis has been shown both in vitro and in vivo60,63.
in the context of osteoclast differentiation11 (FIG. 2). The NFATc1 promoter contains NFAT binding sites and
RANK lacks intrinsic enzymatic activity in its intra- NFATc1 specifically autoregulates its own promoter dur-
cellular domain, and transduces signalling by recruiting ing osteoclastogenesis and enables the robust induction
adaptor molecules such as the TNF-receptor-associated of NFATc1 (REF. 63). AP1 (containing cFOS) and continu-
factor (TRAF) family of proteins47. Among the TRAF ous activation of calcium signalling is crucial for this
proteins, gene-disruption studies48,49 followed by inten- auto-amplification60,64. NFATc1 regulates a number of
sive molecular analyses identified TRAF6 as the main osteoclast-specific genes, such as cathepsin K, tartrate-
adaptor molecule that links RANK to the differentiation resistant acid phosphatase (TRAP), calcitonin receptor,
and function of osteoclasts50–52. By an as-yet-unknown osteoclast-associated receptor (OSCAR) and β3-integrin,
mechanism, RANKL binding to RANK induces the in cooperation with other transcription factors such as
trimerization of RANK and TRAF6, which leads to AP1, PU.1 and MITF60,65–67. A recent study indicated
the activation of NF-κB and mitogen-activated kinases that CREB activated by CaMKIV also cooperates with

296 | APRIL 2007 | VOLUME 7 www.nature.com/reviews/immunol


© 2007 Nature Publishing Group
REVIEWS

NFATc1 in the activation of osteoclast-specific genes59. Interplay with other cytokine systems. Osteoclasts are
Compared with the wealth of information on the role derived from the haematopoietic lineage and are there-
of RANKL in osteoclasts, it remains unclear whether fore naturally influenced by cytokines that direct haema-
RANKL uses the same signalling mechanism when it topoietic-cell differentiation. Crosstalk among cytokines
exerts its functions in the immune system40,41,44. is a target of much interest for osteoimmunology, and

Osteoblast

RANKL Unknown
M-CSF
OPG Ig-like
RANK receptor cFMS

Osteoclast TRAF6 DAP12 or FcRγ


CD40L
IL-1 SYK PI3K GRB2 VAV3
cSRC
IFNγ cCBL
IKKs MAPKs PLCγ AKT ERK

Ca2+

IFNβ cFOS CREB CaMKIV Calcineurin Proliferation


and survival

TNF
NF-κB AP1 Cytoskeletal
reorganization
LPS ?

NFATc1 Auto-amplification Bone


• DC-STAMP resorption
AP1 Fusion
PU.1 • ATP6V0D2
NFATc1
MITF
CREB
• Cathepsin K
• MMP9
• ATP6I
Osteoclast • ClC7
differentiation
Figure 3 | Osteoimmunological interactions in osteoclasts and osteoblasts. The immune and skeletal systems
share cytokines, receptors, signalling molecules and transcription factors, all of which cooperatively regulate
osteoclasts and osteoblasts as well as their interactions. Osteoblasts regulate osteoclastogenesis through
receptor activator of nuclear factor-κB (NF-κB) ligand (RANKL)–RANK (and its decoy receptor osteoprotegerin
(OPG)) interactions, macrophage colony-stimulating factor (M-CSF)–cFMS interactions and immunoglobulin
(Ig)-like receptors associated with immunoreceptor tyrosine-based activation motif (ITAM)-harbouring adaptor
molecules (such as DAP12 and Fc-receptor common γ-subunit (FcRγ), the ligands of which are not well
characterized). Although not depicted, semaphorin 6D and its receptor plexin A1, and ephrin receptor B4 and
ephrin B2 were newly identified as mediators of osteoblast–osteoclast interactions. There are extensive signalling
pathways in osteoclasts. RANK and Ig-like receptors stimulate downstream signalling cascades (such as tumour-
necrosis factor (TNF) receptor-associated factor 6 (TRAF6), NF-κB, mitogen-activated protein kinases (MAPKs),
activator protein 1 (AP1), calcineurin and nuclear factor of activated T cells, cytoplasmic 1 (NFATc1)), which are
influenced by a number of immunoregulatory molecules including CD40 ligand (CD40L), interleukin-1 (IL-1),
interferon-β (IFNβ), IFNγ, TNF and lipopolysaccharide (LPS). Dendritic-cell-specific transmembrane protein
(DC-STAMP) and ATP6V0D2 are necessary for the fusion of osteoclast precursor cells. Phosphoinositide 3-kinase
(PI3K)–AKT and GRB2–ERK (growth-factor-receptor-bound protein 2–extracellular-signal-regulated kinase)
pathways are important for the proliferation and survival of the osteoclast lineage, whereas VAV3, cSRC and
Casitas B-lineage lymphoma (cCBL) are included in the molecules required for cytoskeletal reorganization and
bone-resorbing osteoclasts. Osteoclast activity is dependent on acidifying proton pump ATP6I and chloride
channel ClC7, as well as matrix-degrading enzymes such as cathepsin K and matrix metalloproteinase 9 (MMP9).
The following molecules are known to be involved in both the bone system and the immune system: NF-κB,
RANKL, RANK, OPG, cFMS, M-CSF, Ig-like receptors, FcRγ, DAP12, TRAF6, MAPKs, AP1, calcineurin, NFATc1,
CD40L, IL-1, IFNγ, IFNβ, TNF, LPS, DC-STAMP, PI3K, AKT, ERK, VAV3, cSRC and cCBL. CaMKIV, calcium/
calmodulin-dependent protein kinase type IV; CREB, cyclic AMP responsive-element-binding protein;
PLC, phospholipase C; MITF, microphthalmia-associated transcription factor; IKK, inhibitor of NF-κB (IκB) kinase;
SYK, spleen tyrosine kinase.

NATURE REVIEWS | IMMUNOLOGY VOLUME 7 | APRIL 2007 | 297


© 2007 Nature Publishing Group
REVIEWS

is of great clinical relevance. Inflammatory cytokines of NF-κB during osteoclastogenesis63. Signals induced
that are mainly produced by macrophages, such as IL-1, by the pro-inflammatory cytokines probably converge at
TNF and IL-6, promote osteoclastogenesis, and are also the activation of NF-κB, which therefore has an integral
called osteolytic cytokines on the basis of their bone- role in osteoimmunological interactions. The function
resorbing effect in vivo24,25. IL-1 stimulates TRAF6 (and of classical and alternative NF-κB signalling pathways
therefore activates NF-κB and MAPKs) and synergizes has been studied using mice deficient in inhibitor of
with RANKL to induce mature osteoclasts to carry out NF-κB (IκB) kinases (IKKs)75 or NF-κB-inducing kinase
bone-resorbing activity, but, interestingly, IL-1 alone (NIK)76, but further studies will be needed to obtain a
cannot induce differentiation, indicating that TRAF6 complete understanding of their specific functions in
activation is not sufficient. IL-1 also indirectly facilitates osteoimmunology.
osteoclastogenesis by acting on the osteoblasts through As mentioned above, the AP1 component cFOS is
the induction of PGE2, and induces the expression of essential for the RANKL-mediated induction of NFATc1.
RANKL25. TNF stimulates the activation of NF-κB The AP1 transcription factor is a dimeric complex
mainly through TRAF2. Although TNF alone cannot composed of the FOS-family proteins (cFOS, FOSB,
induce osteoclastogenesis in vivo, nor can TNF over- FOS-related antigen 1 (FRA1) and FRA2), JUN-family
expression rescue the deficiency of RANKL68,69, it is proteins (cJUN, JUNB and JUND) and activating trans-
reported that TNF with transforming growth factor-β cription factor (ATF)-family proteins58. The specific and
(TGFβ) induces in vitro osteoclastogenesis even in the indispensable functions of AP1 transcription factors in
absence of RANK or TRAF6 (REF. 70). These results the regulation of osteoclasts and osteoblasts (for exam-
indicate that TNF has a pivotal role in the pathologi- ple, the role of cFOS and FRA1 in bone resorption and
cal activation of osteoclasts that are associated with of FRA1 and ATF4 in bone formation) have been well
inflammation. established by a series of studies from genetically modi-
Type I IFNs (that is, IFNα and IFNβ) are essential fied mice58, but emerging evidence indicates that they
for host defence against pathogens such as viruses. also have an unexpectedly interesting role in the immune
The induction mechanism of type I IFNs in response to system (for example, the role of FOS in lipopolysaccha-
pathogen-associated molecular patterns has been well ride signalling)77,78, which indicates the importance of
documented71, and recent studies on TLR signalling AP1 in the osteoimmune system. Although the precise
have provided new insights into the mechanism and the composition of AP1 dimers is not well characterized in
function of the IFN system57. Similar to TLRs, RANKL the physiological context, a recent study showed that
induces the IFNB gene in osteoclast precursor cells, and an AP1 dimer composed of FOS and any JUN protein
IFNβ functions as a negative-feedback regulator that induces osteoclastogenesis, but that FRA1 has different
inhibits the differentiation of osteoclasts by interfer- partners79.
ing with the RANKL-induced expression of cFOS72. The NFAT transcription-factor family was originally
The importance of type I IFNs in bone homeostasis was identified in T cells, and comprises five known members:
underscored by the observation that mice deficient in the NFATc1 (also known as NFAT2), NFATc2 (also known
type-I-IFN-receptor component IFNAR1 spontaneously as NFAT1), NFATc3 (also known as NFAT4), NFATc4
develop marked osteopaenia, accompanied by enhanced (also known as NFAT3) and NFAT5 (REFS 61,62). This
osteoclastogenesis72. family of transcription factors developed along with the
As T-cell activation is often associated with evolution of vertebrates, and is involved in the regulation
abnormal bone resorption, the regulation of bone of various biological systems, such as the cardiovascular
metabolism by T cells is one of the central subjects of and muscular systems, in addition to the bone system
osteoimmunology 8,14,73. T cells express RANKL, but the and the immune system. As the activation of NFATc1–
main T-cell cytokines such as IFNγ, IL-4 and IL-10 are NFATc4 is mediated by the phosphatase calcineurin,
all inhibitory to in vitro osteoclastogenesis, and IL-12 which is activated by calcium–calmodulin signalling,
and IL-18 also negatively affect osteoclastogenesis6,8, calcineurin inhibitors such as FK506 and cyclosporin A
indicating a complex mechanism working in vivo, strongly inhibit osteoclastogenesis60. The necessary and
which will be discussed below. sufficient role of the NFATc1 gene in osteoclastogen-
esis was established using a genetic approach, and the
Immune mediators in osteoimmunology robust induction of NFATc1 by an auto-amplification
Transcription factors. NF-κB is a family of dimeric mechanism is dependent on the epigenetically deter-
trans cription factors including REL (cREL), RELA mined chromatin structure of the NFATc1 and NFATc2
(p65), RELB, NF-κB1 (p50) and NF-κB2 (p52, which promoters63. It is notable that NFATc1 has an exclusive
is processed from its precursor, p100)74. p50 and p52 role in osteoclasts but that NFATc1 and NFATc2 have
function by heterodimerization with any of the three a redundant role in T cells80. The importance of NFAT
REL proteins containing transcriptional activation in the regulation of bone homeostasis is also supported
domains. Mice deficient in both p50 and p52 develop by the recent findings that NFAT and calcineurin regu-
osteopetrosis owing to a defect in osteoclast differentia- late osteoblast differentiation and bone formation81–83.
tion, which indicates a crucial (but possibly overlapping) It has been shown that NFAT regulates bone formation
Osteopaenia
function of p50 and p52 in osteoclastogenesis53,54. NF-κB through interaction with OSTERIX81, but further stud-
A decrease in bone mineral is required for NFATc1 induction, but it is not clear ies are needed to clarify what activates calcineurin and
density. whether NFATc1 is the exclusive transcriptional target NFAT in osteoblasts.

298 | APRIL 2007 | VOLUME 7 www.nature.com/reviews/immunol


© 2007 Nature Publishing Group
REVIEWS

Inhibitor of DNA binding 2 (ID2) is a helix–loop– receptor (OSCAR; an immunoglobulin-like receptor)92,


helix protein, which often functions as a negative DC-specific transmembrane protein (DC-STAMP;
regulator of helix–loop–helix transcription factors. Id2- a seven-transmembrane-domain-containing receptor
deficient mice have no lymph nodes or Peyer’s patches84, that mediates cell–cell fusion of osteoclasts)93,94 and
and have a defect in mammary-gland formation as well B7-H3 (a member of the B7 superfamily; also known as
as a defective development of natural killer cells85. In this CD276)95, in addition to those involved in axon guid-
respect, Id2–/– mice resemble RANKL–/– mice in many ance, including the ephrin96 and semaphorin receptors97.
ways, but, interestingly, osteoclastogenesis is enhanced In vitro studies also have shown that osteoclast-inhibitory
in Id2–/– cells86. ID2 therefore functions downstream of lectin (OCIL) and 4-1BB (also known as CD137) might
RANKL, but, depending on the context, it can be both be involved in the regulation of osteoclastogenesis. These
a positive or negative regulator of RANKL signalling; findings indicate that osteoclast differentiation is regu-
this indicates a unique osteoimmunological function for lated by additional receptor systems that cooperate with
this molecule. RANK and cFMS, and many of them are involved in the
Signal transducer and activator of transcription 1 interaction between the osteoblast and osteoclast line-
(STAT1) has a crucial role in the immune system as a ages. Therefore, an increasing number of ligand–receptor
mediator of signal transduction and a regulator of gene systems originally identified in the immune system have
transcription in both type I and II IFN systems87. In the also been found to be functional in the skeletal system
skeletal system, both types of IFN inhibit osteoclasto- (FIG. 3; TABLE 1; Supplementary information S1 (table)).
genesis through STAT1, but bone mass was increased FcRγ and DAP12 are adaptor molecules that associate
in STAT1-deficient mice, despite increased osteoclasto- with immunoglobulin-like receptors, classically helping
genesis. Subsequently, STAT1 was found to have a these receptors to express themselves on the membrane,
unique, non-canonical function as a cytoplasmic attenu- and transduce signals through the immunoreceptor
ator of RUNX2, a key transcription factor for osteoblast tyrosine-based activation motif (ITAM) in natural
differentiation88. Therefore, the loss of STAT1 results in killer and myeloid cells98. An osteopetrotic phenotype
excessive RUNX2 activation and osteoblast differen- observed in mice deficient in both FcRγ and DAP12 led
tiation, thereby tipping the balance in favour of bone to the designation of immunoglobulin-like receptors as
formation over bone resorption. This is an interesting crucial co-stimulatory molecules for RANK99,100. The
example of an immunologically latent transcription fac- immunoglobulin-like receptors identified in osteoclast-
tor having a distinct function in the skeletal system. As precursor cells include OSCAR, triggering receptor
expected, the suppressors of cytokine signalling (SOCS) expressed in myeloid cells 2 (TREM2), signal-regulatory
proteins, which are crucial regulators of IFN and STAT protein β1 (SIRPβ1) and paired immunoglobulin-like
signal transduction, are also involved in the diverse receptor A (PIRA), although the ligands and function of
regulatory mechanisms of osteoimmunology 89,90. each of these receptors remain to be elucidated. ITAM-
Schnurri-3 (SHN3), a zinc-finger protein, was first mediated signals cooperate with RANK to stimulate
identified as a DNA-binding protein involved in the calcium signalling through ITAM phosphorylation and
regulation of V(D)J recombination of immunoglobulin the resulting activation of spleen tyrosine kinase (SYK)
genes. However, SHN3 has been shown to have an and phospholipase Cγ (PLCγ)101. As this pathway is
important role in the control of bone formation through crucial for the robust induction of NFATc1 that leads
proteolytic degradation of RUNX2, to which the activ- to osteoclastogenesis, but because ITAM signals alone
ity of WWP1 (WW domain-containing E3 ubiquitin cannot induce osteoclastogenesis, these signals should
protein ligase 1) might also contribute91. be called co-stimulatory signals for RANK5,99.
Therefore, accumulating evidence indicates that a The role of the immunoreceptor tyrosine-based
great number of transcriptional regulators are shared inhibitory motif (ITIM) in osteoclast differentia-
by the immune and the bone systems, and they con- tion remains to be elucidated, but osteoclastogenesis
trol not only osteoclasts but also osteoblasts (FIG. 3). is enhanced in mice lacking phosphatases such as
Osteoblast-mediated bone formation is also affected SH2-domain-containing protein tyrosine phosphatase 1
by soluble factors such as cytokines in the immune sys- (SHP1)102,103 or SH2-domain-containing inositol-5-
tem, including TNF, IL-1 and IL-4 (REF. 6); osteoblasts phosphatase 1 (SHIP1)104, which counterbalance the
are therefore influenced by the immune cells, although ITAM signal in the immune system. The importance
the physiological and pathological significance and the of the ITAM-harbouring adaptors and the recep-
molecular mechanisms are less well understood than in tors associated with them in bone metabolism is also
osteoclasts. underscored by reports that mutations in the DAP12
and TREM2 genes cause skeletal and psychotic abnor-
Immunoreceptors and co-stimulation. RANK and malities that are collectively known as Nasu–Hakola
Nasu–Hakola disease cFMS were thought to be necessary and sufficient for disease98. The expression of semaphorins, which are
A rare autosomal-recessive osteoclastogenesis24, but this was only based on the axon-guidance molecules and immunoregulators, has
disease characterized by observation that the addition of RANKL and M-CSF been observed in bone cells, but it has recently been
systemic bone cysts and is sufficient to induce osteoclast formation in a culture shown that semaphorin 6D and its receptor plexin-A1
psychotic symptoms similar to
dementia or schizophrenia
system. Genome-wide screening and gene-disruption might have an important role in the activation of the
(mutations in DAP12 or studies have shed light on hitherto-unknown functions DAP12-mediated ITAM signal through the interactions
TREM2 genes are reported). of immunological receptors, such as osteoclast-associated among plexin-A1, DAP12 and TREM2 (REF. 97).

NATURE REVIEWS | IMMUNOLOGY VOLUME 7 | APRIL 2007 | 299


© 2007 Nature Publishing Group
REVIEWS

Table 2 | Cytokines functioning in the regulation of osteoclast differentiation by T cells


Cytokine Effects on Main producer cells Main target cells in the Functions in the context of osteoimmunology
osteoclastogenesis regulation of osteoclasts
IFNγ Inhibition TH1 cells and NK cells Osteoclast precursor cells Cellular immunity; RANKL signalling inhibition
IL-4 Inhibition TH2 cells and NKT cells Osteoclast precursor cells Humoral immunity; RANKL signalling inhibition
IL-6 Activation TH2 cells and DCs Mesenchymal cells and Inflammatory; RANKL induction on mesenchymal
T cells cells; TH17-cell differentiation
IL-10 Inhibition TH2 cells Osteoclast precursor cells Anti-inflammatory; RANKL signalling inhibition
GM-CSF Inhibition TH1 cells Osteoclast precursor cells Granulocyte differentiation; RANKL signalling
inhibition
IL-12 Inhibition Macrophages and DCs T cells TH1-cell differentiation; IFNγ and GM-CSF
induction
IL-17 Activation TH17 cells and memory Mesenchymal cells Inflammatory; RANKL induction on mesenchymal
T cells cells
IL-18 Inhibition Macrophages and DCs T cells TH1-cell differentiation, IFNγ induction
RANKL Activation T cells and Osteoclast precursor cells Osteoclast differentiation induction
mesenchymal cells
(osteoblasts)
TNF Activation Macrophages and TH1 Osteoclast precursor cells Inflammatory; RANKL induction on mesenchymal
cells and mesenchymal cells cells; synergy with RANKL
DC, dendritic cell; GM-CSF, granulocyte macrophage colony-stimulating factor; IFNγ, interferon-γ; IL, interleukin; NK, natural killer; NKT, natural killer T; RANKL,
receptor activator of nuclear factor-κB ligand; TH, T helper; TNF, tumour-necrosis factor.

Cellular interactions in the osteoimmune system It is worthwhile to define what we believe to be a very
Lymphocyte regulation of osteoclasts. Activation of the rare but pathologically important TH-cell subset respon-
immune system is essential for host defence against sible for abnormal bone resorption as osteoclastogenic
pathogens, but aberrant and/or prolonged activation TH cells. Previous investigations in our laboratory and
under certain pathological conditions results in tis- other studies on synovial T cells in rheumatoid arthritis
sue damage owing to the activation of effector cells. clarified the characteristics of osteoclastogenic TH cells
In autoimmune arthritis, it has long been a challeng- in autoimmune arthritis 8,9. First, osteoclastogenic
ing question as to how the abnormal T-cell activa- TH cells do not produce a large amount of IFNγ. Second,
tion (characterized by infiltration of CD4+ T cells) they trigger local inflammation and the production of
mechanistically induces bone damage8,9. Researchers inflammatory cytokines, including TNF, that induce
had already observed osteoclast-like cells at the bone- RANKL expression on synovial fibroblasts. Third, osteo-
destruction site in the early 1980s (REF. 105), but the clastogenic TH cells express RANKL and might directly
importance of osteoclasts has just recently come into participate in accelerated osteoclastogenesis. Because
general acceptance because of the therapeutic efficacy these TH cells have such osteoclastogenic characteristics,
of osteoclast-targeted therapies14,106 and the protec- they can tip the balance in favour of osteoclastogenesis
tion of osteoclast-deficient mice from inflammatory in various aspects. Although autoimmune arthritis has
bone loss107,108. It remains unclear whether osteoclasts been traditionally categorized as a TH1-type disease,
under physiological and inflammatory conditions have TH1 cells do not have such characteristics, indicating that
distinct characteristics, but osteoclasts are partly resist- the osteoclastogenic TH cells might belong to an as-yet
ant to apoptosis induced by bisphosphonates in the unknown subset.
presence of inflammatory cytokines. Recent data from our laboratory indicate that an
As RANKL is expressed in activated T cells, T cells IL-17-producing TH-cell subset (TH17 cells) represents
might have the capacity to induce osteoclast differentia- the long-sought-after osteoclastogenic TH-cell subset,
tion by directly acting on osteoclast-precursor cells14,109. fulfilling all the criteria mentioned above111. Therefore,
However, the IFNγ produced by T cells potently sup- the infiltration of TH17 cells into the inflammatory lesion
presses RANKL signalling through downregulation of links the abnormal T-cell response to bone damage
TRAF6 (REF. 73). T cells also secrete various cytokines (FIG. 4), and the pathogenesis of autoimmune arthritis
such as IL-4 and IL-17 (REF. 110) (TABLE 2), so the effects should be reconsidered in the context of a TH17-type
of T cells on osteoclastogenesis should be dependent disease. Clearly, this subset will be an auspicious target of
on the balance between positive and negative factors future therapy, and cytokines related to TH17-cell differ-
expressed by the T cells. As the well-known effector entiation and expansion, such as IL-6, TGFβ and IL-23,
CD4+ T helper (TH)-cell subsets TH1 and TH2 produce will be of great clinical importance.
IFNγ and IL-4, respectively, both of which are anti- Compared with the established pathological
osteoclastogenic, it has been a paradox that activated role of T cells in arthritis, the role of T cells in non-
CD4+ T cells in arthritis enhance osteoclastogenesis in inflammatory metabolic bone diseases such as postmeno-
the presence of these cytokines. pausal osteoporosis has recently been proposed112 but is

300 | APRIL 2007 | VOLUME 7 www.nature.com/reviews/immunol


© 2007 Nature Publishing Group
REVIEWS

Rheumatoid joint Normal joint


Inflamed
synovium
Bone

Osteoclastogenesis
and bone destruction
Synovial Osteoclast
membrane

Cartilage
Osteoclast
precursor cell
Synovial
macrophage
Inflammatory
Blood cytokines such
vessel as TNF, IL-1 and IL-6

IL-17
Increased
RANKL
Synovial fibroblast
Synovial
T cell
(TH17 cell)

Figure 4 | How T cells induce osteoclastogenesis in autoimmune arthritis. In rheumatoid arthritis, inflammatory
synovium invades and destroys bone; this is mediated by osteoclasts that are induced by receptor activator of nuclear
factor-κB ligand (RANKL). Cells in the synovium include synovial macrophages and fibroblasts in addition to the dendritic
cells, plasma cells, endothelial cells and infiltrating T cells. Although CD4+ T-cell infiltration is a hallmark of the
pathogenesis of arthritis, the link between T cells and the activation of osteoclast-mediated bone resorption has only
recently been shown: interleukin-17 (IL-17)-producing T helper cells (TH17 cells) are the only osteoclastogenic TH-cell
subset characterized so far. TH17 cells do not produce interferon-γ (IFNγ), which suppresses RANKL signalling, but secrete
a huge amount of IL-17 that induces RANKL on synovial fibroblasts. IL-17 also stimulates the local inflammation and
activates synovial macrophages to secrete pro-inflammatory cytokines such as tumour-necrosis factor (TNF), IL-1 and IL-6.
These cytokines activate osteoclastogenesis by either directly acting on osteoclast precursor cells or inducing RANKL on
synovial fibroblasts. TH17 cells also express RANKL on their membrane, which partly contributes to the enhanced
osteoclastogenesis.

not yet recognized by immunologists. The possibil- between osteoblasts and HSCs is mediated by molecular
ity that oestrogen regulates T-cell immune responses interactions, such as N-cadherin–β-catenin, Jagged–
and that T-cell-mediated autoimmunity is involved Notch1 and angiopoietin-1–tyrosine kinase receptor 2
in postmenopausal osteoporosis is intriguing, and interactions15,16,18. It is possible that the osteoblast and
further points to the notion of an intricate interplay niche functions are under the control of bi-potential
between the immune and skeletal systems, but this regulators such as WNT 116. The CXCL12–CXCR4
concept is still highly controversial113 and will not be (CXC-chemokine ligand 12 (also known as SDF1)–
proved until the autoantigen that causes osteoporosis CXC-chemokine receptor 4) system is also involved in
is identified114. the colonization of bone marrow by HSCs and in their
retention in the bone marrow, but the localization of
Osteoblasts as a stem-cell niche. Osteoimmunology CXCL12-expressing cells is not consistent with that
is in the process of extending beyond the function of of osteoblasts on the trabecular bone surface117.
immune mediators in bone cells — increasing atten- Therefore, the detailed localization and characteriza-
tion is currently being given to the regulation of HSCs tion of cells representing the stem-cell niche in the bone
by bone cells. The role of osteoblasts in haematopoiesis marrow remain to be clarified in the future. As it is
was proposed in a previous study, in which impaired unclear whether the bone-forming capacity is related
haematopoiesis was observed after the chemical abla- to the function required for the niche, it is necessary to
Trabecular bone tion of osteoblasts115. HSCs interact with specialized carefully reconsider the definition of osteoblasts. More
A spongy and low-density type microenvironments, known as stem-cell niches, to recently, osteoclasts have also been indicated to be
of osseous tissue with high
surface area, which fills up the
maintain the ability of self-renewal and pluripotency. involved in the mobilization of HSCs17, further support-
inner cavity of long bones and Osteoblasts on the trabecular bone surface have emerged ing the intimate relationship between the haematopoietic
vertebrae. as a crucial component of the niche, and the interaction and skeletal systems.

NATURE REVIEWS | IMMUNOLOGY VOLUME 7 | APRIL 2007 | 301


© 2007 Nature Publishing Group
REVIEWS

Clinical implications osteoclastogenesis by suppressing the induction of


A detailed osteoimmunological understanding of the NFATc1 (REF. 121). Therefore, the osteoimmunological
pathogenesis of bone destruction in many contexts perspective has enabled us to gain profound insights into
will lead to novel strategies for the treatment of various the understanding of the mode of action of therapeutic
diseases, including rheumatoid arthritis, periodontal dis- drugs as well as a novel strategy for drug design.
ease, Paget’s disease, osteoporosis, osteoarthritis, multiple
myeloma and metastatic bone tumours. Rheumatologists Concluding remarks and future perspectives
are now aware of the remarkable impact of TNF-specific The bone system and the immune system share an abun-
antibody therapy on the treatment of rheumatoid arthri- dance of molecules and regulatory mechanisms, and this,
tis, and other cytokines will soon be targeted by similar together with the importance of the interactions between
strategies using biological agents19. The action of TNF the two systems might have resulted in the emergence
and other inflammatory cytokines is not limited to the of the field of osteoimmunology, but more attention
induction of local inflammation, but is directly and indi- should be paid to the differences in the future: how are
rectly involved in the activation of osteoclasts, as men- distinct functions achieved through similar mechanisms?
tioned above. Such osteoimmunological pleiotropy might Similarly, genetically modified mice deficient in a mol-
explain the dramatic efficacy of the biological agent to ecule originally characterized in one system should be
prevent or even ameliorate bone destruction. In addition, analysed in another system to clarify the functions in the
the efficacy of a RANKL-specific antibody for postmeno- context of the osteoimmune system. This will surely lead
pausal osteoporosis and rheumatoid arthritis in clinical to an understanding of the molecular basis for the cell-
trials has been reported118,119, and its apparent preventive lineage specification and, perhaps more importantly,
effect on bone metastasis is enormously promising 46. cell-type-specific treatment despite the similarity of
Interestingly, severe adverse effects of RANKL inhibi- the two systems. The novel functions of bone cells in the
tion on the immune system have never been reported. regulation of cells outside the bone will have an impact
The clinical relevance of newly recognized cytokines on many genres of biomedical studies. For example, the
such as IL-17 and IL-23 has yet to be established in crucial function of bone cells in the control of HSCs might
humans, but IL-6 inhibition, which is also under provide new methods to modulate the HSC mobilization
clinical trial with successful results, might have a dual through bone cells. Therefore, there is reason to believe
impact on TH17-mediated immune responses and that both clinical and non-clinical researchers will benefit
bone120. Although it is difficult to specifically target from the emerging insights that are leading to an inte-
transcription factors and signalling molecules in drug grated understanding of the osteoimmune system and its
treatment, some of the anti-rheumatic drugs inhibit complex interactions.

1. Horton, J. E., Raisz, L. G., Simmons, H. A., 13. Yoshida, H. et al. The murine mutation osteopetrosis 24. Boyle, W. J., Simonet, W. S. & Lacey, D. L. Osteoclast
Oppenheim, J. J. & Mergenhagen, S. E. is in the coding region of the macrophage colony differentiation and activation. Nature 423, 337–342
Bone resorbing activity in supernatant fluid stimulating factor gene. Nature 345, 442–444 (2003).
from cultured human peripheral blood leukocytes. (1990). 25. Suda, T. et al. Modulation of osteoclast differentiation
Science 177, 793–795 (1972). 14. Kong, Y. Y. et al. Activated T cells regulate bone loss and function by the new members of the tumor
2. Mundy, G. R., Raisz, L. G., Cooper, R. A., and joint destruction in adjuvant arthritis through necrosis factor receptor and ligand families.
Schechter, G. P. & Salmon, S. E. Evidence for the osteoprotegerin ligand. Nature 402, 304–309 Endocr. Rev. 20, 345–357 (1999).
secretion of an osteoclast stimulating factor in (1999). 26. Teitelbaum, S. L. & Ross, F. P. Genetic regulation of
myeloma. N. Engl. J. Med. 291, 1041–1046 (1974). 15. Arai, F. et al. Tie2/angiopoietin-1 signaling regulates osteoclast development and function. Nature Rev.
3. Dewhirst, F. E., Stashenko, P. P., Mole, J. E. & hematopoietic stem cell quiescence in the bone Genet. 4, 638–649 (2003).
Tsurumachi, T. Purification and partial sequence marrow niche. Cell 118, 149–161 (2004). 27. Tondravi, M. M. et al. Osteopetrosis in mice
of human osteoclast-activating factor: identity with 16. Calvi, L. M. et al. Osteoblastic cells regulate lacking haematopoietic transcription factor PU.1.
interleukin 1 β. J. Immunol. 135, 2562–2568 (1985). the haematopoietic stem cell niche. Nature Nature 386, 81–84 (1997).
4. Horowitz, M., Vignery, A., Gershon, R. K. & Baron, R. 425, 841–846 (2003). 28. McGill, G. G. et al. Bcl2 regulation by the melanocyte
Thymus-derived lymphocytes and their interactions This is one of the initial studies showing that master regulator Mitf modulates lineage survival
with macrophages are required for the production of osteoblasts function as a haematopoietic and melanoma cell viability. Cell 109, 707–718.
osteoclast-activating factor in the mouse. Proc. Natl stem-cell niche. (2002).
Acad. Sci. USA 81, 2181–2185 (1984). 17. Kollet, O. et al. Osteoclasts degrade endosteal 29. Lagasse, E. & Weissman, I. L. Enforced expression
5. Takayanagi, H. Mechanistic insight into osteoclast components and promote mobilization of of Bcl-2 in monocytes rescues macrophages and
differentiation in osteoimmunology. J. Mol. Med. hematopoietic progenitor cells. Nature Med. partially reverses osteopetrosis in op/op mice.
83, 170–179 (2005). 12, 657–664 (2006). Cell 89, 1021–1031 (1997).
6. Walsh, M. C. et al. Osteoimmunology: interplay 18. Zhang, J. et al. Identification of the haematopoietic This is one of the important studies that
between the immune system and bone metabolism. stem cell niche and control of the niche size. determines the role of M-CSF in osteoclasts.
Annu. Rev. Immunol. 24, 33–63 (2006). Nature 425, 836–841 (2003). 30. Anderson, D. M. et al. A homologue of the TNF
7. Arron, J. R. & Choi, Y. Bone versus immune system. 19. Palladino, M. A., Bahjat, F. R., Theodorakis, E. A. & receptor and its ligand enhance T-cell growth and
Nature 408, 535–536 (2000). Moldawer, L. L. Anti-TNF-α therapies: the next dendritic-cell function. Nature 390, 175–179
8. Sato, K. & Takayanagi, H. Osteoclasts, rheumatoid generation. Nature Rev. Drug Discov. 2, 736–746 (1997).
arthritis, and osteoimmunology. Curr. Opin. (2003). 31. Lacey, D. L. et al. Osteoprotegerin ligand is a
Rheumatol. 18, 419–426 (2006). 20. Seeman, E. & Delmas, P. D. Bone quality—the cytokine that regulates osteoclast differentiation
9. Takayanagi, H. Inflammatory bone destruction and material and structural basis of bone strength and and activation. Cell 93, 165–176 (1998).
osteoimmunology. J. Periodontal Res. 40, 287–293 fragility. N. Engl. J. Med. 354, 2250–2261 (2006). 32. Wong, B. R. et al. TRANCE is a novel ligand of the
(2005). 21. Harada, S. & Rodan, G. A. Control of osteoblast tumor necrosis factor receptor family that activates
10. Ross, F. P. & Teitelbaum, S. L. αvβ3 and macrophage function and regulation of bone mass. Nature c-Jun N-terminal kinase in T cells. J. Biol. Chem.
colony-stimulating factor: partners in osteoclast 423, 349–355 (2003). 272, 25190–25194 (1997).
biology. Immunol. Rev. 208, 88–105 (2005). 22. Karsenty, G. & Wagner, E. F. Reaching a genetic and 33. Yasuda, H. et al. Osteoclast differentiation factor
11. Asagiri, M. & Takayanagi, H. The molecular molecular understanding of skeletal development. is a ligand for osteoprotegerin/osteoclastogenesis-
understanding of osteoclast differentiation. Dev. Cell 2, 389–406 (2002). inhibitory factor and is identical to TRANCE/RANKL.
Bone 40, 251–264 (2007). 23. Nakashima, K. et al. The novel zinc finger-containing Proc. Natl Acad. Sci. USA 95, 3597–3602 (1998).
12. Theill, L. E., Boyle, W. J. & Penninger, J. M. transcription factor osterix is required for osteoblast 34. Simonet, W. S. et al. Osteoprotegerin: a novel
RANK-L and RANK: T cells, bone loss, and mammalian differentiation and bone formation. Cell 108, 17–29 secreted protein involved in the regulation of bone
evolution. Annu. Rev. Immunol. 20, 795–823 (2002). (2002). density. Cell 89, 309–319 (1997).

302 | APRIL 2007 | VOLUME 7 www.nature.com/reviews/immunol


© 2007 Nature Publishing Group
REVIEWS

35. Tsuda, E. et al. Isolation of a novel cytokine 59. Sato, K. et al. Regulation of osteoclast differentiation 84. Yokota, Y. et al. Development of peripheral lymphoid
from human fibroblasts that specifically inhibits and function by the CaMK–CREB pathway. Nature organs and natural killer cells depends on the helix-
osteoclastogenesis. Biochem. Biophys. Res. Med. 12, 1410–1416 (2006). loop-helix inhibitor Id2. Nature 397, 702–706
Commun. 234, 137–142 (1997). 60. Takayanagi, H. et al. Induction and activation of the (1999).
36. Kong, Y. Y. et al. OPGL is a key regulator of transcription factor NFATc1 (NFAT2) integrate RANKL 85. Kim, N. S. et al. Receptor activator of NF-κB
osteoclastogenesis, lymphocyte development and signaling for terminal differentiation of osteoclasts. ligand regulates the proliferation of mammary
lymph-node organogenesis. Nature 397, 315–323 Dev. Cell 3, 889–901 (2002). epithelial cells via Id2. Mol. Cell. Biol. 26, 1002–1013
(1999). 61. Crabtree, G. R. & Olson, E. N. NFAT signaling: (2006).
This study provides the first genetic evidence for choreographing the social lives of cells. 86. Lee, J. et al. Id helix-loop-helix proteins negatively
the crucial role of RANKL in both immune and Cell 109, S67–S79 (2002). regulate TRANCE-mediated osteoclast differentiation.
bone systems. 62. Hogan, P. G., Chen, L., Nardone, J. & Rao, A. Blood 107, 2686–2693 (2006).
37. Whyte, M. P. & Mumm, S. Heritable disorders Transcriptional regulation by calcium, calcineurin, 87. Takayanagi, H., Kim, S., Koga, T. & Taniguchi, T.
of the RANKL/OPG/RANK signaling pathway. and NFAT. Genes Dev. 17, 2205–2232 (2003). Stat1-mediated cytoplasmic attenuation in
J. Musculoskelet. Neuronal. Interact. 4, 254–267 63. Asagiri, M. et al. Autoamplification of NFATc1 osteoimmunology. J. Cell. Biochem. 94, 232–240
(2004). expression determines its essential role in bone (2005).
38. Hikita, A. et al. Negative regulation of homeostasis. J. Exp. Med. 202, 1261–1269 (2005). 88. Kim, S. et al. Stat1 functions as a cytoplasmic
osteoclastogenesis by ectodomain shedding of Using chimeric mice, this report was the first to attenuator of Runx2 in the transcriptional
receptor activator of NF-κB ligand. J. Biol. Chem. provide in vivo evidence that NFATc1 is essential program of osteoblast differentiation. Genes Dev.
281, 36846–36855 (2006). for osteoclast differentiation. 17, 1979–1991 (2003).
The authors show the functional difference 64. Matsuo, K. et al. Nuclear factor of activated T-cells 89. Hayashi, T., Kaneda, T., Toyama, Y., Kumegawa, M. &
between membrane-bound and soluble RANKL in (NFAT) rescues osteoclastogenesis in precursors Hakeda, Y. Regulation of receptor activator of NF-κB
osteoclastogenesis. lacking c-Fos. J. Biol. Chem. 279, 26475–26480 ligand-induced osteoclastogenesis by endogenous
39. Fata, J. E. et al. The osteoclast differentiation factor (2004). interferon-β (INF-β) and suppressors of cytokine
osteoprotegerin-ligand is essential for mammary 65. Crotti, T. N. et al. NFATc1 regulation of the human signaling (SOCS). The possible counteracting role
gland development. Cell 103, 41–50 (2000). β3 integrin promoter in osteoclast differentiation. of SOCSs in IFN-β-inhibited osteoclast formation.
40. Josien, R. et al. TRANCE, a tumor necrosis factor Gene 372, 92–102 (2006). J. Biol. Chem. 277, 27880–27886 (2002).
family member, enhances the longevity and adjuvant 66. Kim, Y. et al. Contribution of nuclear factor of 90. Ohishi, M. et al. Suppressors of cytokine signaling-1
properties of dendritic cells in vivo. J. Exp. Med. activated T cells c1 to the transcriptional control and -3 regulate osteoclastogenesis in the presence of
191, 495–502 (2000). of immunoreceptor osteoclast-associated receptor inflammatory cytokines. J. Immunol. 174, 3024–3031
41. Wong, B. R. et al. TRANCE, a TNF family member, but not triggering receptor expressed by myeloid (2005).
activates Akt/PKB through a signaling complex cells-2 during osteoclastogenesis. J. Biol. Chem. 91. Jones, D. C. et al. Regulation of adult bone mass
involving TRAF6 and c-Src. Mol. Cell 4, 1041–1049 280, 32905–32913 (2005). by the zinc finger adapter protein Schnurri-3.
(1999). 67. Matsumoto, M. et al. Essential role of p38 mitogen- Science 312, 1223–1227 (2006).
42. Bachmann, M. F. et al. TRANCE, a tumor necrosis activated protein kinase in cathepsin K gene This paper reveals RUNX2 to be regulated by
factor family member critical for CD40 ligand- expression during osteoclastogenesis through immunomodulatory proteins.
independent T helper cell activation. J. Exp. Med. association of NFATc1 and PU.1. J. Biol. Chem. 92. Kim, N., Takami, M., Rho, J., Josien, R. & Choi, Y.
189, 1025–1031 (1999). 279, 45969–45979 (2004). A novel member of the leukocyte receptor complex
43. Ashcroft, A. J. et al. Colonic dendritic cells, intestinal 68. Lam, J. et al. TNF-α induces osteoclastogenesis regulates osteoclast differentiation. J. Exp. Med.
inflammation, and T cell-mediated bone destruction by direct stimulation of macrophages exposed to 195, 201–209 (2002).
are modulated by recombinant osteoprotegerin. permissive levels of RANK ligand. J. Clin. Invest. 93. Kukita, T. et al. RANKL-induced DC-STAMP is essential
Immunity 19, 849–861 (2003). 106, 1481–1488 (2000). for osteoclastogenesis. J. Exp. Med. 200, 941–946
44. Green, E. A., Choi, Y. & Flavell, R. A. Pancreatic lymph 69. Li, P. et al. RANK signaling is not required for (2004).
node-derived CD4+CD25+ T reg cells: highly potent TNFα-mediated increase in CD11hi osteoclast 94. Yagi, M. et al. DC-STAMP is essential for cell–cell
regulators of diabetes that require TRANCE–RANK precursors but is essential for mature osteoclast fusion in osteoclasts and foreign body giant cells.
signals. Immunity 16, 183–191 (2002). formation in TNFα-mediated inflammatory arthritis. J. Exp. Med. 202, 345–351 (2005).
This study indicates an immunosuppressive role of J. Bone Miner. Res. 19, 207–213 (2004). This was the first in vivo evidence for the role
RANKL through the induction of regulatory T cells. 70. Kim, N. et al. Osteoclast differentiation independent of DC-STAMP in the regulation of osteoclast
45. Loser, K. et al. Epidermal RANKL controls regulatory of the TRANCE–RANK–TRAF6 axis. J. Exp. Med. fusion.
T-cell numbers via activation of dendritic cells. 202, 589–595 (2005). 95. Suh, W. K. et al. The immune regulatory protein
Nature Med. 12, 1372–1379 (2006). 71. Takayanagi, H., Sato, K., Takaoka, A. & Taniguchi, T. B7-H3 promotes osteoblast differentiation and
46. Jones, D. H. et al. Regulation of cancer cell Interplay between interferon and other cytokine bone mineralization. Proc. Natl Acad. Sci. USA
migration and bone metastasis by RANKL. Nature systems in bone metabolism. Immunol. Rev. 101, 12969–12973 (2004).
440, 692–696 (2006). 208, 181–193 (2005). 96. Zhao, C. et al. Bidirectional ephrinB2–EphB4
The authors point to the notion that the 72. Takayanagi, H. et al. RANKL maintains bone signaling controls bone homeostasis. Cell Metab.
chemokine-like function of RANKL contributes to homeostasis through c-Fos-dependent induction 4, 111–121 (2006).
tumour metastasis. of interferon-β. Nature 416, 744–749 (2002). This report was the first to describe the
47. Wong, B. R. et al. The TRAF family of signal 73. Takayanagi, H. et al. T cell-mediated regulation of bidirectional function of the ephrin–EPH-family
transducers mediates NF-κB activation by the osteoclastogenesis by signalling cross-talk between molecules in osteoclast–osteoblast interactions.
TRANCE receptor. J. Biol. Chem. 273, 28355–28359 RANKL and IFN-γ. Nature 408, 600–605 (2000). 97. Takegahara, N. et al. Plexin-A1 and its
(1998). 74. Jimi, E. & Ghosh, S. Role of nuclear factor-κB in the interaction with DAP12 in immune responses and
48. Lomaga, M. A. et al. TRAF6 deficiency results in immune system and bone. Immunol. Rev. 208, 80–87 bone homeostasis. Nature Cell Biol. 8, 615–622
osteopetrosis and defective interleukin-1, CD40, and (2005). (2006).
LPS signaling. Genes Dev. 13, 1015–1024 (1999). 75. Ruocco, M. G. et al. IκB kinase (IKK)β, but not IKKα, 98. Humphrey, M. B., Lanier, L. L. & Nakamura, M. C.
49. Naito, A. et al. Severe osteopetrosis, defective is a critical mediator of osteoclast survival and is Role of ITAM-containing adapter proteins and their
interleukin-1 signalling and lymph node required for inflammation-induced bone loss. receptors in the immune system and bone. Immunol.
organogenesis in TRAF6-deficient mice. Genes Cells J. Exp. Med. 201, 1677–1687 (2005). Rev. 208, 50–65 (2005).
4, 353–362 (1999). 76. Novack, D. V. et al. The IκB function of NF-κB2 p100 99. Koga, T. et al. Costimulatory signals mediated
50. Gohda, J. et al. RANK-mediated amplification of controls stimulated osteoclastogenesis. J. Exp. Med. by the ITAM motif cooperate with RANKL for
TRAF6 signaling leads to NFATc1 induction during 198, 771–781 (2003). bone homeostasis. Nature 428, 758–763 (2004).
osteoclastogenesis. EMBO J. 24, 790–799 (2005). 77. Ray, N. et al. c-Fos suppresses systemic inflammatory This study shows that, in addition to RANKL and
51. Kadono, Y. et al. Strength of TRAF6 signalling response to endotoxin. Int. Immunol. 18, 671–677 M-CSF, immunoglobulin-like receptor signals
determines osteoclastogenesis. EMBO Rep. (2006). provide essential co-stimulation in osteoclast
6, 17–176 (2005). 78. Zenz, R. et al. Psoriasis-like skin disease and arthritis differentiation.
52. Kobayashi, N. et al. Segregation of TRAF6-mediated caused by inducible epidermal deletion of Jun 100. Mocsai, A. et al. The immunomodulatory adapter
signaling pathways clarifies its role in proteins. Nature 437, 369–375 (2005). proteins DAP12 and Fc receptor γ-chain (FcRγ)
osteoclastogenesis. EMBO J. 20, 1271–1280 (2001). 79. Bakiri, L. et al. Role of heterodimerization of c-Fos regulate development of functional osteoclasts
53. Franzoso, G. et al. Requirement for NF-κB in and Fra1 proteins in osteoclast differentiation. through the Syk tyrosine kinase. Proc. Natl Acad. Sci.
osteoclast and B-cell development. Genes Dev. Bone 23 Dec 2006 (doi:10.1016/j.bone.2006.11.005). USA 101, 6158–6163 (2004).
11, 3482–3496 (1997). 80. Peng, S. L., Gerth, A. J., Ranger, A. M. & 101. Mao, D., Epple, H., Uthgenannt, B., Novack, D. V. &
54. Iotsova, V. et al. Osteopetrosis in mice lacking NF-κB1 Glimcher, L. H. NFATc1 and NFATc2 together Faccio, R. PLCγ2 regulates osteoclastogenesis
and NF-κB2. Nature Med. 3, 1285–1289 (1997). control both T and B cell activation and via its interaction with ITAM proteins and GAB2.
55. Chen, Z. J. Ubiquitin signalling in the NF-κB pathway. differentiation. Immunity 14, 13–20 (2001). J. Clin. Invest. 116, 2869–2879 (2006).
Nature Cell Biol. 7, 758–765 (2005). 81. Koga, T. et al. NFAT and Osterix cooperatively 102. Aoki, K. et al. The tyrosine phosphatase SHP-1
56. Wada, T. et al. The molecular scaffold Gab2 is a crucial regulate bone formation. Nature Med. 11, 880–885 is a negative regulator of osteoclastogenesis and
component of RANK signaling and osteoclastogenesis. (2005). osteoclast resorbing activity: increased resorption
Nature Med. 11, 394–399 (2005). 82. Sun, L. et al. Calcineurin regulates bone formation and osteopenia in mev/mev mutant mice. Bone
57. Akira, S. & Takeda, K. Toll-like receptor signalling. by the osteoblast. Proc. Natl Acad. Sci. USA 25, 261–267 (1999).
Nature Rev. Immunol. 4, 499–511 (2004). 102, 17130–17135 (2005). 103. Umeda, S. et al. Deficiency of SHP-1 protein-tyrosine
58. Wagner, E. F. & Eferl, R. Fos/AP-1 proteins in bone and 83. Winslow, M. M. et al. Calcineurin/NFAT signaling phosphatase activity results in heightened osteoclast
the immune system. Immunol. Rev. 208, 126–140 in osteoblasts regulates bone mass. Dev. Cell function and decreased bone density. Am. J. Pathol.
(2005). 10, 771–782 (2006). 155, 223–233 (1999).

NATURE REVIEWS | IMMUNOLOGY VOLUME 7 | APRIL 2007 | 303


© 2007 Nature Publishing Group
REVIEWS

104. Takeshita, S. et al. SHIP-deficient mice are severely 113. Lee, S. K. et al. T lymphocyte-deficient mice lose Acknowledgements
osteoporotic due to increased numbers of hyper- trabecular bone mass with ovariectomy. J. Bone We thank Y. Choi, G. Karsenty, N. Takahashi, K. Matsuo,
resorptive osteoclasts. Nature Med. 8, 943–949 Miner. Res. 21, 1704–1712 (2006). T. Nakashima, M. Asagiri, T. Koga and M. Shinohara for
(2002). 114. Teitelbaum, S. L. Postmenopausal osteoporosis, critical reading of the manuscript and fruitful discussion.
105. Bromley, M. & Woolley, D. E. Chondroclasts and T cells, and immune dysfunction. Proc. Natl Acad. Sci. The work was supported in part by Grants-in-Aid for Creative
osteoclasts at subchondral sites of erosion in the USA 101, 16711–16712 (2004). Scientific Research from Japan Society for the Promotion of
rheumatoid joint. Arthritis Rheum. 27, 968–975 115. Visnjic, D. et al. Conditional ablation of the osteoblast Science, SORST program of Japan Science and Technology
(1984). lineage in Col2.3∆tk transgenic mice. J. Bone Miner. Agency, Grants-in-Aid for the 21st century COE program
106. Takayanagi, H. et al. Suppression of arthritic bone Res. 16, 2222–2231 (2001). and Genome Network Project from Ministry of Education,
destruction by adenovirus-mediated csk gene transfer 116. Rattis, F. M., Voermans, C. & Reya, T. Wnt signaling in Culture, Sports, Science, and Technology of Japan (MEXT),
to synoviocytes and osteoclasts. J. Clin. Invest. 104, the stem cell niche. Curr. Opin. Hematol. 11, 88–94 Grants-in-Aid for Scientific Research from MEXT, and Health
137–146 (1999). (2004). Sciences Research Grants from the Ministry of Health,
107. Pettit, A. R. et al. TRANCE/RANKL knockout mice are 117. Tokoyoda, K., Egawa, T., Sugiyama, T., Choi, B. I. & Labour and Welfare of Japan.
protected from bone erosion in a serum transfer model Nagasawa, T. Cellular niches controlling B lymphocyte
of arthritis. Am. J. Pathol. 159, 1689–1699 (2001). behavior within bone marrow during development. Competing interests statement
108. Redlich, K. et al. Osteoclasts are essential for Immunity 20, 707–718 (2004). The authors declare no competing financial interests.
TNF-α-mediated joint destruction. J. Clin. Invest. 118. McClung, M. R. et al. Denosumab in postmenopausal
110, 1419–1427 (2002). women with low bone mineral density. N. Engl. J. Med.
109. Horwood, N. J. et al. Activated T lymphocytes support 354, 821–831 (2006). DATABASES
osteoclast formation in vitro. Biochem. Biophys. Res. 119. Lane, N. E. et al. RANKL inhibition with Denosumab The following terms in this article are linked online to:
Commun. 265, 144–150 (1999). decreases markers of bone and cartilage turnover in Entrez Gene:
110. Kotake, S. et al. IL-17 in synovial fluids from patients patients with rheumatoid arthritis. Arthritis Rheum. http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?db=gene
with rheumatoid arthritis is a potent stimulator of 54, S225–S226 (2006). DAP12 | IL-1 | M-CSF | NF-κB | OSTERIX | RANKL | RUNX2
osteoclastogenesis. J. Clin. Invest. 109, 1345–1352 120. Nishimoto, N. & Kishimoto, T. Interleukin 6: from
(1999). bench to bedside. Nature Clin. Pract. Rheumatol. FURTHER INFORMATION
111. Sato, K. et al. TH17 functions as an osteoclastogenic 2, 619–626 (2006). Hiroshi Takayanagi’s homepage:
helper T cell subset that links T cell activation and bone 121. Urushibara, M. et al. The antirheumatic drug http://osteoimmunology.com/
destruction. J. Exp. Med. 203, 2673–2682 (2006). leflunomide inhibits osteoclastogenesis by
112. Cenci, S. et al. Estrogen deficiency induces bone loss interfering with receptor activator of NF-κB SUPPLEMENTARY INFORMATION
by increasing T cell proliferation and lifespan through ligand-stimulated induction of nuclear factor of See online article: S1 (table)
IFN-γ-induced class II transactivator. Proc. Natl Acad. activated T cells c1. Arthritis Rheum. 50, 794–804 Access to this links box is available online.
Sci. USA 100, 10405–10410 (2003). (2004).

304 | APRIL 2007 | VOLUME 7 www.nature.com/reviews/immunol


© 2007 Nature Publishing Group

You might also like