Cytokines and the pathogenesis of osteoporosis






Receptor activator of nuclear factor-κB ligand, receptor activator of nuclear factor-κB, and osteoprotegerin


Receptor activator of nuclear factor-κB (RANK) ligand (RANKL) is tumor necrosis factor (TNF) superfamily ligand 11 (TNFSF11) and together with its RANK and osteoprotegerin (OPG) (TNFRSF11A and TNFRSF11B, respectively) forms the critical paracrine system, which regulates osteoclast formation .


The discovery of RANKL, which has potent activity to increase the formation of osteoclasts from precursor cells and the bone-resorbing activity and life span of mature osteoclasts, defined a new paradigm for how a variety of cells in the bone marrow microenvironment regulate bone resorption . RANKL directs the terminal differentiation of osteoclast precursor cells. In addition, it stimulates and maintains the resorptive activity of mature cells. Most importantly, this activity of RANKL is reproduced in vitro in the absence of bone marrow stromal cells .


RANKL-deficient mice have significant osteopetrosis and no osteoclasts, but a normal number of monocyte/macrophages . They also fail to erupt teeth, which is a common finding in all causes of developmental osteopetrosis. Marrow stromal cells, hypertrophic chondrocytes, osteoblasts, and osteocytes produce RANKL . In addition, the production of RANKL by B-lymphocytes is important for regulating the enhanced bone resorption and decreased bone mass that occurs after ovariectomy in mice , while T-lymphocyte-generated RANKL may mediate bone loss in patients with HIV infection . Targeted gene deletion of RANKL in the osteocytes and maturing osteoblasts of mice demonstrated the critical nature of RANKL expression by these cells for the maintenance of osteoclastogenesis after fetal development . Recently, it was found that membrane-bound RANKL can bind RANK on vesicles, which are secreted by maturing osteoclasts, and reverse signal into osteoblasts to regulate their activity .


RANKL exists either in a cell membrane-bound or soluble form . Loss of soluble RANKL increased bone mass and decreased osteoclast number in adult, but not in growing mice. However, deletion of soluble RANKL had no effect on the bone loss that occurs after ovariectomy . Many well-known resorption stimulators, including cytokines and hormones, exert their primary osteoclastogenic activity by inducing RANKL expression in mesenchymal lineage cells . Conversely, the shedding of membrane-bound RANKL is a mechanism for inhibiting osteoblast-mediated osteoclast formation by removing RANKL from the cell surface of osteoblasts . This process depends on expression of matrix metalloproteinase (MMP) 14 , since mice that lacked this enzyme have increased osteoclast number.


OPG is a novel, secreted inhibitor of osteoclast formation that acts as a decoy receptor for RANKL . It was initially identified as a soluble factor, having inhibitory effects on osteoclastogenesis in vitro , and an inducer of osteopetrosis when it was transgenically overexpressed in mice . In marrow, it is produced by a variety of cells including stromal cells, B-lymphocytes, and dendritic cells . Besides RANKL, OPG also binds the TNF-like ligand TRAIL (TNF-related apoptosis inducing ligand) . Mice that lack OPG have severe osteoporosis, an increased number of osteoclasts and arterial calcification . The latter finding highlights a potential genetic link between osteoporosis and vascular calcification . Overexpression of OPG in transgenic mice caused osteopetrosis, decreased osteoclast numbers, and extramedullary hematopoiesis . Serum levels of OPG increase with age in humans and are directly correlated with whole body bone mass . Conversely, serum titers of OPG antibodies are inversely correlated with bone mass . Genetic inactivation of OPG is a cause of juvenile Paget’s disease , while decreases in OPG are associated with glucocorticoid-induced osteoporosis in animal and in vitro models .


The biologically active receptor for RANKL is RANK. It was first identified on dendritic cells , but it is also present on osteoclast precursors and mature osteoclasts . RANK expression at the RNA level is detected in a variety of cell types and tissues . RANK-deficient mice phenocopy the defective osteoclast development of RANKL-deficient mice, confirming the exclusive specificity of RANKL for RANK . In humans, gain-of-function mutations in RANK are associated with familial expansile osteolysis, expansible skeletal hyperphosphatasia, and juvenile Paget’s disease, all of which are diseases that are caused by excessive formation and activity of osteoclasts .


While osteoclast-like cells can form in vitro in the absence of RANK or TNF receptor associated factor (TRAF) 6 signaling when exposed to a cocktail of cytokines and growth factors , the significance of this in vitro finding is controversal since osteoclasts are generally absent in RANK-deficient animals . In most instances, cytokines and growth factors other than RANKL, which are produced at sites of inflammation or physiologically during bone turnover, act as cofactors that enhance or modulate the response of osteoclasts and their precursors to RANKL–RANK stimulation . However, it was demonstrated that in the absence of the p100 precursor of NF-κB, TNFα was a potent stimulator of osteoclastogenesis . In addition, in inflammatory arthritis RANKL-independent osteoclastogenesis in vivo has been observed .



Colony-stimulating factor-1


In addition to RANKL, colony-stimulating factor (CSF)-1 (also known as macrophage CSF) is critical for normal osteoclast formation. This cytokine was originally identified by its ability to regulate macrophage formation . However, it was subsequently shown that a spontaneous mouse mutant (the op / op mouse) with a phenotype of absent osteoclasts and defective macrophage/monocyte formation was deficient in CSF-1 . Injection of CSF-1 into op / op mice corrected the defect in osteoclast formation and bone resorption , as did the expression of CSF-1 protein specifically in osteoblastic cells .


Stimulators of bone resorption can increase the production of CSF-1 in bone , and multiple transcripts of CSF-1 are produced by alternative splicing resulting in either a soluble or membrane-bound form of the protein . In vivo treatment with CSF-1 increased osteoclast number and bone resorption as well as the rate of fracture repair . Expression of the membrane-bound form of CSF-1 is regulated by stimulators of resorption and facilitates the differentiation of precursor cells into mature osteoclasts . However, the deletion of just the soluble form of CSF-1 in mice prevented the bone loss that occurs with estrogen deficiency . This result demonstrates the critical importance of soluble CSF-1 in mediating bone loss in this animal model of postmenopausal osteoporosis.


CSF-1 has multiple effects on osteoclast precursors. It stimulates their replication and differentiation , and it regulates their motility . In mature osteoclasts, CSF-1 augments RANKL-induced resorptive activity . Environmental factors appear to modulate the response of osteoclast precursor cells to CSF-1 since it was found that there were differences in in vitro responses depending on whether cells were cultured on plastic or bone .


CSF-1 receptor (CSF-1R, also known as c-Fms), which binds CSF-1 to activate intracellular signaling, is a tyrosine kinase . Signaling through CSF-1R involves the immunoreceptor tyrosine-based activation motif–containing protein, DAP12, and β-catenin .


The role of CSF-1 in regulating osteoclast apoptosis has also been examined. Addition of CSF-1 to mature osteoclast cultures prolongs their survival . This response may be important for the development of the osteopetrotic phenotype in op / op mice, since transgenic expression of B-cell CLL/lymphoma 2 (Bcl-2), which blocks apoptosis in myeloid cells, partially reversed the defects in osteoclast and macrophage development in these animals . The effects of CSF-1 on osteoclasts has been linked to the activation of a Na/HCO cotransporter . CSF-1 is also a potent stimulator of RANK expression in osteoclast precursor cells and is critical for expanding the osteoclast precursor pool size . Strategies are being developed to inhibit CSF-1 signaling as a mechanism to treat disease .


Most recently, interleukin (IL)-34 was found to be an additional ligand for CSF-1R . Like CSF-1, it can be added with RANKL to in vitro cultures to stimulate osteoclastogenesis . Furthermore, injection of IL-34 into mice increased the proportion of CD11b-positive cells, which contain osteoclast precursors, and decreased trabecular bone mass . Production of IL-34 may be responsible for the spontaneous rescue of the osteoclast phenotype that occurs in op/op mice with age and may also explain why the CSF-1R deficient mouse has a more severe phenotype than the op/op mouse . Differences in the ability of CSF-1R and IL-34 to polarize macrophages into M1 and M2 subsets have been identified .



Additional colony stimulating factors


Like CSF-1, the CSFs, granulocyte macrophage CSF (GM-CSF) and IL-3, affect osteoclast differentiation . Both have complex actions that are dependent on the lineage stage of the myeloid precursor cells with which they interact. IL-3 is produced by osteoblasts . It has multiple effects on in vitro osteoclastogenesis , which have been associated with both stimulatory and inhibitory responses, depending on the cells that are examined and the culture conditions . One mechanism by which IL-3 inhibits osteoclastogenesis is through the regulation of c-Fos and Id protein expression . Another is through differential regulation of soluble and membrane-bound RANKL expression in osteoblastic lineage cells . IL-3 is also reported to inhibit osteoblast differentiation in multiple myeloma and to enhance mesenchymal stem cell differentiation into osteoblasts .


In early multipotential myeloid precursors, GM-CSF inhibits RANKL-mediated osteoclastogenesis and enhances the number of osteoclast precursor cells . It does this by directing the common myeloid precursor cell toward the dendritic cell lineage . One mechanism for this effect is increased shedding of CSF-1R through upregulation of “a disintegrin and metalloproteinase 17” (ADAM17), which is also known as TNFα converting enzyme . Interestingly, GM-CSF-expanded dendritic lineage cells can still maintain their capacity to differentiate into osteoclasts and may be a source of osteoclast progenitors in inflammatory conditions .


GM-CSF also inhibits the expression of monocyte chemotactic protein 1 [MCP-1, C-C motif chemokine ligand (CCL)2] by osteoclast precursor cells . MCP-1 is a chemokine involved in osteoclast motility. Both GM-CSF and IL-3 inhibit the expression of TNF receptors on myeloid precursor cells . However, in prefusion osteoclast precursors, which are myeloid cells that have been stimulated with RANKL and CSF-1 for 3 days to a point where they will shortly (within 6 hours) fuse into osteoclasts, treatment with GM-CSF+CSF-1 enhanced osteoclastogenesis and mimicked the response to RANKL+CSF-1 . This increased osteoclastogenesis was mediated by upregulation of the cell fusion protein, dendritic cells-specific transmembrane protein in the prefusion osteoclasts . It has also been shown that if multipotential myeloid precursor cells are cultured sequentially with GM-CSF and then with CSF-1+RANKL, they form osteoclasts and, in some instances, act as dendritic cells, which present antigen to T-lymphocytes and initiate the adaptive immune response . IL-3 and GM-CSF may also support osteoclast differentiation by stimulating CSF-1 production .


At relatively high doses granulocyte-colony stimulating factor (G-CSF) decreased bone mass in rodents when injected systemically . This response appeared to result from increased osteoclast formation, decreased osteoblast function, and increased osteoblast apoptosis. Similar effects are seen in humans . G-CSF also mobilized the migration of hematopoietic precursor cells from the bone marrow into the circulation and increased the number of circulating osteoclast precursor cells , which may be related to its ability to increase osteoclast resorptive activity.


In mice, overexpression of G-CSF inhibited the ability of osteoblasts to respond to bone morphogenetic protein . Short-term treatment of mice with G-CSF decreased endosteal and trabecular osteoblasts by increasing their apoptosis and inhibiting osteoblast precursor cell differentiation . Mice overexpressing G-CSF had increased bone resorption, which, in contrast to wild type mice, was not increased with ovariectomy . Targeted deletion of ADAM17 in mice using Cre/Lox technology and the Sox9 promoter to target Cre recombinase to chondroprogenitor cells, produced a phenotype of enhanced G-CSF production, osteoporosis and extramedullary hematopoiesis . These results suggest that the production of ADAM17 on osteoblasts and/or chondroblasts regulates the expression of G-CSF.



Interleukin-1


There are two separate IL-1 genes, IL-1α and IL-1β, which have identical activities . IL-1 is a potent peptide stimulator of bone resorption . It directly affects RANKL-primed osteoclasts to resorb by a mechanism dependent on microphthalmia transcription factor . It also has indirect resorptive actions through its ability to stimulate RANKL production . In addition, both RANKL- and 1,25-dihydroxyvitamin D 3 -stimulated osteoclast formation in vitro was mediated, in part, by their effects on IL-1 production . IL-1 also increases prostaglandin synthesis in bone , which may enhance its resorptive activity since prostaglandins are potent resorption stimuli . Direct stimulation of osteoclastogenesis by IL-1 in mixed murine stromal and hematopoietic cell cultures is dependent on RANKL but not TNFα expression in the stromal/osteoblastic cells .


In mouse models, IL-1 appears to be involved in normal growth plate development and bone turnover . It also may be essential for the systemic bone loss that is seen in some inflammatory conditions due to high TNF production . In addition, it mediates some of the effects of estrogen withdrawal on bone loss in both mice and humans . In humans, levels of IL-1have been negatively correlated to measurements of bone mass .


IL-1 is produced in bone , and its activity is present in bone marrow serum . One source of bone cell-derived IL-1 is osteoclast precursor cells, which produce IL-1 when they interact with bone matrix . There is also a natural inhibitor of IL-1, IL-1 receptor antagonist, which is an analog of IL-1 that binds, but does not activate the biologically important type I IL-1 receptors .


There are two known receptors for IL-1: type I and type II . All known biologic responses to IL-1 appear to be mediated exclusively through the type I receptor . IL-1 receptor type I requires interaction with a second protein, IL-1 receptor accessory protein, to generate postreceptor signals . Signaling through type I receptors involves the activation of specific TRAFs and NF-κB . IL-1 receptor type II is a decoy receptor that prevents activation of type I receptors . One report found a decrease in the bone mass of mice that were deficient in the bioactive type I IL-1 receptor . However, this has not been our experience .


Expression of myeloid differentiation factor 88 but not Toll/IL-1 receptor domain-containing adapter inducing interferon-beta (TRIF) was necessary for IL-1 to stimulate RANKL production in osteoblasts and to prolong the survival of osteoclasts . Survival of osteoclasts by treatment with IL-1 required PI3-kinase/AKT and extracellular signal-regulated kinase (ERK) .


The effects of IL-1 on bone in inflammatory states, such as rheumatoid arthritis, are multiple and mediated by both direct and indirect mechanisms. IL-1 stimulates bone resorption and inhibits bone formation through its effects on osteoclasts and osteoblasts, respectively . In inflammatory conditions, it is directly involved in the production of a relatively unique population of very active osteoclasts . In addition, it stimulates the production of a variety of secondary factors in the bone microenvironment including prostaglandins and GM-CSF, which have complex effects on bone cells themselves . IL-1 also has been reported both to inhibit and to stimulate the production of OPG in various osteoblastic cell models in vitro . IL-1’s effects on osteoclast precursor cell migration are mediated by its ability to stimulate production of the chemokine CX3CL1 .


IL-1 induces the differentiation of mesenchymal stem cells toward osteoblasts via the noncanonical Wnt-5a/Ror2 pathway . However, it also inhibits osteoblast migration and the ability of mesenchymal stem cells to promote tissue regeneration . IL-1 also is involved in the mechanisms of mechanosensing in bone . Osteocytes regulate osteoclastogenesis by producing a variety of factors including RANKL . Treatment of the MLO-Y4 osteocyte-like cell line with IL-1 enhanced RANKL and decreased OPG production, which was reversed by mechanically loading the cells .



Tumor necrosis factor


Like IL-1, TNF represents a family of two related polypeptides (α and β) that are the products of separate genes . TNFα and TNFβ have similar biologic activities and are both potent stimulators of bone resorption .


In vivo administration of TNFα was shown to increase the serum calcium of mice and to stimulate new osteoclast formation and bone resorption . Like IL-1, TNF also enhances the formation of osteoclast-like cells in bone marrow culture through its ability to increase RANKL production . However, RANKL is not the only cytokine that TNF stimulates in bone, and many of these enhance the response to RANKL. For example, TNF stimulates osteoclast formation in mixed stromal cell/osteoclast precursor cell cultures by a mechanism that was partially dependent on the production of IL-1 . In addition, TNF-induced osteolysis was found to be dependent on CSF-1 production .


TNF can also directly stimulate osteoclast formation in vitro, independent of RANK, since it occurs in cells from RANK-deficient mice . However, the significance of this in vitro finding is controversial. In vivo administration of TNF to RANK-deficient mice caused only an occasional osteoclast to form . In addition, RANK-deficient mice that also overexpressed TNF had severe osteopetrosis and no osteoclasts . It was also demonstrated that TNF can stimulate osteoclastogenesis in mice that are deficient in the p100 precursor protein of NF-κB, which is a critical signaling molecule in RANKL-mediated stimulation of osteoclastogenesis and bone resorption . These results demonstrate that even though both TNF and RANKL are members of the same cytokine superfamily and share multiple overlapping signaling pathways, there are crucial differences between their downstream signaling molecules in osteoclast precursor cells. It has also been shown that in cherubism, a disease with elevated production of TNF due to a gain-of-function mutation in the SRC homology 3 domain binding protein 2 (Sh3bp2) gene, bone resorption can occur through the action of cells that are not classic osteoclasts . There may also be a role for sclerostin, a Wnt signaling inhibitor, in the ability of TNF to induce inflammatory arthritis since deletion or inhibition of sclerostin exacerbated the inflammatory joint destruction seen in TNF transgenic mice .


As with IL-1, TNF binds to two cell surface receptors, TNF receptor 1 or p55, and TNF receptor 2 or p75 . In contrast to IL-1, both receptors transmit biologic responses. However, the principal effects on bone cells appear to be mediated through TNF receptor 1 . Mice deficient in both TNF receptor 1 and TNF receptor 2 have been produced . These animals appear healthy and are not reported to have an abnormal bone phenotype.


TNF can stimulate the expression of the CSF-1R in osteoclast precursor cells and, through this mechanism, increase their number . It also enhances RANK signaling, which activates osteoclasts and their precursor cells and enhances the expression of the costimulatory molecule, paired Ig-like receptor A, which enhances nuclear factor of activated T cells cytoplasmic 1 (NFATc1) activation . Recently, it was found that RANKL enhances TNF-induced osteoclastogenesis independent of effects on TRAF6 by degrading TRAF3 in osteoclast precursor cells . Mice that overexpress TNF have an increased number of CD11b high osteoclast precursor cells in their spleen and blood . The enhancement of osteoclastogenesis that occurs with TNF signaling involves multiple pathways, including phosphoinositide 3-kinase/AKT (PI3K/Akt)-mediated Blimp1 expression and recombination signal binding protein for immunoglobulin kappa J region-mediated regulation of miR-182 .


TNF has biphasic effects on bone formation and osteoblast function, which may be dose and time related . At lower doses, it stimulated the differentiation of mesenchymal precursor cells into osteoblasts , while at higher concentrations it inhibited osteoblast function and bone formation . It was reported to promote fracture repair by enhancing the recruitment of precursor cells to the osteoblastic lineage . The inhibitory effects of TNF on osteoblasts appear to be direct and mediated by downregulation of the critical transcription factor genes, RUNX2 and osterix . TNF has many actions on osteoblasts including inhibiting type 1 collagen and osteocalcin synthesis , which are essential for differentiated osteoblast function. It also stimulated osteoblast apoptosis , suppressed production of insulin-like growth factor (IGF)-1 and downregulated EphB4 signaling . Some of the inhibitory effects of TNF on osteoblast differentiation are mediated by activated transcription factor 3 through a pathway involving c-Jun N-terminal kinase (JNK) .


Most recently, patients with inflammatory diseases who were treated with anti-TNF therapy were found to have increased bone mineral density . However, in a study of patients with inflammatory bowel disease who were treated with anti-TNF therapy, the incidence of fractures was not affected even though bone mass increased . This result suggests that circulating TNF, which is produced by localized inflammatory pathology, has systemic effects on bone mass but not necessarily on bone strength. Production of TNF on B-lymphocytes may also contribute to the osteoclastogenesis produced in periodontitis . Finally, a polymorphism in the TNF-α gene has been associated with the risk of developing osteoporosis in women .



Additional tumor necrosis factor superfamily members



Fas-ligand


Fas-ligand (FasL), which binds to its receptor Fas on responsive cells, regulates apoptosis and other cellular processes in multiple cell types . In osteoblasts, FasL inhibits differentiation through a caspase 8-mediated mechanism . In osteoclasts, addition of FasL to cultures of osteoclast precursor cells, which were also treated with CSF-1 and RANKL, increased osteoclast formation. Osteoclast precursors and mature osteoclasts express Fas and FasL . Expression of Fas was upregulated by RANKL treatment in the RAW 264.7 osteoclast precursor cell line and treatment of mature osteoclasts with Fas-induced apoptosis . However, in contrast to their similar effects on osteoclastogenesis in cultures of precursor cells, there appears to be divergent roles of RANKL and FasL on mature osteoclast apoptosis. At high concentrations, RANKL inhibited the ability of FasL to induce apoptosis . The effect that FasL deficiency has on bone mass is controversial. One group has found that bone mass is decreased in FasL-deficient mice while another found it to be increased . However, the significance of studying bone mass in Fas or FasL-deficient mice is questionable since these models have a generalized lymphroliferative disorder, which activates a wide variety of immune responses that affect bone and makes it difficult to interpret the results of these studies.


It appears that Fas signaling is involved in the effects of estrogen on bone . However, there has been controversy about the exact role that FasL has in this response. One group found that stimulation of estrogen receptor α (ERα) in osteoclasts in mice enhanced FasL production, which, in turn, reduced the rates of bone loss by increasing osteoclast apoptosis . In contrast, a second group failed to detect expression of FasL in osteoclasts . Rather, they found that estrogen enhanced FasL production in osteoblasts. They speculated that estrogen-induced increases in FasL production in osteoblasts regulated osteoclast apoptosis through a paracrine mechanism . More recently, Fas receptor was shown to be required for estrogen deficiency-induced bone loss . Estrogens, interacting through ERα, stimulated metalloproteinase 3 expression on osteoblasts, which induced FasL cleavage from osteoblasts. In turn, this solubilization of FasL produced osteoclast apoptosis . Fas–Fas ligand interactions may also mediate some effects of interferon (INF) γ on bone . FasL also stimulated MMP2 expression in osteoblasts .



Tumor necrosis factor–related apoptosis inducing ligand


TRAIL is another TNF-superfamily member that has a wide variety of activities. Its effects on osteoclast function and bone are also controversial. Some groups have found that treatment of osteoclasts with TRAIL induced apoptosis through effects that were mediated by the receptor TRAIL-R2, which is also known as “death receptor 5” (DR5) . Others have found that TRAIL stimulated osteoclast differentiation through a TRAF-6-dependent mechanism . In vivo, the injection of TRAIL for 8 days in 4-week-old mice induced an increase in bone mass. In vitro, this effect was associated with an increase in the cyclin-dependent kinase inhibitor, p27 Kip1 through the effects of TRAIL on the ubiquitin-proteasome pathway . TRAIL may also be a factor in the effects that myeloma and microgravity have on osteoclasts . However, other groups have failed to find either in vitro or in vivo effects of recombinant TRAIL on osteoclasts or in vivo effects on bone mass .


In cultured human osteoblasts, the ability of TRAIL to induce apoptosis was dependent on their differentiation state with early cells being more responsive than more mature cells . This affect was regulated by differential expression of the active DR5 and decoy DcR2 TRAIL receptors during osteoblast differentiation.



CD40 ligand


CD40 ligand (CD40L) is involved in the differentiation of naïve T-lymphocytes into T H 1 effector cells . In humans, deficiency of CD40L causes X-liked hyper IgM (XHIM) syndrome. Bones of XHIM patients develop spontaneous fractures and are osteopenic . Activated T-lymphocytes from XHIM patients have normal amounts of RANKL but deficient INF-γ production, which may contribute to their decreased bone mass . In addition, the expression of CD40L in rheumatoid arthritis synovial cells induced RANKL expression and enhanced their ability to stimulate osteoclastogenesis. This result suggests that this mechanism is involved in the effects of rheumatoid arthritis on bone . CD40L was also found to accelerate the osteoclastogenesis that is induced by RANKL and lipopolysaccharide (LPS) . The ability of parathyroid hormone (PTH) to stimulate osteoclastogenesis has been reported to involve induction of CD40L on T-lymphocytes and the subsequent induction of responses in stromal cells, expressing the receptor CD40 . CD40L has also been implicated in the bone loss that occurs after ovariectomy in mice and the anabolic response to intermittent PTH . Recently, T-lymphocyte-derived CD40L has been implicated in the ability of osteoclasts to suppress T-lymphocyte activation by a mechanism involving indoleamine 2,3-dioxygenase production in osteoclasts . A gene association study found correlation between bone mineral density and single nucleotide polymorphisms (SNPs) in both CD40 and CD40L .



Interleukin-6


IL-6, like IL-1 and TNF, has a wide variety of activities on immune cell function and on the replication and differentiation of a number of cell types . Osteoblastic cells (both rodent and human) produce IL-6 and IL-6 receptors . Another source of IL-6 in the bone microenvironment is bone marrow stromal cells, which can produce IL-6 after they are stimulated with IL-1 and TNF . The receptor for IL-6 is composed of two parts: a specific IL-6 binding protein (IL-6 receptor), which can be either membrane-bound or soluble, and gp130, an activator protein that is common to a number of cytokine receptors . Soluble IL-6 receptor binds IL-6, and this complex can then activate cells that contain the gp130 signal peptide . The shedding of IL-6 receptor from osteoblasts is stimulated by IL-1 and TNFα .


The ability of IL-6 to affect bone resorption in vitro is variable and depends on the assay system that is used as both stimulatory and inhibitory effects have been observed . It appears that a major effect of IL-6 is to regulate the differentiation of osteoclast progenitor cells into mature osteoclasts . IL-6 also directly stimulates both RANKL and OPG mRNA production in bone and enhances production of prostaglandins . In addition, one publication suggested that IL-6 can stimulate osteoclastogenesis in vitro by a RANKL-independent mechanism . In contrast, two other publications found IL-6 to directly inhibit RANKL signaling in osteoclast precursor cells and decrease osteoclast formation .


Examination of IL-6-deficient mice at 8 months of age demonstrated that bone mass was increased, as was tartrate-resistant acid phosphatase (TRAP)-positive osteoclast number and alkaline phosphatase activity in osteoblasts. However, curiously, cathepsin K-positive osteoclasts were decreased in IL-6-deficient mice suggesting that loss of IL-6 inhibited osteoclast maturation and in this way enhanced bone mass. Some of this discrepancy may be related to differences in the responses of cortical and trabecular bone to membrane-bound and soluble IL-6 receptor . There was also enhanced apoptosis of osteoclasts in IL-6-deficient mice . Deletion of IL-6 in mice promoted fracture healing , and IL-6 appears to influence the bone loss that is seen in mice on a high fat diet .


IL-6 has a variety of effects on mesenchymal cells. It promotes osteogenic differentiation of bone marrow mesenchymal cells but inhibits more mature osteoblast differentiation through the activation of the Janus kinase/signal transduction and activators of transcription (JAK/STAT), Src homology region 2 domain-containing phosphatase-2/dual specificity mitogen-activated protein kinase kinase 2 (SHP2/MEK2) and SHP2/AKT pathways . It also inhibits osteogenesis in glucocorticoid-treated mice . Osterix, a key transcription factor in early osteoblast differentiation, appears to inhibit IL-6 production since mice heterozygous for osterix deficiency had increased IL-6 production . In contrast, bone morphogenetic protein, which enhances osteoblast differentiation, was shown in vitro to enhance IL-6 production in osteoblasts .


IL-6 may mediate some of the bone pathology that is seen with aging and in a variety of clinical syndromes including Paget’s disease , hypercalcemia of malignancy , fibrous dysplasia , giant cell tumors of bone inflammatory states mediated by TNF or RANKL and Gorham–Stout disease . There has been conflicting data about the role of IL-6 in PTH-mediated responses in bone as some investigators have found it critical while others have not . Inhibition of the IL-6 receptor blocked osteoclastogenesis in vitro and in vivo .


In mice, increased IL-6 expression augmented the effects of p62/sequestosome-1 mutations (which is linked to the development of Paget’s disease in humans) but did not fully reproduce the Pagetic phenotype, suggesting that additional mechanisms are involved . It was also shown in mice that measles virus nuclear capsid protein, which is also implicated as driving the development of Paget’s disease, increased IL-6 expression .


IL-6 is a mediator of inflammation, and IL-6 inhibition is now used therapeutically to treat inflammatory diseases, such as rheumatoid arthritis . Recently, anti-IL-6 therapy was shown to reduce systemic bone loss and osteoclast precursor cell number in a mouse model of rheumatoid arthritis . Furthermore, in humans with rheumatoid arthritis, serum levels of IL-6 inversely correlated with bone mass and directly correlated with disease activity .



Additional interleukin-6 family members


IL-6 is a member of a group of cytokines that share the gp130 activator protein in their receptor complex . Each family member utilizes unique ligand receptors to generate specific binding. Signal transduction through these receptors utilizes the JAK/STAT pathway .



Interleukin-11


IL-11 is produced by bone cells in response to a variety of resorptive stimuli . It stimulates osteoclast formation in murine bone marrow cultures and bone resorption in a variety of in vitro assays . Interestingly, it has no effect on isolated mature osteoclasts. In mice deficient in the specific IL-11 receptor, trabecular bone mass is increased. This effect appears to result from decreased bone turnover, which is associated with decreased in vitro osteoclast formation and resorption . After ovariectomy and subsequent estrogen withdrawal, IL-11 receptor deficient and wild type mice lost bone mass at similar rates. This result argues that IL-11 signaling is not involved in the effects of estrogen on bone mass . However, IL-11 does appear to be involved in the ability of mechanical force to stimulate osteoblast activity in vivo through its effects on Wnt signaling . It has also been shown that IL-11 mediates the stimulatory effects of PTH and mechanical stress on osteoblast differentiation via activator protein 1 and SMAD signaling .


Mutations in IL-11 receptor or IL-11 have been linked to human disease. The mutation in patients with craniosynostosis was found to generate a defective IL-11 receptor, which, in turn, caused premature closure of cranial bone sutures . In addition, an SNP, which alters the stability of IL11, was linked to short stature .



Leukemia inhibitory factor


Leukemia inhibitory factor (LIF) is produced by bone cells in response to a number of resorption stimuli . The effects of LIF on bone resorption are variable. In a number of in vitro model systems, LIF stimulated resorption by a prostaglandin-dependent mechanism . However, it also was found to have inhibitory effects in vitro . In neonatal murine calvaria cultures, LIF stimulated production of both RANKL and OPG .


The effects of LIF on osteoblast differentiation in vitro are complex and appear dependent on the dose administered and the differentiation state of the osteoblastogenic cultures that are studied . Local injection of LIF in vivo over the calvaria of mice was shown to increase both resorption and formation parameters, as well as the thickness of the treated calvaria . Expression of LIF appears downregulated during osteoblast differentiation by a mechanism that is mediated by micro-RNAs (miRs) . In mice that lacked the specific LIF receptor (LIF-R) and, hence, could not respond to LIF, bone volume was reduced and osteoclast number was increased sixfold . LIF may mediate some of its actions on bone in vivo through the inhibition of sclerostin production . Animals lacking LIF are characterized by giant osteoclasts, which are produced through mechanisms involving Fos-related antigen 2, hypoxia, hypoxia-induced factor (HIF)1α and Bcl-2 . LIF may also be involved in chondroclast production .



Oncostatin M


Oncostatin M (OSM) was demonstrated to stimulate multinuclear cell formation in murine and human bone marrow cultures . However, these cells appeared to be macrophage polykaryons and not osteoclasts . In contrast, OSM inhibited osteoclast-like cell formation that was stimulated by 1,25-dihydroxyvitamin D 3 in human marrow cultures , and it decreased bone resorption rates in fetal mouse long bone cultures . In vivo, overexpression of OSM in transgenic mice induced a phenotype of osteopetrosis . Hence, it appears that OSM is predominantly an inhibitor of osteoclast formation and bone resorption . However, OSM can affect cellular responses in bone by binding to either the OSM receptor, which produces inhibitory effects on resorption or the LIF-R, which promotes bone formation through inhibition of sclerostin expression .


OSM stimulates mesenchymal cells to differentiate toward osteoblasts and osteocytes and inhibits their differentiation toward adipocytes , an effect that may be mediated through induction of MCP-1 . Monocytes can produce OSM and drive osteoblast differentiation through a mechanism that is dependent on STAT3 . Finally, it has also been shown that OSM contributes to the anabolic response of bone to intermittent administration of PTH .


The role of all IL-6 family members in osteoclast formation has to be examined in the light of data demonstrating that mice lacking the gp130 activator protein have an increased number of osteoclasts in their bones compared with normal animals . Since gp130 is an activator of signal transduction for all members of the IL-6 family, this result argues that at least some IL-6 family members have a predominantly inhibitory effect on osteoclast formation and bone resorption.



Interleukin-7


IL-7 is a cytokine that has diverse effects on the hematopoietic and immunologic systems and is best known for its nonredundant role in supporting B- and T-lymphopoiesis. Studies have demonstrated that IL-7 also plays an important role in the regulation of bone homeostasis . However, the precise nature of how IL-7 affects osteoclasts and osteoblasts is controversial, since it has a variety of actions in different target cells. Systemic administration of IL-7 upregulated osteoclast formation from human peripheral blood cells by increasing osteoclastogenic cytokine production in T-lymphocytes . Furthermore, mice with global overexpression of IL-7 had a phenotype of decreased bone mass with increased osteoclast and no change in osteoblast number . Significantly, IL-7 did not induce bone resorption and bone loss in T-cell-deficient nude mice in vivo . In addition, treatment of mice with a neutralizing anti-IL-7 antibody inhibited ovariectomy-induced proliferation of early T-cell precursors in the thymus, demonstrating that ovariectomy upregulates T-cell development through IL-7. This latter effect may be a mechanism by which IL-7 regulates ovariectomy-induced bone loss . However, the interpretation of results from in vivo IL-7 treatment studies is complicated by secondary effects of IL-7, which result from the production of bone-resorbing cytokines by T-cells in response to activation by IL-7 .


In contrast with previously reported studies , we found differential effects of IL-7 on osteoclastogenesis . IL-7 inhibited osteoclast formation in murine bone marrow cells that were cultured for 5 days with CSF-1 and RANKL . Furthermore, IL-7-deficient mice had markedly increased osteoclast number and decreased trabecular bone mass compared to wild type controls . In addition, we found that trabecular bone loss after ovariectomy was similar in wild type and IL-7-deficient mice . Other investigators have found that IL-7 could stimulate osteoclastogenesis by a mechanism that was independent of RANKL but dependent on STAT5, and IL-7 was found to decrease bone mass and increase RANKL through a mechanism dependent on cFos/c-Jun .


IL-7 mRNA levels in bone increase with ovariectomy and this effect may be linked to alterations in osteoblast function with estrogen withdrawal . Treatment of newborn murine calvaria cultures with IL-7 inhibited bone formation, as did the injection of IL-7 above the calvaria of mice in vivo . When IL-7 was overexpressed locally by osteoblasts, trabecular bone mass was increased compared with wild type mice . Furthermore, targeted overexpression of IL-7 in IL-7-deficient mice rescued the osteoporotic bone phenotype of the IL-7-deficient mice . These studies indicated that the actions of IL-7 on bone cells are dependent on whether IL-7 is delivered systemically or locally. Production of IL-7 by osteoblasts appeared critical for normal B-lymphopoiesis and was dependent on mechanistic target of rapamycin complex 1 signaling in osterix-expressing cells . In sepsis, B-lymphopoiesis decreases through a mechanism in which osteoblastic cells are depleted as presumably is IL-7 production by these cells . The induction of this cytokines in osteoblasts is mediated by G s α-dependent signaling. Osteoclast mediated bone resorption can influence B-lymphopoiesis through effects on local IL-7 production in the bone marrow .



Interleukin-8 and other chemokines


Recruitment and homing of myeloid cells often occurs under the direction of chemokines and their receptors. This family of relatively small proteins induces interactions through cognate G-protein-coupled receptors to initiate cytoskeletal rearrangement, adhesion, and directional migration . Chemokines can be divided into four branches, depending on the spacing and sequence motifs of their first cysteine (C) residues. These are CXC, CC, C, and CX 3 C, where X is any other amino acid . The majority of chemokine receptor interactions occur through the CC and CXC chemokines, which are referred to as major, while C and CX 3 C chemokines are referred to as minor.



Interleukin-8


Many cells produce chemokines that bind specific G-protein coupled receptors. IL-8, a CXC chemokine, is produced by osteoclasts and stimulates osteoclastogenesis and bone resorption by a mechanism that is reported to be independent of the RANKL pathway . IL-8 may also be produced by certain cancers and stimulate lytic bone lesions in metastatic disease . Effects of IL-8 on bone may be partially mediated by upregulation of nitric oxide synthase expression in osteoclasts . IL-8 production is stimulated by RANKL in osteoclast precursors and may be a critical element of RANKL-induced osteoclast formation .



C-C motif chemokine ligand 2


CCL2 (MCP-1) is a potent chemokine for monocytes, and a variety of other immune cells. Its receptor is C-C motif chemokine receptor (CCR)2, which is expressed at high levels on monocytes . CCL2 is produced by osteoblasts in response to PTH and proinflammatory cytokines. One of its functions appears to be to regulate the recruitment of osteoclast precursors to bone . CCL2 is induced by RANKL in mononuclear precursor cells and enhances the ability of RANKL to stimulate osteoclast-like cell formation in these cells . The treatment of monocytes with CCL2 alone induced the production of multinucleated- and calcitonin receptor-positive cells . However, these cells did not resorb bone unless they were also exposed to RANKL .


Mice that are deficient in CCL2 have an elevated bone mass, decreased resorptive activity, a lower number of osteoclasts but normal bone formation activity . It was recently reported that the anabolic actions of intermittent PTH treatment in mice were mediated to a significant degree by CCL2 .


CCL2 may also be involved in tooth eruption, since dental follicle cells express it . Among the factors that stimulate CCL2 in the dental follicle are PTH-related protein , platelet-derived growth factor BB, and fibroblast growth factor 2 (FGF-2) . However, CCL2 is not critical for tooth eruption since there were only minor changes in the temporal pattern of this process in CCL2-deficient mice .



C-C motif chemokine ligand 3


CCL3 (macrophage inflammatory protein-1 α, MIP-1α) is a direct stimulator of osteoclastogenesis that is expressed in bone and bone marrow cells by a mechanism that is proposed to be independent of RANK activation . In addition, it can enhance RANKL expression by stromal cells and osteoblasts in the bone microenvironment . CCL3 mediates some of the osteolytic activity and the inhibition of osteoblastic bone formation that is induced by multiple myeloma. It also mediated the bone resorption that was seen in a murine model of inflammatory arthritis . Activation of osteoclastogenesis by CCL3 involves CCR1 and CCR5 . CCL3 and IL-8 also stimulate motility but suppress resorption in mature osteoclasts . CCL3 and its receptor CCR1 are reported to mediate the bone remodeling that occurs during orthodontic tooth movement and may also regulate osteoclastogenesis in osteomyelitis .



C-C motif chemokine ligand 9


CCL9 [macrophage inflammatory peptide gamma (MIP-1γ)], like CCL3, binds the receptor CCR1 and regulates osteoclast function . Injection of CSF-1 to induce osteoclastogenesis and bone resorption in osteopetrotic tl/tl rats, which lack CSF-1, caused a rapid (within 2 days) upregulation of CCL9 and CCR1 in the bones, and a rapid increase in osteoclastogenesis . Significantly, treatment of tl/tl rats with antibodies to CCL9 ameliorated the ability of CSF-1 injections to stimulate osteoclastogenesis. RANKL appears to be an inducer of CCL9 and CCR1 in osteoclasts . Induction of CCR1 by RANKL is dependent on NFATc1 expression . CCL9, and other chemokines that bind CCR1 (CCL3, CCL5, and CCL7) are produced by osteoclasts, osteoblasts, and their precursors in bone. In addition, expression of these chemokines in differentiating osteoblasts is induced by proinflammatory cytokines such as IL-1 and TNF .



C-X-C motif chemokine ligand 12 and C-X-C motif chemokine receptor 4


C-X-C motif chemokine ligand 12 (CXCL12) (stromal cell-derived factor-1) and its receptor C-X-C motif chemokine receptor 4 (CXCR4) are involved in a variety of cellular processes including hematopoietic cell homeostasis and immune responses , which are regulated by osteoblasts and their precursor cells . Osteoclast precursor cells express CXCR4 , but the level of this receptor is downregulated during differentiation toward mature osteoclast . Osteoclast precursor cells from CXCR4-deficient mice had accelerated osteoclastogenesis and enhanced resorbing activity . Furthermore, treatment of human osteoclast precursor cells with CXCL12-stimulated migration and enhanced osteoclastogenesis in response to RANKL and CSF-1 . CXCL12-induced expression of matrix MMP9 in RAW 264.7 cells, which have the characteristics of osteoclast precursor cells, and this may be a mechanism for the migration of osteoclast precursor cells toward bone . Expression of CXCL12 is upregulated in osteoclasts when they differentiate on a calcium phosphate matrix . In addition, the production of CXCL12 may be involved in the recruitment of precursor cells to form giant cell tumors of bone , the increased osteolysis that is seen in multiple myeloma , the osteoclastogenic response to LPS , and the growth of solid tumors in bone .


CXCL12 is a product of CAR (CXCL12-abundant reticular) cells in bone marrow, which maintain the hematopoietic stem cell niche and are regulated by expression of the transcription factor early B-cell factor 3 (Ebf3) .


CXCR4 signaling also seems critical for osteoblast development and function. Mice with targeted deficiency of CXCR4 in early osteoblast precursor cells had reduced bone mass and mineral apposition rates as well as abnormal growth plate architecture and enhanced adipogenesis . In contrast, in mice with targeted deletion of CXCR4 in more mature osteoblasts, bone mass was decreased and bone resorption was increased , but adiposity in the bone marrow was not affected . CXCL12 is produced in high levels by osteoblasts through a mechanism that is dependent on the transcription factor Slug . Bone morphogenic protein-2 (BMP2) stimulates osteogenic differentiation of mesenchymal precursor cells by a mechanism that appears dependent on CXCL12 . In one study, CXCL12 levels in serum were negatively associated with bone mass in the hip. However, these authors failed to find a significant association of serum CXCL12 levels with hip fracture incidence .



CX3CR1


CX3CR1 is a chemokine receptor that is present on most early myeloid lineage cells including osteoclast precursors . Its ligand is CX3CL1, which is also known as fractalkine. CX3CL1 is produced by osteoblasts and appears to stimulate the migration of osteoclast precursors to the bone surface . Antibody neutralization of CX3CR1 in both in vitro and in vivo models blocked osteoclast formation while anti-CX3CL1 antibody blocked joint destruction in a murine experimental arthritis model . Mice deficient in CX3CR1 had a slight increase in trabecular and cortical bone thickness, reduced osteoclast precursor cell and mature osteoclast number, and increased osteoid formation rates . Expression of CX3CL1 in vascular cells is reported to mediate the enhanced bone resorption that is seen after lethal irradiation in mice . A subset of inflammatory osteoclasts express CX3CR1 while osteoclasts produced during homeostasis do not .



C-C motif chemokine receptor 1


Ligands for CCR1 include CCL3 (MIP-1α), CCL5 regulated on activation, normal T cell expressed and secreted (RANTES), CCL7 (MCP-3), CCL13 (MCP-4), CCL14 (HCC-1), CCL15 (LKN-1), and CCL23 (MPIF-1) . The inhibition of CCR1 expression with small interfering RNA or by blocking NFATc1 activation with cyclosporin A, inhibited migration of RAW 264.7 cells (a model for osteoclast precursors) and murine bone marrow cells in Boyden chambers . Furthermore, the inhibition of CCR1 signaling with a mutated form of CCL5, which blocks the binding of CCR1 to its ligands, prevented osteoclast-like cell formation in murine bone marrow cultures . In addition, antibody neutralization of CCL9 inhibited RANKL-induced osteoclastogenesis by 60%–70% in murine bone marrow cultures . Mice that were deficient in CCR1 had decreased bone remodeling during orthodontic tooth movement that was induced by mechanical loading .



C-C motif chemokine receptor 2


CCR2, which binds CCL2 and CCL7 (MCP-3) appears to have major effects on osteoclasts. Mice deficient in CCR2 have increased bone mass, decreased osteoclast number, size, and resorptive activity and no defect in osteoblast function . In addition, osteoclast formation in vitro was attenuated in CCR2-deficient mice and these animals lost less bone with ovariectomy than did wild type mice . In addition, deletion of CCL2 or inhibition of CCR2 abrogated the ability of mechanical force to exacerbate inflammatory arthritis . In another example of the interactions of the CCL2/CCR2 axis and mechanical force, orthodontic tooth movement and its resultant TRAP-positive cell formation were significantly decreased in CCR2 deficient mice . Additional chemokine receptors that are produced on osteoclasts include CCR3 and CCR5 .



Interleukin-10


IL-10 is produced by activated T- and B-lymphocytes . It is a direct inhibitor of osteoclastogenesis and osteoblastogenesis , which are effects that are associated with increased tyrosine phosphorylation of multiple proteins in osteoclast precursor cells . The direct effects of IL-10 on RANKL-stimulated osteoclastogenesis include decreases in NFATc1 expression, reduced translocation of this transcription factor into the nucleus and suppressed c-Fos and c-Jun expression . Administration of IL-10 may have utility as a mechanism to control wear-induced osteolysis and the alveolar bone loss of periodontal disease . In dental follicle cells, which function to regulate tooth eruption, in vitro treatment with IL-10 inhibited RANKL production and enhance OPG . Hence, there appears to also be an indirect effect of IL-10 on osteoclastogenesis that is mediated by its ability to regulate RANKL and OPG production.


Treatment of bone marrow cell cultures with IL-10 suppressed the production of osteoblastic proteins and prevented the onset of mineralization . IL-10 also inhibited osteoclast formation in bone marrow cultures without affecting macrophage formation or the resorptive activity of mature osteoclasts . This effect appears to involve the production of novel phosphotyrosine proteins in osteoclast precursor cells . IL-10 also stimulates a novel inducible nitric oxide synthase . IL-10-deficient mice have decreased alveolar bone and decreased indices of osteoblast differentiation . A polymorphism in the IL-10 gene has been linked to postmenopausal osteoporosis .


4-1BB is an inducible T-cell costimulatory molecule, which interacts with 4-1BB ligand. In vitro treatment of RANKL-stimulated osteoclast precursor cells with 4-1BB ligand enhanced IL-10 production. In addition, expression of IL-10 was greater in RANKL-stimulated wild type osteoclast precursor cell cultures than in cultured cells from 41-BB-deficient mice . These results imply that some effects of IL-10 on osteoclasts may be mediated through the interactions of 4-1BB with 4-1BB ligand.



Interleukin-12, interleukin-23, interleukin-27, and interleukin-35


IL-12 is a cytokine that is produced by myeloid and other cell types. It induces T H 1 differentiation in T-lymphocytes and the subsequent expression of INF γ . Most authors have found that IL-12 has an inhibitory effect on osteoclastogenesis. However, the mechanisms by which this effect occurs in vitro are controversial. Some authors demonstrated direct inhibitory effects of IL-12 on RANKL-stimulated osteoclastogenesis in purified primary osteoclast precursors and RAW 264.7 cells . This was associated with the inhibition of NFATc1 expression in the osteoclast precursor cells. Interestingly, the inhibitory effects of IL-12 on osteoclastogenesis were absent in cells that were pretreated with RANKL . In contrast, others found that the inhibitory effects of IL-12 on osteoclastogenesis are indirect. One group demonstrated that the inhibitory effects of IL-12 are mediated by T-lymphocytes and do not involve production of INF γ . A second group disputes this result and found inhibition of osteoclastogenesis by IL-12 in cells from T-lymphocyte depleted cultures and cells from T-lymphocyte-deficient nude mice . The latter authors also demonstrated that antibody neutralization of INF γ blocked some of the inhibitory effect of IL-12 on RANKL-stimulated osteoclast formation. In contrast to the majority of reports that IL-12 is an inhibitor of osteoclastogenesis, one group demonstrated that IL-12 induced RANKL expression in human periodontal ligament cells .


The effects of IL-12 on TNFα-induced osteoclastogenesis have been examined in vivo . It was found that osteoclastogenesis, which was stimulated by the injection of TNFα over the calvaria of mice, was decreased when the mice were also treated with IL-12. Furthermore, this effect was not altered by antibody neutralization of T-lymphocytes in the mice. Induction of Fas by TNFα and FasL by IL12 in bone was critical for this response


IL-23 is an IL-12-related cytokine composed of one subunit of p40, which it shares with IL-12, and one subunit of p19, which is unique . It is critical for the differentiation of the T H 17 subset of T-lymphocytes along with TNFβ (TGFβ) and IL-6 . IL-23 appears most important for expanding the population of T H 17 T-lymphocytes. This subset of T-lymphocytes, which produces RANKL, has a high osteoclastogenic potential, which is mediated by their production of IL-17 . In an LPS-induced model of inflammatory bone destruction, it was found that there was markedly less bone loss in mice that were deficient either in IL-17 or IL-23 . Hence, the production of both is involved in the bone loss in this model. IL-23 induces RANKL expression in CD4 T-lymphocytes and RANK expression in osteoclast precursor cells . However, the actions of IL-23 on bone in vivo are controversial. IL-23-deficient mice have decreased bone mass in one report but increased bone mass in another . In some studies, IL-23 inhibited osteoclastogenesis through actions that were mediated by CD4 T-lymphocytes . In another study, IL-23 stimulated osteoclastogenesis in mixed osteoblast–osteoclast precursor cultures . One study identified upregulation of leukotriene B4 as a mechanism by which IL-23 regulated osteoclasts .


Curiously, individual neutralization of either IL-17 or IL-23 was found to be more efficacious than their combination as an inhibitor of ovariectomy-induced bone loss . IL-23 has also been identified as a critical mediator of the actions of ankylosing spondylitis on bone .


Another IL-12-related cytokine, IL-27, was found to have inhibitory effects on osteoclastogenesis in murine bone marrow cultures that were mediated by T-lymphocytes . However, the direct inhibitory effects of IL-27 on RANKL-stimulated osteoclastogenesis were identified as being mediated by inhibition of c-Fos . Recent data have implicated INF γ as a mediator of IL-27’s inhibitory effect on osteoclastogenesis , and IL-27 can also inhibit RANKL expression in CD4 T-lymphocytes . In osteoblasts, IL-27 inhibited apoptosis through a mechanism dependent on induction of early growth response-2 gene . Bone cells can be a source of IL-27 during inflammation .


IL-35, the most recently discovered IL-12 family member, is a direct inhibitor of osteoclastogenesis .



Interleukin-15


IL-15, like IL-7, is a member of the IL-2 superfamily and shares many activities with IL-2 including the ability to stimulate lymphocytes. It has been shown to enhance osteoclast progenitor cell number in culture . Its receptor is composed of a unique IL-15 receptor α and the β and γ chains of the IL-2 receptor . Deletion of IL-15 receptor α in mice produced a phenotype of decreased bone mineralization . IL-15 production by T-lymphocytes has been linked to the increased osteoclastogenesis and bone destruction seen in rheumatoid arthritis . In animal models of inflammatory bowel disease and staphylococcus aureus sepsis, lack of IL-15 or treatment with an IL-15 inhibitor reduced bone loss and the severity of the disease . In vitro, IL-15 treatment of mixed murine bone marrow and osteoblast cocultures demonstrated a role of natural killer (NK) cells in the ability of IL-15 to induce apoptosis in osteoblasts . Polymorphisms of the IL-15 gene have also been linked to variations in bone mineral density in women . In rheumatoid arthritis, IL-15 is reported to promote osteoclastogenesis via a pathway that is dependent on phospholipase D1 .



Interleukin-17 and interleukin-25


IL-17 is a family of related cytokines, which are unique and contain at least six members (A–F) IL-17E is also called IL-25 . These cytokines are central for the development of the adaptive immune response and the products of a subset of CD4 T-lymphocytes with a unique cytokine expression profile, termed T H 17. This contrasts with the more established T-lymphocyte cytokine-expressing subsets T H 1 and T H 2.


IL-17A was initially identified as a stimulator of osteoclastogenesis in mixed cultures of mouse hematopoietic cells and osteoblasts . This enhanced resorptive activity was mediated through the increased production of prostaglandin and RANKL . The direct effects of IL-17A on the differentiation of osteoclast precursor cells is controversial with some investigators finding stimulatory effects and others finding it to be inhibitory . One report found that low levels of IL-17 regulated osteoclast precursor autophagy . In another report, IL-17 stimulated rheumatoid synoviocytes to produce RANKL only with 1,25 dihydroxyvitamin D 3 and prostaglandin E 2 . The production of IL-17A in rheumatoid arthritis appears to be involved in the production of activated osteoclasts and bone destruction in involved joints . Effects of IL-17 on osteoclastogenesis and bone resorption are enhanced by TNFα, which is also produced in the inflamed joints of patients with rheumatoid arthritis . Inhibition of IL-17A in an antigen-induced arthritis model reduced the joint and bone destruction that is typically seen and decreased the production of RANKL, IL-1 β, and TNFα in the involved lesions . Multiple reports have now implicated IL-17 as a critical mediator of the bone loss that occurs in animal models after estrogen withdrawal . One of these suggested that studies of IL-17 neutralization be initiated to determine its role as a potential therapy to reverse postmenopausal bone loss in humans and to enhance bone regeneration after fracture . Data also implicate T-cell IL-17 production in the ability of PTH to stimulate bone resorption .


The effects of IL-17 on bone formation are complex. It stimulates mesenchymal cell proliferation and the expression of genes associated with early stages of osteoblast differentiation , an effect that may be mediated by the production of IL-17 by γδ T-cells . However, it inhibits mature osteoblast differentiation in vitro and the reparative response to a calvarial critical size defect in vivo , possibly through effects on Wnt signaling . Paradoxically, one study found that it augmented the osteogenic response to BMP-2 .



Interleukin-18, interleukin-33, and interleukin-37


IL-18 is similar to IL-1 in its structure and a member of the IL-1 superfamily . IL-18 synergizes with IL-12 to induce INF γ production , and its levels are increased at sites of inflammation such as rheumatoid arthritis . Osteoblastic cells express IL-18, and its production is induced by treatment with endothelin-1 . IL-18 inhibits osteoclast formation through a variety of mechanisms. These include its ability to stimulate GM-CSF , which is produced by T-cells in response to IL-18 treatment . It also stimulates INF γ production in vivo in bone , and its inhibitory effects on osteoclastogenesis and bone resorption are enhanced by cotreatment with IL-12 . IL-18 has been shown to indirectly stimulate osteoclastogenesis through its effects on T-lymphocytes . Finally, IL-18 is reported to increase production of OPG . In IL-18 overexpressing transgenic mice, osteoclasts were decreased, although, curiously, so was bone mass. These results indicate that there also may be effects of IL-18 on bone growth . In confirmation of this hypothesis, it was demonstrated that PTH treatment of osteoblasts stimulated IL-18 production. In addition, the anabolic effect of intermittent PTH treatment on trabecular bone mass in IL-18-deficient mice was reduced . IL-18 is also a mitogen for osteoblastic cells in vitro .


IL-18 binding protein (IL-18BP) is an antagonist of IL-18 action with antiinflammatory actions. Treatment of ovariectomized mice with IL1-BP prevented bone loss. It was also found that osteoporotic women had decreased IL-18BP levels and increased the amounts of serum IL-18 .


IL-33 is another member of the IL-1 family that has primarily been studied for its effects on T-lymphocytes . Its specific receptor is the orphan IL-1 receptor ST2 (also called IL-1R-like-1) . IL-33 is expressed by osteoblasts and production in these cells is stimulated by PTH and OSM . Its effects on bone cells are varied. One report found it to stimulate osteoclastogenesis , while multiple others have found it to be inhibitory ( Table 33.1 ) . In addition, there is a report that it had no effects on bone remodeling although these same authors suggested that it may be involved in the development of osteonecrosis of the femoral head caused by inadequate blood supply . The activation of HIF1α in osteoblasts resulted in increased IL-33 production . Osteoclastogenesis was decreased in transgenic mice that overexpress IL-33 in osteoblasts, . Mice deficient in IL-33 or ST2 had decreased bone mass . Interestingly, loss of ST2 in mice prevented ovariectomy-induced bone loss in the maxilla but not in the femur .



Table 33.1

Effects of major cytokines and cytokine receptors on bone cells.


































































































































Cytokine Receptor Actions on bone cells
RANKL (TNFSF11) RANK (TNFSFR11A), OPG (TNFSFR11B) RANKL is the principal stimulator of osteoclast formation and osteoclast-mediated bone resorption.
OPG is a soluble decoy receptor for RANKL that inhibits its interaction with the cellular receptor RANK.
CSF-1 (M-CSF) CSF-1R (c-fms) CSF-1 stimulates the proliferation of the osteoclast precursor and together with RANKL stimulates osteoclastogenesis and bone resorption.
IL-34 CSF-1R (c-fms) IL-34 can replicate the actions of CSF-1 on osteoclast precursors and mature osteoclasts
GM-CSF GM-CSFR In early myeloid precursors GM-CSF inhibits RANKL-mediated osteoclastogenesis while stimulating their differentiation into dendritic cells. In more mature osteoclast precursors, it has stimulatory effects on maturation and resorptive activity.
IL-1 α and β IL-1R1, IL-1R2 IL-1 is a major proinflammatory cytokine that stimulates osteoclastogenesis and bone resorption through a variety of both direct and indirect actions on osteoclasts and their precursors. IL-1 also inhibits osteoblast-mediated bone formation. IL-1R1 is the major cellular receptor. IL-1R2 is a cell membrane decoy receptor.
TNF α and β TNFR1, TNFR2 Like IL-1, TNF is a major proinflammatory cytokine that stimulates osteoclastogenesis and bone resorption through a variety of both direct and indirect actions on osteoclasts and their precursors. In addition, it is an inhibitor of osteoblasts and bone formation. Both TNF receptors are active. Although, most of the effects of TNF on bone cells are mediated by TNFR1.
Fas-ligand Fas Inhibits osteoblast differentiation and stimulates osteoclastogenesis.
TRAIL TRAIL-R2 Effects on osteoclastogenesis are variable.
CD40-ligand CD40 Accelerate RANKL-induced osteoclastogenesis.
IL-4 IL-4R Inhibits osteoclast and osteoblast activity.
IL-6 IL-6R, gp130 Variable effects on bone resorption and formation.
IL-11 IL-11R, gb130 Stimulates osteoclastogenesis and osteoblast differentiation.
LIF LIFR, gp130 Variable effects on resorption and formation.
Oncostatin-M OSMR, LIFR, gp130 Inhibits osteoclasts formation and resorption. Stimulates osteoblast differentiation
IL-7 IL-7R, common γ chain Variable effects on bone resorption and formation.
IL-8 CXCR1, CXCR2 Stimulates bone resorption.
CCL2 (MCP-1) CCR2, CCR4 Stimulates bone resorption.
CCL3 (MIP-1α) CCR1 CCR5 Stimulates osteoclastogenesis and bone resorption, inhibits bone formation.
CCL9 (MIP-1γ) CCR1 Stimulates osteoclastogenesis and bone resorption.
CXCL12 CXCR4 Stimulates osteoclast precursor cell migration and osteoclastogenesis.
CX3CL1 CX3CR1 Stimulates osteoclast precursor cell migration and osteoclastogenesis.
IL-10 IL-10R Inhibits osteoclastogenesis and osteoblastogenesis.
IL-12 IL-12R Inhibits osteoclastogenesis.
IL-13 IL-13R Inhibits osteoclast and osteoblast activity.
IL-15 IL-15R Stimulates osteoclastogenesis.
IL-17 (A–F) IL-17R Variable effects on osteoclastogenesis.
IL-18 IL-18R Inhibits osteoclastogenesis, variable effects on osteoblasts.
INF-α and β (type 1) IFNAR1 Inhibits osteoclasts and osteoblasts.
INF-γ (type II) IFNGR1, IFNGR2 Variable effects on osteoclasts and osteoblasts.
MIF CD74 Variable effects, including direct inhibition of osteoclastogenesis.

CCL , C-C motif chemokine ligand; CSF-1 , colony-stimulating factor 1; CSF-1R , colony-stimulating factor-1 receptor; CXCR , C-X-C motif chemokine receptor; GM-CSF , granulocyte macrophage colony-stimulating factor; IL , interleukin; LIF , leukemia inhibitory factor; M-CSF , macrophage colony-stimulating factor; MCP-1 , monocyte chemotactic protein 1; MIF , migration inhibitory factor; OPG , osteoprotegerin; RANK , receptor activator of nuclear factor-κB; RANKL , receptor activator of nuclear factor-κB ligand; TNF , tumor necrosis factor; TNFSF11 , tumor necrosis factor superfamily ligand 11; TRAIL , tumor necrosis factor–related apoptosis inducing ligand.


IL-37 is also an IL-1 family member with osteoclast inhibitory activity . Serum levels of IL-37 are increased in patients with rheumatoid arthritis and ankylosing spondylitis and correlate with disease activity .



Interferons


Interferons (INF) γ is a type II interferon with a wide variety of biologic activities. In vitro, INF γ has generally been found to have inhibitory actions on bone resorption . These appear to be direct and are mediated by its effects on osteoclast progenitor cells. INF γ inhibits the ability of 1,25-dihydroxyvitamin D 3 , PTH and IL-1 to stimulate the formation of osteoclast-like cells in cultures of human bone marrow . INF γ also inhibits RANK signaling by accelerating the degradation of TRAF6 through the activation of the ubiquitin/proteasome system , by inhibiting NFATc1 expression and by activating the NF-κB and JNK pathways . Curiously, it is reported to not inhibit resorption in mature osteoclasts . However, INF γ is also reported to have stimulatory effects on resorption through its ability to increase RANKL and TNF-α production in T-lymphocytes and through its ability to enhance the fusion of preosteoclasts . It also appears to mediate the ability of γδ T-lymphocytes and IL-27 to inhibit osteoclastogenesis and resorptive activity .


In osteoblasts INF γ is an inhibitor of proliferation and has variable effects on differentiation .


The effects of INF γ on bone in vivo are also variable, as both inhibitory and stimulatory effects have been reported. In mice with collagen-induced arthritis, loss of the INF γ receptor (INF γR) leads to increased bone destruction . Similarly, in mice that are injected over their calvaria with bacterial endotoxin, which activates toll-like receptors (TLRs), loss of INF γR resulted in an enhanced resorptive response . This result is consistent with more recent findings demonstrating that the inhibitory effects of INF γ on osteoclastogenesis are enhanced by activation of TLRs . The ability of M1 macrophages and Foxp3+CD8 T-cells to inhibit osteoclastogenesis was dependent on their production of INF γ . Finally, in mice that underwent ovariectomy to induce estrogen withdrawal, the administration of INF γ enhanced bone mass and prevented the development of the bone loss that is otherwise seen in this condition .


In contrast, intraperitoneal injection of INF γ for 8 days in rats induced osteopenia . In patients who have osteopetrosis, because they produce defective osteoclasts, administration of INF γ stimulated bone resorption and appeared to partially reverse the disease . The latter effects are possibly due to the ability of INF γ to stimulate osteoclast superoxide synthesis , osteoclast formation in vivo , or a generalized immune response .


Type I interferons (INF α and INF β) are typically produced in response to invading pathogens . Mice deficient in the INF α/β receptor component interferon alpha and beta receptor subunit 1 (IFNAR1) have reduced trabecular bone mass and an increased number of osteoclasts . RANKL induces INF β in osteoclasts, and INF β, in turn, inhibits RANKL-mediated osteoclastogenesis by decreasing c-fos expression and inducing the production of miR-155 . Osteocytes are a source of INF α . INF β has also been shown to inhibit bone resorption in vitro although its mechanism of action is not as well studied as that of INF γ and α . In vivo, INF α had no effect on bone turnover .



Additional cytokines


IL-4 and IL-13 are members of a group of locally acting factors that have been termed “inhibitory cytokines.” The effects of IL-4 and IL-13 seem related and appear to affect both osteoblasts and osteoclasts. Transgenic mice that overexpress IL-4 had an osteoporotic phenotype . This effect may result from both an inhibition of osteoclast formation and activity and an inhibition of bone formation . IL-13 and IL-4 inhibited IL-1-stimulated bone resorption by decreasing the production of prostaglandins and the activity of cyclooxygenase-2 . The direct inhibitory effects of IL-4 on osteoclast precursor cell maturation are more potent than that of IL-13 and involve effects on STAT6, NF-κB, peroxisome proliferator-activated receptor γ1, mitogen-activated protein kinase signaling, Ca ++ signaling, NFATc1, and c-Fos . IL-4 along with GM-CSF induces multipotential myeloid cell differentiation toward the dendritic cell lineage and away from the osteoclast lineage .


IL-13 and IL-4 induce cell migration (chemotaxis) in osteoblastic cells , and they regulate the ability of osteoblasts and vascular endothelial cells to control OPG and RANKL production .


IL-32 is a cytokine that is involved in innate and adoptive immunity. It is produced by T-lymphocytes, NK cells and epithelial cells and has six different splice variants . IL-32 stimulated the formation of multinuclear cells that were TRAP- and vitronectin receptor-positive but did not resorb. In addition, it inhibited resorption that was stimulated by RANKL . IL-32 and IL-17 can reciprocally stimulate each other in inflamed synovium and influence osteoclastic resorption . In osteoblasts IL-32γ stimulated bone formation through a mechanism dependent on the miR, miR-29a .


Macrophage migration inhibitory factor (MIF) was initially identified as an activity in conditioned medium from activated T-lymphocytes that inhibited macrophage migration in capillary tube assays . Once purified and cloned , it became available for functional studies and was shown to have a variety of activities. In addition to T-lymphocytes, it is produced by pituitary cells and activated macrophages. MIF is a direct inhibitor of osteoclastogenesis in vitro through its ability to activate Lyn tyrosine kinase .


In vivo, MIF’s effects are complex. Mice that overexpress MIF globally have high turnover osteoporosis while, curiously, mice deficient in MIF have low turnover osteoporosis with decreased serum indices of bone resorption and bone formation . MIF-deficient mice are also reported to not lose bone mass or increase osteoblast or osteoclast number in bone with ovariectomy . Hence, MIF may be another mediator of the effects that estrogen withdrawal has on bone. Estrogen downregulates MIF expression in activated macrophages . Hence, a similar response may occur in bone or bone marrow and mediate some of the effects that ovariectomy has on bone mass. MIF also mediates the homing of osteoclast precursor cells to osteolytic sites .


MIF is made by osteoblasts , and its production in these cells was upregulated by a variety of factors including TGF-β, FGF-2, IGF-II, and fetal calf serum . In vitro, MIF increased MMP9 and MMP13 expression in osteoblasts and inhibited RANKL-stimulated osteoclastogenesis by decreasing the fusion of precursors, possibly through its ability to inhibit the migration of these cells .


Deletion of CD74, a putative MIF receptor, in mice produced a phenotype of enhanced osteoclastogenesis and decreased bone mass . In human studies a polymorphism in the MIF gene has been linked to osteoporosis , and it may mediate some of the pathology of ankylosing spondylosis .



References




  • 1. Suda T., Takahashi N., Udagawa N., Jimi E., Gillespie M.T., Martin T.J.: Modulation of osteoclast differentiation and function by the new members of the tumor necrosis factor receptor and ligand families. Endocr Rev 1999; 20: pp. 345-357.



  • 2. Teitelbaum S.L.: Bone resorption by osteoclasts. Science 2000; 289: pp. 1504-1508.



  • 3. Walsh M.C., Kim N., Kadono Y., Rho J., Lee S.Y., Lorenzo J., et. al.: Osteoimmunology: interplay between the immune system and bone metabolism. Annu Rev Immunol 2006; 24: pp. 33-63.



  • 4. Fuller K., Wong B., Fox S., Choi Y., Chambers T.J.: TRANCE is necessary and sufficient for osteoblast-mediated activation of bone resorption in osteoclasts. J Exp Med 1998; 188: pp. 997-1001.



  • 5. Lacey D.L., Timms E., Tan H.L., Kelley M.J., Dunstan C.R., Burgess T., et. al.: Osteoprotegerin ligand is a cytokine that regulates osteoclast differentiation and activation. Cell 1998; 93: pp. 165-176.



  • 6. Yasuda H., Shima N., Nakagawa N., Yamaguchi K., Kinosaki M., Mochizuki S., et. al.: Osteoclast differentiation factor is a ligand for osteoprotegerin osteoclastogenesis-inhibitory factor and is identical to TRANCE/RANKL. Proc Natl Acad Sci USA 1998; 95: pp. 3597-3602.



  • 7. Kong Y.Y., Yoshida H., Sarosi I., Tan H.L., Timms E., Capparelli C., et. al.: OPGL is a key regulator of osteoclastogenesis, lymphocyte development and lymph-node organogenesis. Nature 1999; 397: pp. 315-323.



  • 8. Nakashima T., Hayashi M., Fukunaga T., Kurata K., Oh-Hora M., Feng J.Q., et. al.: Evidence for osteocyte regulation of bone homeostasis through RANKL expression. Nat Med 2011; 17: pp. 1231-1234.



  • 9. Xiong J., Onal M., Jilka R.L., Weinstein R.S., Manolagas S.C., O’Brien C.A.: Matrix-embedded cells control osteoclast formation. Nat Med 2011; 17: pp. 1235-1241.



  • 10. Onal M., Xiong J., Chen X., Thostenson J.D., Almeida M., Manolagas S.C., et. al.: RANKL expression by B lymphocytes contributes to ovariectomy-induced bone loss. J Biol Chem 2012; 287:



  • 11. Titanji K., Vunnava A., Foster A., Sheth A.N., Lennox J.L., Knezevic A., et. al.: T-cell receptor activator of nuclear factor-kappaB ligand/osteoprotegerin imbalance is associated with HIV-induced bone loss in patients with higher CD4+ T-cell counts. AIDS (London, Engl) 2018; 32: pp. 885-894.



  • 12. Xiong J., Piemontese M., Thostenson J.D., Weinstein R.S., Manolagas S.C., O’Brien C.A.: Osteocyte-derived RANKL is a critical mediator of the increased bone resorption caused by dietary calcium deficiency. Bone 2014; 66C: pp. 146-154.



  • 13. Zhang S., Wang X., Li G., Chong Y., Zhang J., Guo X., et. al.: Osteoclast regulation of osteoblasts via RANKRANKL reverse signal transduction in vitro. Mol Med Rep 2017; 16: pp. 3994-4000.



  • 14. Ikebuchi Y., Aoki S., Honma M., Hayashi M., Sugamori Y., Khan M., et. al.: Coupling of bone resorption and formation by RANKL reverse signalling. Nature 2018; 561: pp. 195-200.



  • 15. Nakashima T., Kobayashi Y., Yamasaki S., Kawakami A., Eguchi K., Sasaki H., et. al.: Protein expression and functional difference of membrane-bound and soluble receptor activator of NF-kappaB ligand: modulation of the expression by osteotropic factors and cytokines. [In Process Citation] Biochem Biophys Res Commun 2000; 275: pp. 768-775.



  • 16. Xiong J., Cawley K., Piemontese M., Fujiwara Y., Zhao H., Goellner J.J., et. al.: Soluble RANKL contributes to osteoclast formation in adult mice but not ovariectomy-induced bone loss. Nat Commun 2018; 9: pp. 2909.



  • 17. Boyle W.J., Simonet W.S., Lacey D.L.: Osteoclast differentiation and activation. Nature 2003; 423: pp. 337-342.



  • 18. Hikita A., Yana I., Wakeyama H., Nakamura M., Kadono Y., Oshima Y., et. al.: Negative regulation of osteoclastogenesis by ectodomain shedding of receptor activator of NF-kappaB ligand. J Biol Chem 2006; 281: pp. 36846-36855.



  • 19. Simonet W.S., Lacey D.L., Dunstan C.R., Kelley M., Chang M.S., Luthy R., et. al.: Osteoprotegerin: a novel secreted protein involved in the regulation of bone density. Cell 1997; 89: pp. 309-319.



  • 20. Tsuda E., Goto M., Mochizuki S., Yano K., Kobayashi F., Morinaga T., et. al.: Isolation of a novel cytokine from human fibroblasts that specifically inhibits osteoclastogenesis. Biochem Biophys Res Commun 1997; 234: pp. 137-142.



  • 21. Yun T.J., Chaudhary P.M., Shu G.L., Frazer J.K., Ewings M.K., Schwartz S.M., et. al.: OPG/FDCR-1, a TNF receptor family member, is expressed in lymphoid cells and is up-regulated by ligating CD40. J Immunol 1998; 161: pp. 6113-6121.



  • 22. Emery J.G., McDonnell P., Burke M.B., Deen K.C., Lyn S., Silverman C., et. al.: Osteoprotegerin is a receptor for the cytotoxic ligand TRAIL. [In Process Citation] J Biol Chem 1998; 273: pp. 14363-14367.



  • 23. Bucay N., Sarosi I., Dunstan C.R., Morony S., Tarpley J., Capparelli C., et. al.: Osteoprotegerin-deficient mice develop early onset osteoporosis and arterial calcification. Genes Dev 1998; 12: pp. 1260-1268.



  • 24. Mizuno A., Amizuka N., Irie K., Murakami A., Fujise N., Kanno T., et. al.: Severe osteoporosis in mice lacking osteoclastogenesis inhibitory factor/osteoprotegerin. [In Process Citation] Biochem Biophys Res Commun 1998; 247: pp. 610-615.



  • 25. Coulson J., Bagley L., Barnouin Y., Bradburn S., Butler-Browne G., Gapeyeva H., et. al.: Circulating levels of dickkopf-1, osteoprotegerin and sclerostin are higher in old compared with young men and women and positively associated with whole-body bone mineral density in older adults. Osteoporos Int 2017; 28: pp. 2683-2689.



  • 26. Hauser B., Zhao S., Visconti M.R., Riches P.L., Fraser W.D., Piec I., et. al.: Autoantibodies to osteoprotegerin are associated with low hip bone mineral density and history of fractures in axial spondyloarthritis: a cross-sectional observational study. Calcif Tissue Int 2017; 101: pp. 375-383.



  • 27. Whyte M.P., Obrecht S.E., Finnegan P.M., Jones J.L., Podgornik M.N., McAlister W.H., et. al.: Osteoprotegerin deficiency and juvenile Paget’s disease. N Engl J Med 2002; 347: pp. 175-184.



  • 28. Piemontese M., Xiong J., Fujiwara Y., Thostenson J.D., O’Brien C.A.: Cortical bone loss caused by glucocorticoid excess requires RANKL production by osteocytes and is associated with reduced OPG expression in mice. Am J Physiol Endocrinol Metab 2016; 311: pp. E587-E593.



  • 29. Hofbauer L.C., Khosla S., Dunstan C.R., Lacey D.L., Spelsberg T.C., Riggs B.L.: Estrogen stimulates gene expression and protein production of osteoprotegerin in human osteoblastic cells. Endocrinology 1999; 140: pp. 4367-4370.



  • 30. Anderson D.M., Maraskovsky E., Billingsley W.L., Dougall W.C., Tometsko M.E., Roux E.R., et. al.: A homologue of the TNF receptor and its ligand enhance T-cell growth and dendritic-cell function. Nature 1997; 390: pp. 175-179.



  • 31. Dougall W.C., Glaccum M., Charrier K., Rohrbach K., Brasel K., De Smedt T., et. al.: RANK is essential for osteoclast and lymph node development. Genes Dev 1999; 13: pp. 2412-2424.



  • 32. Cecchini M.G., Hofstetter W., Halasy J., Wetterwald A., Felix R.: Role of CSF-1 in bone and bone marrow development. Mol Reprod Dev 1997; 46: pp. 75-84.



  • 33. Hayashi S., Miyamoto A., Yamane T., Kataoka H., Ogawa M., Sugawara S., et. al.: Osteoclast precursors in bone marrow and peritoneal cavity. J Cell Physiol 1997; 170: pp. 241-247.

    Only gold members can continue reading. Log In or Register to continue

    Stay updated, free articles. Join our Telegram channel

Oct 27, 2020 | Posted by in ENDOCRINOLOGY | Comments Off on Cytokines and the pathogenesis of osteoporosis

Full access? Get Clinical Tree

Get Clinical Tree app for offline access