Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2023 Nov 19.
Published in final edited form as: Bone. 2022 Aug 27;164:116540. doi: 10.1016/j.bone.2022.116540

Cytokine-mediated immunomodulation of osteoclastogenesis

Pengcheng Zhou a,e,1,*, Ting Zheng b,1, Baohong Zhao b,c,d
PMCID: PMC10657632  NIHMSID: NIHMS1864270  PMID: 36031187

Abstract

Cytokines are an important set of proteins regulating bone homeostasis. In inflammation induced bone resorption, cytokines, such as RANKL, TNF-α, M-CSF, are indispensable for the differentiation and activation of resorption-driving osteoclasts, the process we know as osteoclastogenesis. On the other hand, immune system produces a number of regulatory cytokines, including IL-4, IL-10 and IFNs, and limits excessive activation of osteoclastogenesis and bone loss during inflammation. These unique properties make cytokines powerful targets as rheostat to maintain bone homeostasis and for potential immunotherapies of inflammatory bone diseases. In this review, we summarize recent advances in cytokine-mediated regulation of osteoclastogenesis and provide insights of potential translational impact of bench-side research into clinical treatment of bone disease.

Keywords: Osteoclastogenesis, Cytokines, Immunomodulation, Bone loss

1. Introduction

Bone homeostasis is critical for health and disease. As an organ harboring a variety type of cells, bone is continuously remodeling through the dynamic equilibrium between osteoclasts, responsible for bone resorption, and osteoblasts/osteocytes, responsible for bone formation. This process is tightly regulated by local and systemic factors, including the body's innate and adaptive immune cells, through direct interaction and via intermediate cellular response. Interrupted bone and skeletal homeostasis often lead to diseases, such as osteoporosis, osteoarthritis (OA), rheumatoid arthritis (RA) and Paget disease, which immensely affect the quality of life and increase the risk of mortality in patients. Notably, osteoporosis is one of the most common degenerative diseases in middle-aged and elderly people, causing a vast health burden worldwide. Characterized by significant bone loss and deterioration of skeletal microstructure, osteoporosis results in weak bone loading function and a significantly increased risk of bone fracture. Nearly one third of women and 1/5 men aged over 50 are suffering from or have a higher chance of experiencing fragility fracture. These account for almost 9 million fragility fractures that occur each year, involving nearly 1.6 million hip fractures, which can cause life-threatening complications [1].

The crosstalk between skeletal and immune systems is essential for the onset and development of osteoporosis [2]. Indeed, osteoimmunology, a concept coined in 2000, has revolutionized our understanding of the osteoporosis pathophysiology and demonstrated a key role of inflammatory cytokines in influencing the fine-tuned balance between bone formation and resorption [3,4]. Simply put, excessive bone resorption caused by hyperactivated osteoclasts (OC) impairs the normal bone remodeling which laid on the foundation of the balance between osteoclasts and osteoblasts, resulting in the significant bone loss in osteoporosis [5]. These osteoclasts are originated from the myeloid cell progenitors, particularly monocytes and macrophages, which are part of innate immune cells (Fig. 1). During osteoclast differentiation, or termed as osteoclastogenesis, myeloid cells receive signals guided by key cytokines such as receptor activator of nuclear factor kappa-B ligand (RANKL), and undergo transcriptional alterations with the ultimate fate as the multinucleated mature osteoclast cells. Adaptive immune cells, mainly T cells, either promote this process or inhibit it from osteoclastic onset of differentiation. Activated T cells are one of the major sources of receptor activator of nuclear factor kappa-B ligand (RANKL) that is essential for osteoclastogenesis during tissue inflammation. Conversely, regulatory T (TREG) cells inhibit osteoclastogenesis and subsequently reduce bone loss through either contact-directed or intermediate cytokine-modulated regulation, or a combination of both [6,7]. These indispensable connections between skeletal and innate and adaptive immune systems provide a unique perspective to understand the biology of bone remolding, which ultimately allows us to modulate osteoclastogenesis by immunomodulation to treat bone diseases.

Fig. 1. Immune cells produce cytokines to modulate osteoclastogenesis.

Fig. 1.

Both innate and adaptive immune cells regulate osteoclastogenesis by production of cytokines. TH17 cells produce RANKL and IL-17 during bone inflammation in diseases such as osteoporosis and rheumatoid arthritis. Macrophages are also the major source of proinflammatory cytokines such as IL-1β and TNF-α. Cytokines produced by these immune cells markedly enhanced the activation of osteoclastogenesis. On the other hand, regulatory T (TREG) cells, TH2 cells and innate lymphoid cells are counterpart of this process and negatively regulate osteoclastogenesis by producing cytokines such as IL-10, IL-4, IL-5 and IL-13. These immune cells in together, maintain the dynamics of bone remodeling and are key immunomodulators to treat inflammatory bone diseases.

Cytokines are a broad range of secreted proteins necessary for cell signaling. Through the interaction with cell surface receptors, cytokines initiate complex downstream signaling cascades that can direct cell proliferation, survival, differentiation and metabolism. Immune cells are one of the major sources of cytokines. As important bone-affecting factors, cytokines maintain bone homeostasis by direct modulation of bone forming-osteoblasts and bone resorbing-osteoclasts. The excess or insufficiency of key cytokines, for instance RANKL, tumor necrosis factor alpha (TNF-α), and IL-6, are often considered as major immune-related factors causing inflammatory bone diseases. For example, in osteoporosis, a number of proinflammatory cytokines, such as RANKL, IL-1β, TNF-α, IL-6 and IL-17, are increased, which rapidly promotes osteoclast formation. On the other hand, the presence of regulatory cytokine IL-10, which negatively regulates OC development, is often declined in osteoporosis (Table 1). These abnormalities, together, tip the balance towards the inflammatory bone loss and the severe illness of osteoporosis. To be noted, most of these pro-inflammatory cytokines were also increased in patients infected with SARS-CoV-2 [8,9]. Therefore, further understanding of cytokines' role in regulating bone remodeling also enables us to adapt to the fast-ever-changing global health situation in response to COVID-19 and have necessary knowledge available to prevent and treat potential infection induced bone loss. Cytokines are also implemented in clinical diagnosis or evaluation of treatment efficacy in patients with acute or chronic bone disorders. More importantly, beyond diagnosis, emerging progress has been made in developing novel immuno-therapeutics either straightforwardly using cytokines as an immunologic agent or targeting them to restore the balance between bone formation and bone resorption [10]. These potential cytokine-based immunotherapeutics can provide broader options in addition to the current treatments in patients with osteoporosis or broad bone disorders, which are mainly chemical or physical based approaches.

Table 1.

Cytokines in osteoclastogenesis.

Regulation Factor Main source Action Reference
Positive regulation RANKL Osteoblasts, osteocytes, TH17, ILC3 Induces osteoclastogenesis, survival, proliferation and fusion. [19-38]
M-CSF Osteoblasts, MSCs Supports osteoclast precursor survival, proliferation and fusion. [2,44-49]
IL-1β Monocytes, macrophages Induces osteoclastogenesis by RANK/RANKL signaling via TRAF6 and increases RANKL expression. [50-56]
IL-7 Stromal cells, DC Induces osteoclastogenesis by inducing STAT5 activation; enhance the production of TNF-α and RANKL by T cells. [82,83]
IL-8 Macrophages Induces osteoclastogenesis by inducing RANKL-induced NFATc1 activation. [84-89]
IL-15 Monocytes, macrophages Induces osteoclastogenesis by activation of RANK/RANKL signaling; induces RANKL expression. [92-97]
IL-17 TH17, ILC3 Induces RANKL, IL-1β, IL-6, IL-23 and TNF-α expression. [5,39,57-63]
TNF-α Macrophages, T/B/NK cells Induces M-CSF, RANKL and RANK expression. [48,69-80]
Negative regulation IL-4 TH2, ILC2 Inhibits osteoclastogenesis by affecting NF-kB activation in a STAT6-dependent manner or modulating Ca2+ signaling and decreases IL-1, IL-6, TNF-α expression. [27,28,102,106-109]
IL-5 TH2, ILC2 Inhibits osteoclastogenesis. [110]
IL-9 TH2, ILC2 Inhibits osteoclastogenesis. [100]
IL-10 Macrophages, DC, regulatory T/B cells Inhibits osteoclastogenesis by reducing NFATc1 expression and level of RANKL and M-CSF, and upregulating OPG expression; suppresses IL-1β and TNF-α expression. [6,7,116-120]
IL-12 Macrophages, monocytes, DC, B cells Inhibits osteoclastogenesis by inducing Fas/FasL or NO mediated apoptosis. [131-134]
IL-13 TH2, ILC2 Inhibits osteoclastogenesis. [28,101,108]
IL-27 Macrophages, monocytes, DC, B cells Inhibits osteoclastogenesis by suppressing NFATc1-AP-1-MAPK signaling pathway; inhibits STAT1 dependent c-Fos activation; downregulation of RANKL and TREM-1 expression. [135-137]
IFN-β Macrophages, DC Inhibits osteoclastogenesis by c-Fos dependent RANK signaling via TRAF6 and suppresses RANKL expression; increases CXCL11 expression. [26,126-129]
IFN-γ T cells, NK Inhibits osteoclastogenesis by RANK/RANKL signaling via TRAF6. [3,138-142]
Ambiguous regulation IL-6 Macrophages, monocytes Regulates RANK/RANKL signaling via JAK/STAT3 and increases RANKL expression;
Suppresses the osteoclast precursor differentiation via NF-kB signaling.
[145,146]
[147]
IL-23 Macrophages, monocytes, DC, B cells Promotes osteoclast formation by upregulation of RANK expression; activation of DAP12; induces RANKL and IL-17 production. [150-153]
Inhibits osteoclastogenesis by induction of GM-CSF. [154,155]
TGF-β1 Macrophages, monocytes, T/B/NK/DC cells Regulates osteoclast differentiation via SMAD2/3, which mediated by TRAP6-TAB1-TAK1 complex at low-concentration or early stage of differentiation [161-163,165,166]
Inhibits osteoclastogenesis through regulating RANKL/OPG ratio secreted by osteoblasts. [161,163,164,166]

TH2: Type 2 helper T cells, ILC2: Group 2 innate lymphoid cells; TH17: Type 17 helper T cells; ILC3: Group 3 innate lymphoid cells; NK: natural killer cells; DC: dendritic cells.

A key drawback of current medicines on osteoporosis is the severe adverse effect. The risk of potential side effects is particularly higher in those who require long-term clinical therapy [1]. To date, anti-resorptive drugs bisphosphonates and RANKL monoclonal antibody (Denosumab) are most used medications in clinical practice, taking advantages of their relative effectiveness, long-term tolerance and affordable price [11]. However, studies suggest that long-term use of these medicines might induce serious side effects, although rare, including atypical leg fractures, osteonecrosis of the jaw, and esophageal cancer, cardiovascular disorder, osteoarthritis, hypercalcemia and hypocalcemia [12]. Moreover, anabolic drugs parathyroid hormone (NATPARA) and sclerostin monoclonal antibody (Romosozumab) were approved by FDA as additional treatments for osteoporosis in postmenopausal women with high risk of fracture [13-17]. Notwithstanding, potential issues such as osteosarcoma, osteonecrosis and relatively high cost still exist, making these medications “second-line” treatments in patients with severe osteoporosis [13,15]. Moreover, these medications were developed to broadly turn down the osteoclastogenesis and activated immune response, which might induce immunosuppression with increased risk of cancer and opportunistic infection [18]. Therefore, additional and alternative novel clinical treatment options are greatly needed.

In this review, we focus on osteoimmunological cytokines and summarize the advances and current knowledge of cytokine-mediated immunomodulation of cellular and molecular process in osteoclastogenesis. In particular, we comprehensively review how cytokines regulate osteoclastogenesis, a key process for bone resorption. In addition, we will provide insights on how the current studies and their findings have the potential to translate into clinical design of novel immunotherapeutics, and how they might benefit the patients with bone-related disorders.

2. Positive regulation of osteoclastogenesis by cytokines

2.1. RANKL

The receptor activator of nuclear factor kappa-B ligand (RANKL) is a member of the tumor necrosis factor (TNF) superfamily of ligands and instrumental for the ultimate differentiation of osteoclast precursors into osteoclasts. RANKL is secreted or expressed by both immune cells such as type 17 helper T (TH17) cells and skeletal cells such as osteoblast and osteocytes. RANKL interacts with its receptor RANK on osteoclast surface, where trimerized RANK-RANKL complex recruits downstream adaptor molecules such as tumor necrosis factor (TNF) receptor-associated factor (TRAF) 6 to its cytoplasmic domain, resulting in the activation of multiple signaling pathways including IKK complexes (IKKα, IKKβ, IKKγ, and NIK-IKKα) and mitogen-activated protein kinases (MAPKs: extracellular signal-regulated kinase (ERK), p38, and c-Jun terminal kinase (JNK)) in premature osteoclasts [19,20]. Together with activated CaMKIV-CREB signaling pathway, RANKL-transduced signaling cascades promote AP-1 activation, followed by the robust induction of the key transcriptional factor of osteoclastogenesis, nuclear factor of activated T cells cytoplasmic 1 (NFATc1) (Fig. 2) [20-23]. Cytosolic calcium, which can be regulated by signaling receptors including FcγRIII, PIR-A, OSCAR, TREM2 and SIRPβ1, is also essential to activate NFATc1 [5] [24,25]. NFATc1 promotes the expression of Blimp1, which represses anti-osteoclastogenic genes, such as Mafb, BCL-6 and IRF8 [23]. In addition, STAT family members are also critical during the RANKL-mediated osteoclastogenesis. For instance, interferon-β negatively regulates osteoclast differentiation through the STAT1 pathway [19,26]. IL-4 abrogates osteoclastogenesis through STAT6-dependent suppression of NF-kappaB [27,28]. Moreover, STAT5 negatively regulates osteoclastogenesis, by suppressing Dusp1 and Dusp2, two key phosphatases in MAPK pathway [22]. Additionally, sialic acid-binding immunoglobulin-like lectins family molecule Siglec-15 was shown to regulate RANKL induced-osteoclastogenesis [29-31]. Deciphering how cytokines might affect these molecules during osteoclastogenesis could help us develop further treatments to intervene the OC formation.

Fig. 2. Critical signaling pathways activated by cytokines in osteoclasto-genesis.

Fig. 2.

RANKL binds to RANK which recruits TRAF6 to activate signaling pathways including MAPK, IKK and Ca+/CAMKIV. These signaling cascades ultimately induces the translocation and activation of key transcriptional factors such as NFATc1 that targets and initiates the expression of osteoclastogenic genes. Cytokines such as TNF-α, MCSF and IL-1β facilitate osteoclastogenesis by activating other critical signaling pathways such as JAK/STAT, PI3K, NF-κB to promote the maturation, differentiation and survival of osteoclasts.

RANKL not only stimulates the differentiation of osteoclast but also accelerates bone resorption by enhancing function of mature osteoclasts and prolonging their survival [32]. Indeed, mice with injected soluble RANKL showed increased measurable bone resorption within 60 min [33]. Notably, RANKL signaling can be potently suppressed through the negative feedback action coordinated by osteoprotegerin (OPG), a secreted receptor also belonging to the TNF receptor superfamily. OPG, largely produced by bone marrow mesenchymal stem cells (MSCs), osteoblast lineage cells, B cells and dendritic cells, functions as a decoy receptor for RANKL. OPG binds to RANKL to prevent its interaction with RANK receptor [34], thereby limiting osteoclastogenesis. RANKL/OPG ratio is therefore recognized as the one of the hallmarks for patients with osteoporosis and other various bone related diseases, including rheumatoid arthritis, Paget's disease, metastatic bone disease as well as glucocorticoid-induced bone loss [35,36]. In addition to the secretion of OPG, bone marrow MSCs also regulates the OC formation through the signaling involves Cyclin-dependent kinase 8 (CDK8), by which the alteration of CDK8 level resulted in changed STAT1 signaling downstream of RANKL induced osteoclastogenesis [37,38].

TH17 cells promote osteoclastogenesis in inflammation-induced osteoporosis and rheumatoid arthritis, amongst many RANKL-expressing activated CD4+ T cells [39]. This cellular regulation is elegantly coordinated by both secreted RANKL as well as surface expression of RANKL on TH17 cells that can lead to a short but potent synapse-like cell-cell interaction [40]. Importantly, IL-17, the signature cytokine produced by TH17 cells synergizes RANKL produced by TH17 cells and subsequently promotes osteoclastogenesis and bone resorption. Interestingly, a subset of CCR6+ innate lymphoid cells also expresses RANKL on cell surface [41]. Although lacking antigen specific receptors, these group 3 innate lymphoid (ILC3) cells act similarly to their T cell counterparts, TH17 cells, which produce IL-17 and IL-22 in response to IL-1b and IL-23 stimulation during tissue inflammation in gut, lung and skin. However, whether and how these RANKL expressing cells regulate osteoclastogenesis in bone microenvironment remains unaddressed.

2.2. M-CSF

M-CSF plays an important role during the early proliferation, survival and differentiation of osteoclast precursor cells [42,43]. Produced by various cell populations derived from mesenchymal cells, M-CSF binds to its receptor CD115 (c-Fms) and initiates the downstream signaling pathways including phosphoinositide 3 kinase (PI3K), phospholipase C gamma (PLCγ), as well as extracellular signal-regulated kinases 1 and 2 (ERK1/2) signaling (Fig. 2) [2]. These signaling pathways are indispensable for the development of osteoclast precursors during early differentiation.

Indeed, animal studies found that mice with depleted M-CSF receptor c-fms showed reduction in the number of osteoclasts which associated with increased risk of osteopetrosis [44]. Moreover, high level of M-CSF was often observed in pathological bone diseases including osteoporosis, inflammatory osteolysis and rheumatoid arthritis, along with an increased number of osteoclasts [45-47]. In an inflammatory osteolysis model, TNF-α induced-M-CSF production in TNF-responsive stromal cells contributed to the increased osteoclastogenesis [48]. On the other hand, blockade of M-CSF using monoclonal antibodies was able to markedly ameliorate inflammatory and arthritic pain in arthritic diseases in mice [49]. Targeting M-CSF appears to be an alternative approach to limiting excessive activation of osteoclastogenesis. However, due to its equally important function in polarization of macrophages, such as M1 macrophages against infection, immunomodulation of osteoclastogenesis using M-CSF neutralizing antibodies or inhibitors should be particularly cautious with the consideration of the context and regimen to avoid severe immunosuppression.

2.3. IL-1β

IL-1β is a pro-inflammatory cytokine that induces bone destruction in inflammatory bone disease through the activation of osteoclast. IL-1β promotes osteoclast differentiation through its synergetic function with RANKL/RANK signaling to enhance TRAF6 and its downstream signal transduction [50]. IL-1β also facilitates osteoclastogenesis through its receptor IL-1RI and strongly recruits MITF to the promoter region of TRAP and OSCAR, which are osteoclast marker genes (Fig. 2) [51]. In vivo mice studies suggested that IL-1 stimulates osteoclastogenesis since IL-1β deficiency increased bone mass, resulting from decreased osteoclast numbers. Interestingly, the numbers of osteoblasts and areas of osteoid surface per bone surface (OS/BS) were significantly reduced in IL-1β knock-out mice [52]. RANKL expression on stromal cells can be induced by IL-1β, which directly links to the activation of osteoclast precursors [53]. These notions were also observed in patients with postmenopausal osteoporosis and RA patients, where the serum IL-1β and RANKL expression were largely increased in the active phase of diseases, while the reduction of these cytokines were observed after the remission of osteoporosis or RA symptoms in patients [54,55]. Interestingly, different from its common roles in resolving inflammation, TREG cells were found to be osteoclastogenic and became RANKL-expressing TREG cells in the presence of abundant IL-1β to accelerate osteoclast formation and bone erosion [56]. These results further demonstrate the strong immunomodulatory effects of cytokines on osteoclastogenesis.

2.4. IL-17

IL-17 is produced by TH17 cells and group 3 innate lymphoid cells in inflammation and infection. It is also known that IL-17 promotes osteoclastogenesis in arthritis and osteoporosis with enhanced RANKL/OPG ratio in mice and patients [5,57]. As early as in 1999, IL-17 was found to induce COX-2-mediated PGE2 synthesis in osteoblasts/stromal cells in synovial tissues, which subsequently directs RANKL expression and secretion in osteoblasts. The increased RANKL further stimulates osteoclastogenesis [57]. IL-17 was found to coordinate local inflammatory cytokine production, which results in an overall amplified proinflammatory tissue microenvironment. Cytokines, such as TNF-α, IL-1β, IL-6 and IL-23, are rapidly secreted into the inflammatory sites after initial elevation of IL-17, which altogether intensifies RANKL expression and RANKL induced osteoclast differentiation. Indeed, in vivo studies suggested that IL-23/IL-17 axis is critical for inflammation-induced bone destruction, where a pronounced reduction of bone destruction was found in IL-17 and IL-23α knock-out mice [39]. On the other hand, IL-17A can also enhance the production of granulocyte-macrophage colony-stimulating factor (GM-CSF) by osteoblasts, which maintains monocytes in an undifferentiated state by downregulating c-Fos, Fra-1, and nuclear factor of activated T cells 1 (NFATc1) [58,59]. Of note, mice with IL-17A deficiency have normal bone homeostasis and development of functional osteoclasts [60]. This is likely due to the dual role of IL-17A in driving RANKL production while maintaining the possibility to induce osteoblast differentiation and osteogenesis by activating JAK2/STAT3 signaling in the mesenchymal stem cells and other bone-derived cells [61-63]. In some postmenopausal women, serum IL-17 levels were elevated and associated with reduced bone mass and estrogen levels [64-66]. As a signature cytokine of TH17 cells, increased secretion of IL-17 is usually accompanied by other proinflammatory cytokines, such as IL-1β, IL-6, IL-21, IL-22 and IL-23, which all show close relation with osteoclastogenesis and bone disease. This unique feature identifies the amount of TH17 cells as a good diagnostic marker for diseases like osteoporosis and rheumatoid arthritis as well as key immunomodulatory targets conferring prevention and protection for bone disorders.

2.5. TNF-α

Many immune cells produce TNF-α in inflammation and infection. This includes activated macrophages, T cells, B cells, and NK cells [67]. Meanwhile, TNF-α is a critical factor for bone destruction especially during inflammatory bone diseases [68]. Mechanistically, TNF-α directly binds to its receptor on cell surface and synergize with RANKL signaling to promote osteoclastogenesis (Fig. 2) [69]. Moreover, TNF-a stimulated M-CSF gene expression, in turn, induces RANK expression in osteoclast precursors [48]. Importantly, TNF-α often acts in synergy with RANKL and/or together with other inflammatory cytokines, such as IL-6 to promote osteoclastogenesis and bone resorption under inflammatory conditions [70,71]. Consistently, elevated TNF-α was found in postmenopausal women, which contributes to the increased incidents of osteoporosis [72]. Similar bone erosion phenomenon has been observed in many other inflammatory bone diseases, such as RA and periodontitis, all with increased serum level of TNF-α [73-75]. TNF-α also induces the production of Dickkopf-1 (DKK1), a critical inhibitor of WNT pathway. TNF-upregulated DKK1 suppresses bone formation and thus contributes to inflammatory bone loss, such as in RA [76,77]. Of note, TNF-α can suppress osteoblast differentiation by inhibiting the expression of IGF-1, BMP2, osterix (OSX), and osteocalcin (OCN), which result in degradation of Runx2 and inhibition of SMADs in osteoblasts [78-80]. In addition, Xing and colleagues revealed that Notch activation suppressed TNF-α-mediated inhibition of MSC differentiation into osteoblasts via noncanonical NF-κB signaling in inflammatory arthritis [81]. It is thus clear that TNF-α induces bone erosion by both increasing osteoclastic bone resorption and decreasing osteoblastic bone formation in many inflammatory diseases associated with bone loss. It is thus promising to improve bone health through the use of blocking antibodies (infliximab) and inhibitors of TNF-α or its downstream targets as a complementary approach in patients with inflammatory bone diseases.

2.6. IL-7, IL-8 and IL-15

IL-7 is known for the maintenance of immune cell memory. While during the inflammation, stromal cells and osteoblasts increase the secretion of IL-7 in response to IL-1b and TNFa, and this IL-7 might directly function on osteoclast through the activation of STAT5, independent of RANKL [82]. It was shown that IL-7 also potentiated OC formation by promoting the RANKL and TNF-α secretion from activated T cells [83]. Cytokine IL-8 (CXCL8) is important in recruiting neutrophil, monocytes and other immune cells in response to infection [84]. Experiment has found that IL-8 promoted osteoclastogenesis and bone resorption in osteolysis in conjunction with metastatic breast cancer and multiple myeloma [85-87]. IL-8 also can act as an autocrine regulator of osteoclastogenesis, which mediated by RANKL-induced NFATc1 activation. Human osteoclast precursors secreted IL-8 prior to the formation of mature OC, where IL-8 could strengthen osteoclastogenic effect of RANKL [86]. The inhibition of IL-8 with neutralizing antibody or IL-8 receptors, CXCR1 or CXCR2, attenuated RANKL-induced osteoclastogenesis [86]. Interestingly, two studies reported that the serum level of IL-8 was enhanced in arthritis-specific and anticitrullinated protein/peptide antibodies (ACPAs)-positive RA patients. During the differentiation and activation of osteoclast precursors, circulating ACPAs binds to the mature osteoclasts in bone marrow, leading to an increased osteoclast activity and bone resorption. Interestingly, blocking of IL-8 by neutralizing antibodies markedly suppressed the ACPA-induced osteoclast formation [88,89]. IL-15 is a IL-2 family cytokine, and it shares many structural and biological properties with IL-2 [90]. Many cells secrete IL-15, including monocytes, macrophages, dendritic cells, fibroblasts, epithelial cells, and bone marrow stromal cells [91]. IL-15 induces RANKL expression by osteoblasts and stromal cells that subsequently affect osteoclastogenesis [92]. Cell line studies also found that IL-15 promoted RANKL-induced osteoclastogenesis in RAW264.7 (RAW) cells by activating ERK signaling [93]. The blockade of IL-15 reduces the number of osteoclasts in the joint and local bone destruction during arthritis in mice [94,95]. Notwithstanding, inconsistent results also showed that IL-15 could active NK cells that can induce the apoptosis of osteoclast resulting in reduced bone resorption [96,97]. Studies that elucidate the clear function of these cytokines are urgently awaited.

3. Negative regulation of osteoclastogenesis by cytokines

3.1. IL-4, IL-5, IL-9 and IL-13

IL-4, IL-5, IL-9 and IL-13 are cytokines that belong to type 2 inflammation. These cytokines are key immunomodulators during allergic response as well as antiparasitic infections. It is also shown that type 2 inflammation is protective in skewing proinflammation response that occurs in diseases such as obesity [98], diabetes [99], rheumatic arthritis [100] and osteoarthritis [101,102]. Type 2 helper T (TH2) cells and group 2 innate lymphoid (ILC2) cells are major cells producing these cytokines amongst other immune cells [103,104]. Indeed, studies have suggested that TH2 cells and ILC2 cells are protective against bone loss during severe inflammation [100-102]. The immunomodulatory functions of these cells in bone remodeling are mainly mediated by production of cytokines. As a signature cytokine of this kind, IL-4 could directly inhibit bone resorbing activity driven by mature osteoclasts through the suppression of NF-kB activation in a STAT6-dependent manner in osteoclast precursors [27,28]. IL-4 can also indirectly inhibit osteoclast formation through regulation of osteoblast to produce greater amount of osteoprotegerin (OPG). OPG is the soluble decoy receptor of RANKL thus reduces the amount of RANKL in the microenvironment [105]. Interestingly, IL-4 deficient mice are sensitive to RANKL induced hypercalcemia, a condition that can lead to the exacerbated bone resorption by osteoclast [106]. Human clinical study also reported inhibitory effect of IL-4 on RANKL-induced osteoclastogenesis through peroxisome proliferator-activated receptor γ1 (PPARγ1), which laid the foundation, in part, for the beneficial effects of the thiazolidinedione class of PPARγ1 ligand on bone loss in diabetic patients [107]. Meanwhile, others revealed that the serum concentration of IL-4 was markedly lower in patients with postmenopausal osteoporosis, which closely correlated with reduction of bone marrow density [108]. IL-13 shares the same gamma receptor with IL-4, which could suppress RANKL expression while promoting OPG expression in osteoblasts in a STAT signaling-dependent manner together with IL-4 [28]. Interestingly, despite the similar inhibitory function on osteoclast formation, IL-4 exerts stronger inhibition than that of by IL-13 [109]. As mentioned above, newly discovered group 2 innate lymphoid (ILC2) cells also possess the regulatory functions to suppress the differentiation of osteoclasts. These regulatory functions are achieved by ILC2-derived IL-4/13, which considerably suppressed IL-1β secretion by macrophages and bone erosion in the inflamed sites by osteoclasts [102]. Similar immunomodulatory function was found in another cohort of patients with rheumatoid arthritis, where ILC2s produced IL-9 and remarkably resolved the inflammation with altered numbers of osteoclasts and improved bone protection [100]. IL-5 also mediates bone formation through the mobilization and activation of osteogenic progenitors and/or inhibition of recruited osteoclasts [110]. In summary, cytokines that belong to the type 2 immunity/inflammation are remarkable candidates that can elicit protective roles in preventing bone loss. However, caution exists because these cytokines also robustly regulate other immune processes, which might induce unexpected complications, such as allergic response [111], dermatitis [112], and promote IgE isotype class switching and induce other autoimmune diseases, such as lupus and asthma [113]. Therefore, further research on how to precisely modulate osteoclastogenesis and bone remodeling process by type 2 cytokines is greatly needed and currently emerging.

3.2. IL-10

IL-10 is a vital immunomodulatory cytokine with potent anti-inflammatory function. Many immune cells involving M2 macrophage, dendritic cells, regulatory B cells, and regulatory subsets of CD4+ and CD8+ T cells achieve their tuning functions by producing IL-10 [114,115]. As a regulatory cytokine, IL-10 primarily suppresses differentiation and proliferation of the responding cells expressing IL-10 receptor. These include monocytes, macrophages, dendritic cells, neutrophils, B cells and T cells [115]. Numerous studies have found that IL-10 elicited vigorous inhibition on osteoclastogenesis by reducing the intrinsic expression of NFATc1 and the presence of RANKL and M-CSF secreted by other cells [116-118]. In vivo studies found that IL-10 deficient mice developed osteopenia, characterized by decreased bone mass and impaired bone formation [119]. While secretion of a number of inflammatory factors, such as TNF-α, IFN-γ, RANKL, and NO, was remarkably increased in the same mice, decreased expression of markers including OCN, ALP and phosphate-regulating gene endopeptidases (PHRX), known key factors for osteoblasts and osteocytes, was observed [119,120]. In patients with postmenopausal osteoporosis, serum levels of IL-10 are significantly lower than healthy women [121]. IL-10 gene polymorphisms are associated with bone mineral density and the susceptibility to osteoporosis in postmenopausal women [122,123]. This evidence suggests a unique role for those major IL-10 producing cells, such as TREG cells and IL-10-producing innate lymphoid cells, in controlling excessive osteoclastogenesis during bone inflammation. Indeed, recent studies have highlighted that TREG cells exceptionally inhibited differentiation of osteoclast through IL-10 and CD80/CD86 mediated immunomodulatory function [6,7]. As a rapidly growing field, a number of TREG cell-based immunotherapies are currently under clinical trials for treating inflammatory and autoimmune diseases [18,124,125]. Most of these clinical trials require sufficient IL-10 secretion by TREG cells. It would be thus very interesting to see how these therapeutics could be translated into the treatment of inflammatory bone diseases in the near future.

3.3. Interferon-β (IFN-β)

Interferons are critical in regulating innate and adaptive immune responses against infections and can be sub-grouped into three major classes. Type I IFNs are critical for the regulation of osteoclastogenesis in mice. Induction of c-Fos and TRAF6 activates the NF-kB and JNK pathways, which promotes the production of Interferon-β (IFN-β) in osteoclasts [26]. However, it is also known that IFN-β can initiate negative feedback where it can reduce the activity of c-Fos, followed by inhibited osteoclastogenesis [26,126]. Indeed, administration of IFN-β improved bone loss in ovariectomized osteoporotic mice [26]. This negative feedback loop is important for bone homeostasis as IFNAR1 or IFN-β deficient mice with impaired feedback tend to have osteopenia [26]. Interestingly, gene expression profiling revealed that in human osteoclast in vitro cell culture, addition of IFN-β upregulated CXCL11, a chemokine which could further repress osteoclastogenesis (Fig. 2) [127]. Our group has identified that miR182 is an upstream inhibitor whereas Def6 and PKR are activators of IFN-β expression. These factors form a regulatory network to coordinate and fine tune osteoclast differentiation. The miR182-PKR-IFN-β axis and Def6-IFN-β pathway play important roles not only in bone homeostasis but also in pathological bone loss [128,129].

3.4. IL-12 and IL-27

IL-12 and IL-27 both belong to the IL-12 family of cytokines. These cytokines are secreted by macrophages, monocytes, dendritic and B cells in response to bacterial and parasitic infections [130]. IL-12 can inhibit RANKL/TNF-α/LPS-induced osteoclastogenesis in vitro, which is attributed to the FAS/FASL-mediated apoptosis [131-133]. It was also found that IL-12 reduced TNF-α induced osteolysis in vivo [134]. However, it was still not very clear how IL-12 might affect the osteoclastogenesis along with other cytokines. IL-27 has a pleiotropic immunoregulatory functions. It was shown that IL-27 suppressed RANKL-induced osteoclastgenesis in vitro by inhibiting MAPK signaling pathway and NFATc1 expression [135]. IL-27 also functions on STAT1 which subsequently reduced the activation of c-Fos in osteoclast precursor cells [135,136]. A recent in vivo study revealed that IL-27 treatment prevents estrogen-deficient induced bone loss through promotion of early growth response gene 2 (Egr2) and Id2 in mice [137]. It is tempting to expect that more exciting results will be found from bench-to-bedside studies on IL-12 and IL-27-mediated regulation of osteoclastogenesis.

4. Cytokines that show dual roles in osteoclastogenesis

There are many other cytokines involved in the regulation of osteoclastogenesis, for instance, interferon γ, IL-2, IL-6 and TGF-β. These cytokines often play a multifaceted role during bone remodeling. IFN-γ produced by T cells strongly accelerates JAK-STAT1 signaling transduction to induce rapid degradation of TRAF6, which results in interfering with RANKL-RANK signaling to suppress osteoclastogenesis [3]. Moreover, IFN-γ also reduces the expression of c-fms and RANK on the surface of osteoclast precursors to synergistically inhibit osteoclastogenesis with other factors such as Toll-like receptors TLRs (Fig. 2) [138]. Interestingly, IFN-γ not only inhibits osteoclast differentiation directly, but also reduces osteoclastogenesis by promoting osteoblast-derived NO, which could induce the apoptosis of osteoclast via FAS ligand/Fas signaling [139,140]. On the other hand, however, IFN-γ can promote the fusion of osteoclast precursor required for osteoclastogenesis through the induction of the expression of dendritic cell-specific transmembrane protein (DC-stamp), which is often required for immature osteoclast fusion [141]. In vivo studies further support the binary function of IFN-γ on OC formation, where bone volume was found significantly reduced in IFN-γ or IFN-γ receptor deficiency mice, while exogenous IFN-γ administration can lead to severe bone loss in osteoporotic mice [3,142]. Controversial results were also found with regards to IL-2, where IL-2 was found to stimulate osteoclastic activities [143], but the deficiency of IL-2 in mice could lead to severe bone loss [90,144].

IL-6 is another cytokine of such kind, where in some conditions it is pathogenetic for inflammatory bone loss through its promotion of osteoclast precursors into mature osteoclasts [145,146]. On the flip side, IL-6 was suggested to suppress RANKL-mediated NF-kB and JNK activation in during osteoclastogenesis (Fig. 2) [147]. Tocilizumab, the first humanized anti-IL-6 receptor antagonist, has been approved by FDA to treat patients with rheumatoid arthritis [148]. Clinical study also found that Tocilizumab administration in patients with anticitrullinated protein antibody-positive rheumatoid arthritis might prevent bone loss [149], which supports the beneficial effects on bone by blocking IL-6 signaling. Although more clear results should be provided from bed to bench side, IL-6 might serve as good target for bone diseases in the context of severe inflammation.

IL-23 belongs to the IL-12 family of cytokine. Like IL-6, both positive and negative roles were reported regarding to the regulation of OC formation. On the one hand, IL-23 promotes osteoclast formation in vitro by upregulating RANK expression on osteoclast precursors [150]. IL-23 also activates DAP12, which is required for osteoclastogenesis [151]. Indirectly, IL-23 can enhance the osteoclastogenesis by promotion of TH17 cell polarization and IL-17A production, this will lead to the increased production of RANKL required for osteoclastogenesis [150,152]. The anti-IL-23 antibody reduces synovial inflammation and bone destruction in rats with collagen-induced arthritis [153]. On the other hand, IL-23 was reported to inhibit osteoclastogenesis in vitro [154,155]. Indeed, the genetic IL-23p19 deficient mice shows 30 % decrease in bone mass [154]. Moreover, clinical study has found that anti-IL-12/23 p40 monoclonal antibody (ustekinuma) treatment significantly improved the joint damage in patients with active psoriatic arthritis (PsA) [156]. Hence, IL-23 might have context-dependent regulation of regulating osteoclastogenesis.

Aberrant transforming growth factor-β 1(TGF-β1) signaling induces immune cell abnormalities, which is one of the leading causes of inflammation, allergy, autoimmunity and cancer [157]. Indeed, TGF-β1-deficient mice die within 2–3 weeks due to the severe lymphocyte infiltration into multiple vital organs [157]. Intriguingly, TGF-β1 is one of most abundant cytokines in the bone matrix during bone homeostasis, which has been shown to regulate both bone resorption and bone formation [158-160]. Despite lots of studies investigating the effect of TGF-β on osteoclastogenesis, the conclusions are still controversial and contradictory [161-167]. Some studies reported that endogenously produced TGF-β is an essential autocrine factor for osteoclastogenesis in combination with RANKL and M-CSF [165]. TGF-β binds to its receptor on osteoclasts and activates SMAD2/3, which is mediated by TRAF6-TAB1-TAK1 complex and triggers osteoclast differentiation [162]. On the other hand, a previous study reports that endogenous TGF-β1 inhibits bone resorption partly by inducing osteoclast apoptosis through up-regulating Bim in human osteoclast cultures and also partly by stimulating osteoblastic bone formation [164]. However, previous works also show that low-concentration treatment of TGF-β1 promotes RANKL or TNF-α stimulated osteoclast differentiation via direct action, while high level of TGF-β1 indirectly represses osteoclast differentiation by enhancing OPG secretion from osteoblasts in the cell cultures [161,166]. Another study found that TGF-β1 showed both inhibitory and stimulatory effects on human osteoclast differentiation, and that these opposing functions are mediated by SMAD1 and SMAD3 signaling, respectively [163]. TGF-β1 inhibits osteoclastogenesis when added at the early stages of differentiation, suggesting that timing affects the function of TGF-β1 on osteoclastogenesis [163]. The significantly higher level of human TGF-β1 was noticed in subchondral bone of knee joints throughout of osteoarthritis in patients comparing to that of in healthy donors [168]. Moreover, inhibition of TGF-β signaling attenuated cartilage damage and improved the inflammatory bone loss in OA [168]. Taken together, TGF-β1 is an important cytokine in regulating bone homeostasis, while its clear role on osteoclastogenesis requires genetic evidence and further investigation.

5. Conclusion

In this review, we summarized recent progress in cytokine-mediated immunomodulation of osteoclastogenesis and discussed potential therapeutic possibilities of bench-side research into novel therapies to treat bone disease (Table 1). In a fast-growing field of immunotherapy, cytokines serve as an excellent intermediate to connect skeletal system and immune system. Due to the dual role or even multifaceted roles of cytokines in regulating bone remodeling, it might not be practical to develop a single cytokine-based treatment to treat broad and complex conditions of bone disorders. Taken this into account, the heterogenous nature of human genetics also requires more precise and personalized treatment options to confer protection or improvement of disease symptoms with limited adverse events. Novel techniques, such as single cell RNA-sequencing, CRISPR editing and screening, can facilitate the development of new treatments targeting more than one cytokine for the improvement of bone health in patients with different needs and backgrounds. By doing so, more exciting results from both fundamental science and clinical research of osteoimmunology would emerge and are expected.

Acknowledgments

We thank Courtney Ng for critical review of the manuscript.

Footnotes

CRediT authorship contribution statement

P.Z, T.Z and B.Z initiated the topic and wrote the review.

Declaration of competing interest

The authors declare no conflict of interest.

References

  • [1].IOF Compendium of Osteoporosis, 2nd Editions, 2019 (2019). [Google Scholar]
  • [2].Tsukasaki M, Takayanagi H, Osteoimmunology: evolving concepts in bone-immune interactions in health and disease, Nat. Rev. Immunol 19 (10) (2019) 626–642. [DOI] [PubMed] [Google Scholar]
  • [3].Takayanagi H, Ogasawara K, Hida S, Chiba T, Murata S, Sato K, Takaoka A, Yokochi T, Oda H, Tanaka K, Nakamura K, Taniguchi T, T-cell-mediated regulation of osteoclastogenesis by signalling cross-talk between RANKL and IFN-gamma, Nature 408 (6812) (2000) 600–605. [DOI] [PubMed] [Google Scholar]
  • [4].Kobayashi K, Takahashi N, Jimi E, Udagawa N, Takami M, Kotake S, Nakagawa N, Kinosaki M, Yamaguchi K, Shima N, Yasuda H, Morinaga T, Higashio K, Martin TJ, Suda T, Tumor necrosis factor alpha stimulates osteoclast differentiation by a mechanism independent of the ODF/RANKL-RANK interaction, J. Exp. Med 191 (2) (2000) 275–286. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [5].Lubberts E, van den Bersselaar L, Oppers-Walgreen B, Schwarzenberger P, Coenen-de Roo CJ, Rolls JK, Joosten LA, van den Berg WB, IL-17 promotes bone erosion in murine collagen-induced arthritis through loss of the receptor activator of NF-kappa B ligand/osteoprotegerin balance, J. Immunol 170 (5) (2003) 2655–2662. [DOI] [PubMed] [Google Scholar]
  • [6].Zaiss MM, Axmann R, Zwerina J, Polzer K, Guckel E, Skapenko A, Schulze-Koops H, Horwood N, Cope A, Schett G, Treg cells suppress osteoclast formation: a new link between the immune system and bone, Arthritis Rheum. 56 (12) (2007) 4104–4112. [DOI] [PubMed] [Google Scholar]
  • [7].Bozec A, Zaiss MM, Kagwiria R, Voll R, Rauh M, Chen Z, Mueller-Schmucker S, Kroczek RA, Heinzerling L, Moser M, Mellor AL, David JP, Schett G, T cell costimulation molecules CD80/86 inhibit osteoclast differentiation by inducing the IDO/tryptophan pathway, Sci. Transl. Med 6 (235) (2014) 235ra60. [DOI] [PubMed] [Google Scholar]
  • [8].Zhu N, Zhang D, Wang W, Li X, Yang B, Song J, Zhao X, Huang B, Shi W, Lu R, Niu P, Zhan F, Ma X, Wang D, Xu W, Wu G, Gao GF, Tan W, China Novel Coronavirus I, Research T, A novel coronavirus from patients with pneumonia in China, 2019, N. Engl. J. Med 382 (8) (2020) 727–733. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [9].Gong F, Dai Y, Zheng T, Cheng L, Zhao D, Wang H, Liu M, Pei H, Jin T, Yu D, Zhou P, Peripheral CD4+ T cell subsets and antibody response in COVID-19 convalescent individuals, J. Clin. Invest 130 (12) (2020) 6588–6599. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [10].Ferbebouh M, Vallieres F, Benderdour M, Fernandes J, The pathophysiology of immunoporosis: innovative therapeutic targets, Inflamm. Res 70 (8) (2021) 859–875. [DOI] [PubMed] [Google Scholar]
  • [11].Drake MT, Clarke BL, Khosla S, Bisphosphonates: mechanism of action and role in clinical practice, Mayo Clin. Proc 83 (9) (2008) 1032–1045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [12].Whitaker M, Guo J, Kehoe T, Benson G, Bisphosphonates for osteoporosis—where do we go from here? N. Engl. J. Med 366 (22) (2012) 2048–2051. [DOI] [PubMed] [Google Scholar]
  • [13].Kraenzlin ME, Meier C, Parathyroid hormone analogues in the treatment of osteoporosis, Nat. Rev. Endocrinol 7 (11) (2011) 647–656. [DOI] [PubMed] [Google Scholar]
  • [14].Neer RM, Arnaud CD, Zanchetta JR, Prince R, Gaich GA, Reginster JY, Hodsman AB, Eriksen EF, Ish-Shalom S, Genant HK, Wang O, Mitlak BH, Effect of parathyroid hormone (1-34) on fractures and bone mineral density in postmenopausal women with osteoporosis, N. Engl. J. Med 344 (19) (2001) 1434–1441. [DOI] [PubMed] [Google Scholar]
  • [15].Cosman F, Crittenden DB, Adachi JD, Binkley N, Czerwinski E, Ferrari S, Hofbauer LB, Lau E, Lewiecki EM, Miyauchi A, Zerbini CA, Milmont CE, Chen L, Maddox J, Meisner PD, Libanati C, Grauer A, Romosozumab treatment in postmenopausal women with osteoporosis, N. Engl. J. Med 375 (16) (2016) 1532–1543. [DOI] [PubMed] [Google Scholar]
  • [16].McClung MR, Grauer A, Boonen S, Bolognese MA, Brown JP, Diez-Perez A, Langdahl BL, Reginster JY, Zanchetta JR, Wasserman SM, Katz L, Maddox J, Yang YC, Libanati C, Bone HG, Romosozumab in postmenopausal women with low bone mineral density, N. Engl. J. Med 370 (5) (2014) 412–420. [DOI] [PubMed] [Google Scholar]
  • [17].Langdahl BL, Libanati C, Crittenden DB, Bolognese MA, Brown JP, Daizadeh NS, Dokoupilova E, Engelke K, Finkelstein JS, Genant HK, Goemaere S, Hyldstrup L, Jodar-Gimeno E, Keaveny TM, Kendler D, Lakatos P, Maddox J, Malouf J, Massari FE, Molina JF, Ulla MR, Grauer A, Romosozumab (sclerostin monoclonal antibody) versus teriparatide in postmenopausal women with osteoporosis transitioning from oral bisphosphonate therapy: a randomised, open-label, phase 3 trial, Lancet 390 (10102) (2017) 1585–1594. [DOI] [PubMed] [Google Scholar]
  • [18].Zhou P, Chen J, He J, Zheng T, Yunis J, Makota V, Alexandre YO, Gong F, Zhang X, Xie W, Li Y, Shao M, Zhu Y, Sinclair JE, Miao M, Chen Y, Short KR, Mueller SN, Sun X, Yu D, Li Z, Low-dose IL-2 therapy invigorates CD8+ T cells for viral control in systemic lupus erythematosus, PLoS Pathog. 17 (10) (2021), e1009858. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [19].Zheng T, Kang JH, Sim JS, Kim JW, Koh JT, Shin CS, Lim H, Yim M, The farnesoid X receptor negatively regulates osteoclastogenesis in bone remodeling and pathological bone loss, Oncotarget 8 (44) (2017) 76558–76573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [20].Boyle WJ, Simonet WS, Lacey DL, Osteoclast differentiation and activation, Nature 423 (6937) (2003) 337–342. [DOI] [PubMed] [Google Scholar]
  • [21].Kim N, Kadono Y, Takami M, Lee J, Lee SH, Okada F, Kim JH, Kobayashi T, Odgren PR, Nakano H, Yeh WC, Lee SK, Lorenzo JA, Choi Y, Osteoclast differentiation independent of the TRANCE-RANK-TRAF6 axis, J. Exp. Med 202 (5) (2005) 589–595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [22].Jun Hirose HM, Tokuyama Naoto, Omata Yasunori, Matsumoto Takumi, Yasui Tetsuro, Kadono Yuho, Hennighausen Lothar, Tanaka Sakae, Bone resorption is regulated by cell-autonomous negative feedback loop of Stat5–Dusp axis in the osteoclast, J. Exp. Med 211 (1) (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [23].Miyauchi Y, Ninomiya K, Miyamoto H, Sakamoto A, Iwasaki R, Hoshi H, Miyamoto K, Hao W, Yoshida S, Morioka H, Chiba K, Kato S, Tokuhisa T, Saitou M, Toyama Y, Suda T, Miyamoto T, The Blimp1-Bcl6 axis is critical to regulate osteoclast differentiation and bone homeostasis, J. Exp. Med 207 (4) (2010) 751–762. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [24].Walsh MC, Takegahara N, Kim H, Choi Y, Updating osteoimmunology: regulation of bone cells by innate and adaptive immunity, Nat. Rev. Rheumatol 14 (3) (2018) 146–156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [25].Eberl G, Colonna M, Di Santo JP, McKenzie AN, Innate lymphoid cells. Innate lymphoid cells: a new paradigm in immunology, Science 348 (6237) (2015) aaa6566. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [26].Takayanagi H, Kim S, Matsuo K, Suzuki H, Suzuki T, Sato K, Yokochi T, Oda H, Nakamura K, Ida N, Wagner EF, Taniguchi T, RANKL maintains bone homeostasis through c-Fos-dependent induction of interferon-beta, Nature 416 (6882) (2002) 744–749. [DOI] [PubMed] [Google Scholar]
  • [27].Abu-Amer Y, IL-4 abrogates osteoclastogenesis through STAT6-dependent inhibition of NF-kappaB, J. Clin. Invest 107 (11) (2001) 1375–1385. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [28].Stein NC, Kreutzmann C, Zimmermann SP, Niebergall U, Hellmeyer L, Goettsch B, Schoppet M, Hofbauer LC, Interleukin-4 and interleukin-13 stimulate the osteoclast inhibitor osteoprotegerin by human endothelial cells through the STAT6 pathway, J. Bone Miner. Res 23 (5) (2008) 750–758. [DOI] [PubMed] [Google Scholar]
  • [29].Wang J, Sun J, Liu LN, Flies DB, Nie X, Toki M, Zhang J, Song C, Zarr M, Zhou X, Han X, Archer KA, O’Neill T, Herbst RS, Boto AN, Sanmamed MF, Langermann S, Rimm DL, Chen L, Siglec-15 as an immune suppressor and potential target for normalization cancer immunotherapy, Nat. Med 25 (4) (2019) 656–666. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [30].Kameda Y, Takahata M, Komatsu M, Mikuni S, Hatakeyama S, Shimizu T, Angata T, Kinjo M, Minami A, Iwasaki N, Siglec-15 regulates osteoclast differentiation by modulating RANKL-induced phosphatidylinositol 3-kinase/Akt and Erk pathways in association with signaling Adaptor DAP12, J. Bone Miner. Res 28 (12) (2013) 2463–2475. [DOI] [PubMed] [Google Scholar]
  • [31].Zhen G, Dan Y, Wang R, Dou C, Guo Q, Zarr M, Liu LN, Chen L, Deng R, Li Y, Shao Z, Cao X, An antibody against Siglec-15 promotes bone formation and fracture healing by increasing TRAP(+) mononuclear cells and PDGF-BB secretion, Bone Res. 9 (1) (2021) 47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [32].Yasuda H, Discovery of the RANKL/RANK/OPG system, J. Bone Miner. Metab 39 (1) (2021) 2–11. [DOI] [PubMed] [Google Scholar]
  • [33].Burgess TL, Qian Y, Kaufman S, Ring BD, Van G, Capparelli C, Kelley M, Hsu H, Boyle WJ, Dunstan CR, Hu S, Lacey DL, The ligand for osteoprotegerin (OPGL) directly activates mature osteoclasts, J. Cell Biol 145 (3) (1999) 527–538. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [34].Lacey DL, Timms E, Tan HL, Kelley MJ, Dunstan CR, Burgess T, Elliott R, Colombero A, Elliott G, Scully S, Hsu H, Sullivan J, Hawkins N, Davy E, Capparelli C, Eli A, Qian YX, Kaufman S, Sarosi I, Shalhoub V, Senaldi G, Guo J, Delaney J, Boyle WJ, Osteoprotegerin ligand is a cytokine that regulates osteoclast differentiation and activation, Cell 93 (2) (1998) 165–176. [DOI] [PubMed] [Google Scholar]
  • [35].Hofbauer LC, Schoppet M, Clinical implications of the osteoprotegerin/RANKL/RANK system for bone and vascular diseases, JAMA 292 (4) (2004) 490–495. [DOI] [PubMed] [Google Scholar]
  • [36].Buckley L, Humphrey MB, Glucocorticoid-induced osteoporosis, N. Engl. J. Med 379 (26) (2018) 2547–2556. [DOI] [PubMed] [Google Scholar]
  • [37].Yamada T, Fukasawa K, Horie T, Kadota T, Lyu J, Tokumura K, Ochiai S, Iwahashi S, Suzuki A, Park G, Ueda R, Yamamoto M, Kitao T, Shirahase H, Ochi H, Sato S, Iezaki T, Hinoi E, The role of CDK8 in mesenchymal stem cells in controlling osteoclastogenesis and bone homeostasis, Stem Cell Rep. 17 (7) (2022) 1576–1588. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [38].Amirhosseini M, Bernhardsson M, Lang P, Andersson G, Flygare J, Fahlgren A, Cyclin-dependent kinase 8/19 inhibition suppresses osteoclastogenesis by downregulating RANK and promotes osteoblast mineralization and cancellous bone healing, J. Cell. Physiol 234 (9) (2019) 16503–16516. [DOI] [PubMed] [Google Scholar]
  • [39].Sato K, Suematsu A, Okamoto K, Yamaguchi A, Morishita Y, Kadono Y, Tanaka S, Kodama T, Akira S, Iwakura Y, Cua DJ, Takayanagi H, Th17 functions as an osteoclastogenic helper T cell subset that links T cell activation and bone destruction, J. Exp. Med 203 (12) (2006) 2673–2682. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [40].Kikuta J, Wada Y, Kowada T, Wang Z, Sun-Wada GH, Nishiyama I, Mizukami S, Maiya N, Yasuda H, Kumanogoh A, Kikuchi K, Germain RN, Ishii M, Dynamic visualization of RANKL and Th17-mediated osteoclast function, J. Clin. Invest 123 (2) (2013) 866–873. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [41].Bando JK, Gilfillan S, Song C, McDonald KG, Huang SC, Newberry RD, Kobayashi Y, Allan DSJ, Carlyle JR, Cella M, Colonna M, The tumor necrosis factor superfamily member RANKL suppresses effector cytokine production in Group 3 innate lymphoid cells, Immunity 48 (6) (2018) 1208–1219 (e4). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [42].Glantschnig H, Fisher JE, Wesolowski G, Rodan GA, Reszka AA, M-CSF, TNFalpha and RANK ligand promote osteoclast survival by signaling through mTOR/S6 kinase, Cell Death Differ. 10 (10) (2003) 1165–1177. [DOI] [PubMed] [Google Scholar]
  • [43].Hattersley JOG, Flanagan AM, Chambers TJ, Macrophage colony stimulating factor (M-CSF) is essential for osteoclast formation in vitro, Biochem. Biophys. Res. Commun 177 (1) (1991) 526–531. [DOI] [PubMed] [Google Scholar]
  • [44].Dai XM, Ryan GR, Hapel AJ, Dominguez MG, Russell RG, Kapp S, Sylvestre V, Stanley ER, Targeted disruption of the mouse colony-stimulating factor 1 receptor gene results in osteopetrosis, mononuclear phagocyte deficiency, increased primitive progenitor cell frequencies, and reproductive defects, Blood 99 (1) (2002) 111–120. [DOI] [PubMed] [Google Scholar]
  • [45].Takei I, Takagi M, Ida H, Ogino T, Santavirta S, Konttinen YT, High macrophage-colony stimulating factor levels in synovial fluid of loose artificial hip joints, J. Rheumatol 27 (4) (2000) 894–899. [PubMed] [Google Scholar]
  • [46].Yang PT, Kasai H, Xiao WG, Zhao LJ, He LM, Yamashita A, Deng XW, Ito M, Increased expression of macrophage colony-stimulating factor in ankylosing spondylitis and rheumatoid arthritis, Ann. Rheum. Dis 65 (12) (2006) 1671–1672. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [47].Hamilton JA, Tak PP, The dynamics of macrophage lineage populations in inflammatory and autoimmune diseases, Arthritis Rheum. 60 (5) (2009) 1210–1221. [DOI] [PubMed] [Google Scholar]
  • [48].Kitaura H, Zhou P, Kim HJ, Novack DV, Ross FP, Teitelbaum SL, M-CSF mediates TNF-induced inflammatory osteolysis, J. Clin. Invest 115 (12) (2005) 3418–3427. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [49].Saleh R, Lee MC, Khiew SH, Louis C, Fleetwood AJ, Achuthan A, Forster I, Cook AD, Hamilton JA, CSF-1 in inflammatory and arthritic pain development, J. Immunol 201 (7) (2018) 2042–2053. [DOI] [PubMed] [Google Scholar]
  • [50].Amarasekara DS, Yun H, Kim S, Lee N, Kim H, Rho J, Regulation of osteoclast differentiation by cytokine networks, Immune Netw. 18 (1) (2018), e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [51].Kim JH, Jin HM, Kim K, Song I, Youn BU, Matsuo K, Kim N, The mechanism of osteoclast differentiation induced by IL-1, J. Immunol 183 (3) (2009) 1862–1870. [DOI] [PubMed] [Google Scholar]
  • [52].Lee YM, Fujikado N, Manaka H, Yasuda H, Iwakura Y, IL-1 plays an important role in the bone metabolism under physiological conditions, Int. Immunol 22 (10) (2010) 805–816. [DOI] [PubMed] [Google Scholar]
  • [53].Wei S, Kitaura H, Zhou P, Ross FP, Teitelbaum SL, IL-1 mediates TNF-induced osteoclastogenesis, J. Clin. Invest 115 (2) (2005) 282–290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [54].Dundar U, Kavuncu V, Ciftci IH, Evcik D, Solak O, Cakir T, The effect of risedronate treatment on serum cytokines in postmenopausal osteoporosis: a 6-month randomized and controlled study, J. Bone Miner. Metab 27 (4) (2009) 464–470. [DOI] [PubMed] [Google Scholar]
  • [55].Guiducci S, Del Rosso A, Cinelli M, Perfetto F, Livi R, Rossi A, Gabrielli A, Giacomelli R, Iori N, Fibbi G, Del Rosso M, Cerinic MM, Raloxifene reduces urokinase-type plasminogen activator-dependent proliferation of synoviocytes from patients with rheumatoid arthritis, Arthritis Res. Ther 7 (6) (2005) R1244–R1253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [56].Levescot A, Chang MH, Schnell J, Nelson-Maney N, Yan J, Martinez-Bonet M, Grieshaber-Bouyer R, Lee PY, Wei K, Blaustein RB, Morris A, Wactor A, Iwakura Y, Lederer JA, Rao DA, Charles JF, Nigrovic PA, IL-1beta-driven osteoclastogenic Tregs accelerate bone erosion in arthritis, J. Clin. Invest 131 (18) (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [57].Kotake S, Udagawa N, Takahashi N, Matsuzaki K, Itoh K, Ishiyama S, Saito S, Inoue K, Kamatani N, Gillespie MT, Martin TJ, Suda T, IL-17 in synovial fluids from patients with rheumatoid arthritis is a potent stimulator of osteoclastogenesis, J. Clin. Invest 103 (9) (1999) 1345–1352. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [58].Balani D, Aeberli D, Hofstetter W, Seitz M, Interleukin-17A stimulates granulocyte-macrophage colony-stimulating factor release by murine osteoblasts in the presence of 1,25-dihydroxyvitamin D(3) and inhibits murine osteoclast development in vitro, Arthritis Rheum. 65 (2) (2013) 436–446. [DOI] [PubMed] [Google Scholar]
  • [59].Atanga E, Dolder S, Dauwalder T, Wetterwald A, Hofstetter W, TNFalpha inhibits the development of osteoclasts through osteoblast-derived GM-CSF, Bone 49 (5) (2011) 1090–1100. [DOI] [PubMed] [Google Scholar]
  • [60].Adamopoulos IE, Chao CC, Geissler R, Laface D, Blumenschein W, Iwakura Y, McClanahan T, Bowman EP, Interleukin-17A upregulates receptor activator of NF-kappaB on osteoclast precursors, Arthritis Res. Ther 12 (1) (2010) R29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [61].Jo S, Wang SE, Lee YL, Kang S, Lee B, Han J, Sung IH, Park YS, Bae SC, Kim TH, IL-17A induces osteoblast differentiation by activating JAK2/STAT3 in ankylosing spondylitis, Arthritis Res. Ther 20 (1) (2018) 115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [62].Osta B, Lavocat F, Eljaafari A, Miossec P, Effects of interleukin-17A on osteogenic differentiation of isolated human mesenchymal stem cells, Front. Immunol 5 (2014) 425. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [63].Huang H, Kim HJ, Chang EJ, Lee ZH, Hwang SJ, Kim HM, Lee Y, Kim HH, IL-17 stimulates the proliferation and differentiation of human mesenchymal stem cells: implications for bone remodeling, Cell Death Differ. 16 (10) (2009) 1332–1343. [DOI] [PubMed] [Google Scholar]
  • [64].Pacifici R, The role of IL-17 and TH17 cells in the bone catabolic activity of PTH, Front. Immunol 7 (2016) 57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [65].Bhadricha H, Patel V, Singh AK, Savardekar L, Patil A, Surve S, Desai M, Increased frequency of Th17 cells and IL-17 levels are associated with low bone mineral density in postmenopausal women, Sci. Rep 11 (1) (2021) 16155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [66].Molnar I, Bohaty I, Somogyine-Vari E, IL-17A-mediated sRANK ligand elevation involved in postmenopausal osteoporosis, Osteoporos. Int 25 (2) (2014) 783–786. [DOI] [PubMed] [Google Scholar]
  • [67].Kalliolias GD, Ivashkiv LB, TNF biology, pathogenic mechanisms and emerging therapeutic strategies, Nat. Rev. Rheumatol 12 (1) (2016) 49–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [68].Zhao B, TNF and Bone remodeling, Curr. Osteoporos. Rep 15 (3) (2017) 126–134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [69].Zhang YH, Heulsmann A, Tondravi MM, Mukherjee A, Abu-Amer Y, Tumor necrosis factor-alpha (TNF) stimulates RANKL-induced osteoclastogenesis via coupling of TNF type 1 receptor and RANK signaling pathways, J. Biol. Chem 276 (1) (2001) 563–568. [DOI] [PubMed] [Google Scholar]
  • [70].Zhao B, Intrinsic restriction of TNF-mediated inflammatory osteoclastogenesis and bone resorption, Front. Endocrinol. (Lausanne) 11 (2020), 583561. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [71].Teitelbaum SL, Osteoclasts; culprits in inflammatory osteolysis, Arthritis Res. Ther 8 (1) (2006) 201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [72].Zha L, He L, Liang Y, Qin H, Yu B, Chang L, Xue L, TNF-alpha contributes to postmenopausal osteoporosis by synergistically promoting RANKL-induced osteoclast formation, Biomed. Pharmacother. 102 (2018) 369–374. [DOI] [PubMed] [Google Scholar]
  • [73].Osta B, Benedetti G, Miossec P, Classical and paradoxical effects of TNF-alpha on bone homeostasis, Front. Immunol 5 (2014) 48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [74].Schett G, Gravallese E, Bone erosion in rheumatoid arthritis: mechanisms, diagnosis and treatment, Nat. Rev. Rheumatol 8 (11) (2012) 656–664. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [75].Schett G, Teitelbaum SL, Osteoclasts and arthritis, J. Bone Miner. Res 24 (7) (2009) 1142–1146. [DOI] [PubMed] [Google Scholar]
  • [76].Diarra D, Stolina M, Polzer K, Zwerina J, Ominsky MS, Dwyer D, Korb A, Smolen J, Hoffmann M, Scheinecker C, van der Heide D, Landewe R, Lacey D, Richards WG, Schett G, Dickkopf-1 is a master regulator of joint remodeling, Nat. Med 13 (2) (2007) 156–163. [DOI] [PubMed] [Google Scholar]
  • [77].Schett G, Joint remodelling in inflammatory disease, Ann. Rheum. Dis 66 (Suppl. 3) (2007) iii42–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [78].Algate K, Haynes DR, Bartold PM, Crotti TN, Cantley MD, The effects of tumour necrosis factor-alpha on bone cells involved in periodontal alveolar bone loss; osteoclasts, osteoblasts and osteocytes, J. Periodontal Res 51 (5) (2016) 549–566. [DOI] [PubMed] [Google Scholar]
  • [79].Zhao L, Huang J, Zhang H, Wang Y, Matesic LE, Takahata M, Awad H, Chen B, Xing L, Tumor necrosis factor inhibits mesenchymal stem cell differentiation into osteoblasts via the ubiquitin E3 ligase Wwp1, Stem Cells 29 (10) (2011) 1601–1610. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [80].Kaneki H, Guo R, Chen D, Yao Z, Schwarz EM, Zhang YE, Boyce BF, Xing L, Tumor necrosis factor promotes Runx2 degradation through up-regulation of Smurf1 and Smurf2 in osteoblasts, J. Biol. Chem 281 (7) (2006) 4326–4333. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [81].Zhang H, Hilton MJ, Anolik JH, Welle SL, Zhao C, Yao Z, Li X, Wang Z, Boyce BF, Xing L, NOTCH inhibits osteoblast formation in inflammatory arthritis via noncanonical NF-kappaB, J. Clin. Invest 124 (7) (2014) 3200–3214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [82].Kim JH, Sim JH, Lee S, Seol MA, Ye SK, Shin HM, Lee EB, Lee YJ, Choi YJ, Yoo WH, Kim JH, Kim WU, Lee DS, Kim JH, Kang I, Kang SW, Kim HR, Interleukin-7 induces osteoclast formation via STATS, independent of receptor activator of NF-kappaB ligand, Front. Immunol 8 (2017) 1376. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [83].Toraldo G, Roggia C, Qian WP, Pacifici R, Weitzmann MN, IL-7 induces bone loss in vivo by induction of receptor activator of nuclear factor kappa B ligand and tumor necrosis factor alpha from T cells, Proc. Natl. Acad. Sci. U. S. A 100 (1) (2003) 125–130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [84].Bakouny Z, Choueiri TK, IL-8 and cancer prognosis on immunotherapy, Nat. Med 26 (5) (2020) 650–651. [DOI] [PubMed] [Google Scholar]
  • [85].Bendre MS, Montague DC, Peery T, Akel NS, Gaddy D, Suva LJ, Interleukin-8 stimulation of osteoclastogenesis and bone resorption is a mechanism for the increased osteolysis of metastatic bone disease, Bone 33 (1) (2003) 28–37. [DOI] [PubMed] [Google Scholar]
  • [86].Kopesky P, Tiedemann K, Alkekhia D, Zechner C, Millard B, Schoeberl B, Komarova SV, Autocrine signaling is a key regulatory element during osteoclastogenesis, Biol. Open 3 (8) (2014) 767–776. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [87].Herrero AB, Garcia-Gomez A, Garayoa M, Corchete LA, Hernandez JM, San Miguel J, Gutierrez NC, Effects of IL-8 up-regulation on cell survival and osteoclastogenesis in multiple myeloma, Am. J. Pathol 186 (8) (2016) 2171–2182. [DOI] [PubMed] [Google Scholar]
  • [88].Correction: identification of a novel chemokine-dependent molecular mechanism underlying rheumatoid arthritis-associated autoantibody-mediated bone loss, Ann. Rheum. Dis 78 (6) (2019) 866. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [89].Duarte JH, Rheumatoid arthritis: osteoclasts and ACPAs—the joint link, Nat. Rev. Rheumatol 12 (2) (2016) 69. [DOI] [PubMed] [Google Scholar]
  • [90].Zhou P, Emerging mechanisms and applications of low-dose IL-2 therapy in autoimmunity, Cytokine & Growth Factor Rev. (2022) 1–9, 10.1016/j.cytogfr.2022.06.003. In press. [DOI] [PubMed] [Google Scholar]
  • [91].Perera PY, Lichy JH, Waldmann TA, Perera LP, The role of interleukin-15 in inflammation and immune responses to infection: implications for its therapeutic use, Microbes Infect. 14 (3) (2012) 247–261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [92].Ogata Y, Kukita A, Kukita T, Komine M, Miyahara A, Miyazaki S, Kohashi O, A novel role of IL-15 in the development of osteoclasts: inability to replace its activity with IL-2, J. Immunol 162 (5) (1999) 2754–2760. [PubMed] [Google Scholar]
  • [93].Okabe I, Kikuchi T, Mogi M, Takeda H, Aino M, Kamiya Y, Fujimura T, Goto H, Okada K, Hasegawa Y, Noguchi T, Mitani A, IL-15 and RANKL play a synergistically important role in osteoclastogenesis, J. Cell. Biochem 118 (4) (2017) 739–747. [DOI] [PubMed] [Google Scholar]
  • [94].Bergmann B, Jirholt P, Henning P, Lindholm C, Ohlsson C, McInnes IB, Lerner UH, Gjertsson I, Antibiotics with interleukin-15 inhibition reduce joint inflammation and bone erosions but not cartilage destruction in Staphylococcus aureus-induced arthritis, Infect. Immun 86 (5) (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [95].Ferrari-Lacraz S, Zanelli E, Neuberg M, Donskoy E, Kim YS, Zheng XX, Hancock WW, Maslinski W, Li XC, Strom TB, Moll T, Targeting IL-15 receptor-bearing cells with an antagonist mutant IL-15/Fc protein prevents disease development and progression in murine collagen-induced arthritis, J. Immunol 173 (9) (2004) 5818–5826. [DOI] [PubMed] [Google Scholar]
  • [96].Takeda H, Kikuchi T, Soboku K, Okabe I, Mizutani H, Mitani A, Ishihara Y, Noguchi T, Effect of IL-15 and natural killer cells on osteoclasts and osteoblasts in a mouse coculture, Inflammation 37 (3) (2014) 657–669. [DOI] [PubMed] [Google Scholar]
  • [97].Feng S, Madsen SH, Viller NN, Neutzsky-Wulff AV, Geisler C, Karlsson L, Soderstrom K, Interleukin-15-activated natural killer cells kill autologous osteoclasts via LFA-1, DNAM-1 and TRAIL, and inhibit osteoclast-mediated bone erosion in vitro, Immunology 145 (3) (2015) 367–379. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [98].Brestoff JR, Kim BS, Saenz SA, Stine RR, Monticelli LA, Sonnenberg GF, Thome JJ, Farber DL, Lutfy K, Seale P, Artis D, Group 2 innate lymphoid cells promote beiging of white adipose tissue and limit obesity, Nature 519 (7542) (2015) 242–246. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [99].Shafiei-Jahani P, Hurrell BP, Galle-Treger L, Helou DG, Howard E, Painter J, Lo R, Lewis G, Soroosh P, Akbari O, DR3 stimulation of adipose resident ILC2s ameliorates type 2 diabetes mellitus, Nat. Commun 11 (1) (2020) 4718. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [100].Rauber S, Luber M, Weber S, Maul L, Soare A, Wohlfahrt T, Lin N-Y, Dietel K, Bozec A, Herrmann M, Resolution of inflammation by interleukin-9-producing type 2 innate lymphoid cells, nature medicine 23 (8) (2017) 938–944. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [101].Chen Z, Andreev D, Oeser K, Krljanac B, Hueber A, Kleyer A, Voehringer D, Schett A Bozec, Th2 and eosinophil responses suppress inflammatory arthritis, Nat. Commun 7 (1) (2016) 1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [102].Omata Y, Frech M, Primbs T, Lucas S, Andreev D, Scholtysek C, Sarter K, Kindermann M, Yeremenko N, Baeten DL, Group 2 innate lymphoid cells attenuate inflammatory arthritis and protect from bone destruction in mice, Cell Rep. 24 (1) (2018) 169–180. [DOI] [PubMed] [Google Scholar]
  • [103].Klose CS, Artis D, Innate lymphoid cells as regulators of immunity, inflammation and tissue homeostasis, Nat. Immunol 17 (7) (2016) 765–774. [DOI] [PubMed] [Google Scholar]
  • [104].Walker JA, McKenzie ANJ, TH2 cell development and function, Nat. Rev. Immunol 18 (2) (2018) 121–133. [DOI] [PubMed] [Google Scholar]
  • [105].Boyce BF, Xing L, Functions of RANKL/RANK/OPG in bone modeling and remodeling, Archives Biochem. Biophys 473 (2) (2008) 139–146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [106].Mangashetti LS, Khapli SM, Wani MR, IL-4 inhibits bone-resorbing activity of mature osteoclasts by affecting NF-kappa B and Ca2+ signaling, J. Immunol 175 (2) (2005) 917–925. [DOI] [PubMed] [Google Scholar]
  • [107].Bendixen AC, Shevde NK, Dienger KM, Willson TM, Funk CD, Pike JW, IL-4 inhibits osteoclast formation through a direct action on osteoclast precursors via peroxisome proliferator-activated receptor gamma 1, Proc. Natl. Acad. Sci. U. S. A 98 (5) (2001) 2443–2448. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [108].Zhang J, Fu Q, Ren Z, Wang Y, Wang C, Shen T, Wang G, Wu L, Changes of serum cytokines-related Th1/Th2/Th17 concentration in patients with postmenopausal osteoporosis, Gynecol. Endocrinol 31 (3) (2015) 183–190. [DOI] [PubMed] [Google Scholar]
  • [109].Yamada A, Takami M, Kawawa T, Yasuhara R, Zhao B, Mochizuki A, Miyamoto Y, Eto T, Yasuda H, Nakamichi Y, Interleukin-4 inhibition of osteoclast differentiation is stronger than that of interleukin-13 and they are equivalent for induction of osteoprotegerin production from osteoblasts, Immunology 120 (4) (2007) 573–579. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [110].Macias MP, Fitzpatrick LA, Brenneise I, McGarry MP, Lee JJ, Lee NA, Expression of IL-5 alters bone metabolism and induces ossification of the spleen in transgenic mice, J. Clin. Invest 107 (8) (2001) 949–959. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [111].Zhou P, Zheng T, Li Y, Zhang X, Feng J, Wei Y, Wang H, Yao Y, Gong F, Tian W, Chlorinated flame-retardant Dechlorane 602 potentiates type 2 innate lymphoid cells and exacerbates airway inflammation, Environ. Sci. Technol 55 (2) (2020) 1099–1109. [DOI] [PubMed] [Google Scholar]
  • [112].Zheng T, Fan M, Wei Y, Feng J, Zhou P, Sun X, Xue A, Qin CX, Yu D, Huangbai liniment ameliorates skin inflammation in atopic dermatitis, Front. Pharmacol 12 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [113].Gong F, Zheng T, Zhou P, T follicular helper cell subsets and the associated cytokine IL-21 in the pathogenesis and therapy of asthma, Front. Immunol 10 (2019) 2918. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [114].Kamanaka M, Kim ST, Wan YY, Sutterwala FS, Lara-Tejero M, Galán JE, Harhaj E, Flavell RA, Expression of interleukin-10 in intestinal lymphocytes detected by an interleukin-10 reporter knockin tiger mouse, Immunity 25 (6) (2006) 941–952. [DOI] [PubMed] [Google Scholar]
  • [115].Saraiva M, O’Garra A, The regulation of IL-10 production by immune cells, Nat. Rev. Immunol 10 (3) (2010) 170–181. [DOI] [PubMed] [Google Scholar]
  • [116].Evans KE, Fox SW, Interleukin-10 inhibits osteoclastogenesis by reducing NFATc1 expression and preventing its translocation to the nucleus, BMC Cell Biol. 8 (1) (2007) 1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [117].Hong MH, Williams H, Jin CH, Pike JW, The inhibitory effect of interleukin-10 on mouse osteoclast formation involves novel tyrosine-phosphorylated proteins, J. Bone Miner. Res 15 (5) (2000) 911–918. [DOI] [PubMed] [Google Scholar]
  • [118].Liu D, Yao S, Wise GE, Effect of interleukin-10 on gene expression of osteoclastogenic regulatory molecules in the rat dental follicle, Eur. J. Oral Sci 114 (1) (2006) 42–49. [DOI] [PubMed] [Google Scholar]
  • [119].Dresner-Pollak R, Gelb N, Rachmilewitz D, Karmeli F, Weinreb M, Interleukin 10-deficient mice develop osteopenia, decreased bone formation, and mechanical fragility of long bones, Gastroenterology 127 (3) (2004) 792–801. [DOI] [PubMed] [Google Scholar]
  • [120].Claudino M, Garlet TP, Cardoso CR, De Assis GF, Taga R, Cunha FQ, Silva JS, Garlet GP, Down-regulation of expression of osteoblast and osteocyte markers in periodontal tissues associated with the spontaneous alveolar bone loss of interleukin-10 knockout mice, Eur. J. Oral Sci 118 (1) (2010) 19–28. [DOI] [PubMed] [Google Scholar]
  • [121].Gür A, Denli A, Nas K, Cevik R, Karakoc M, Sarac A, Erdogan F, Possible pathogenetic role of new cytokines in postmenopausal osteoporosis and changes during calcitonin plus calcium therapy, Rheumatol. Int 22 (5) (2002) 194–198. [DOI] [PubMed] [Google Scholar]
  • [122].Chen H-Y, Chen W-C, Hsu C-M, Tsai F-J, Tsai C-H, Tumor necrosis factor α, CYP 17, urokinase, and interleukin 10 gene polymorphisms in postmenopausal women: correlation to bone mineral density and susceptibility to osteoporosis, Eur. J. Obstet. Gynecol. Reprod. Biol 122 (1) (2005) 73–78. [DOI] [PubMed] [Google Scholar]
  • [123].Park B-L, Han I-K, Lee H-S, Kim L-H, Kim S-J, Shin J-S, Kim S-Y, Shin H-B, Association of interleukin 10 haplotype with low bone mineral density in Korean postmenopausal women, BMB Rep. 37 (6) (2004) 691–699. [DOI] [PubMed] [Google Scholar]
  • [124].Spolski R, Li P, Leonard WJ, Biology and regulation of IL-2: from molecular mechanisms to human therapy, Nat. Rev. Immunol 18 (10) (2018) 648–659. [DOI] [PubMed] [Google Scholar]
  • [125].Raffin C, Vo LT, Bluestone JA, Treg cell-based therapies: challenges and perspectives, Nat. Rev. Immunol 20 (3) (2020) 158–172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [126].Takayanagi H, Kim S, Koga T, Taniguchi T, Stat1-mediated cytoplasmic attenuation in osteoimmunology, J. Cell. Biochem 94 (2) (2005) 232–240. [DOI] [PubMed] [Google Scholar]
  • [127].Coelho LF, de Freitas Almeida G. Magno, Mennechet FJ, Blangy A, Uze G, Interferon-alpha and -beta differentially regulate osteoclastogenesis: role of differential induction of chemokine CXCL11 expression, Proc. Natl. Acad. Sci. U. S. A 102 (33) (2005) 11917–11922. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [128].Inoue K, Deng Z, Chen Y, Giannopoulou E, Xu R, Gong S, Greenblatt MB, Mangala LS, Lopez-Berestein G, Kirsch DG, Sood AK, Zhao L, Zhao B, Bone protection by inhibition of microRNA-182, Nat. Commun 9 (1) (2018) 4108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [129].Binder N, Miller C, Yoshida M, Inoue K, Nakano S, Hu X, Ivashkiv LB, Schett G, Pernis A, Goldring SR, Ross FP, Zhao B, Def6 restrains osteoclastogenesis and inflammatory bone resorption, J. Immunol 198 (9) (2017) 3436–3447. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [130].Vignali DA, Kuchroo VK, IL-12 family cytokines: immunological playmakers, Nat. Immunol 13 (8) (2012) 722–728. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [131].Nagata N, Kitaura H, Yoshida N, Nakayama K, Inhibition of RANKL-induced osteoclast formation in mouse bone marrow cells by IL-12: involvement of IFN-gamma possibly induced from non-T cell population, Bone 33 (4) (2003) 721–732. [DOI] [PubMed] [Google Scholar]
  • [132].Kitaura H, Nagata N, Fujimura Y, Hotokezaka H, Yoshida N, Nakayama K, Effect of IL-12 on TNF-alpha-mediated osteoclast formation in bone marrow cells: apoptosis mediated by Fas/Fas ligand interaction, J. Immunol 169 (9) (2002) 4732–4738. [DOI] [PubMed] [Google Scholar]
  • [133].Yoshimatsu M, Kitaura H, Fujimura Y, Kohara H, Morita Y, Yoshida N, IL-12 inhibits lipopolysaccharide stimulated osteoclastogenesis in mice, J Immunol Res 2015 (2015), 214878. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [134].Yoshimatsu M, Kitaura H, Fujimura Y, Eguchi T, Kohara H, Morita Y, Yoshida N, IL-12 inhibits TNF-alpha induced osteoclastogenesis via a T cell-independent mechanism in vivo, Bone 45 (5) (2009) 1010–1016. [DOI] [PubMed] [Google Scholar]
  • [135].Kalliolias GD, Zhao B, Triantafyllopoulou A, Park-Min KH, Ivashkiv LB, Interleukin-27 inhibits human osteoclastogenesis by abrogating RANKL-mediated induction of nuclear factor of activated T cells c1 and suppressing proximal RANK signaling, Arthritis Rheum. 62 (2) (2010) 402–413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [136].Furukawa M, Takaishi H, Takito J, Yoda M, Sakai S, Hikata T, Hakozaki A, Uchikawa S, Matsumoto M, Chiba K, Kimura T, Okada Y, Matsuo K, Yoshida Y Toyama, IL-27 abrogates receptor activator of NF-kappa B ligand-mediated osteoclastogenesis of human granulocyte-macrophage colony-forming unit cells through STAT1-dependent inhibition of c-Fos, J. Immunol 183 (4) (2009) 2397–2406. [DOI] [PubMed] [Google Scholar]
  • [137].Shukla P, Mansoori MN, Kakaji M, Shukla M, Gupta SK, Singh D, Interleukin 27 (IL-27) alleviates bone loss in estrogen-deficient conditions by induction of early growth response-2 gene, J. Biol. Chem 292 (11) (2017) 4686–4699. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [138].Ji J-D, Park-Min K-H, Shen Z, Fajardo RJ, Goldring SR, McHugh KP, Ivashkiv LB, Inhibition of RANK expression and osteoclastogenesis by TLRs and IFN-γ in human osteoclast precursors, J. Immunol 183 (11) (2009) 7223–7233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [139].Wang L, Liu S, Zhao Y, Liu D, Liu Y, Chen C, Karray S, Shi S, Jin Y, Osteoblast-induced osteoclast apoptosis by fas ligand/FAS pathway is required for maintenance of bone mass, Cell Death Different. 22 (10) (2015) 1654–1664. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [140].Van’t Hof RJ, Ralston SH, Cytokine-induced nitric oxide inhibits bone resorption by inducing apoptosis of osteoclast progenitors and suppressing osteoclast activity, J. Bone Miner. Res 12 (11) (1997) 1797–1804. [DOI] [PubMed] [Google Scholar]
  • [141].Kim J-W, Lee M-S, Lee C-H, Kim H-Y, Chae S-U, Kwak H-B, Oh J-M, Effect of interferon-γ on the fusion of mononuclear osteoclasts into bone-resorbing osteoclasts, BMB Rep. 45 (5) (2012) 281–286. [DOI] [PubMed] [Google Scholar]
  • [142].Duque G, Huang DC, Dion N, Macoritto M, Rivas D, Li W, Yang XF, Li J, Lian J, Marino FT, Interferon-γ plays a role in bone formation in vivo and rescues osteoporosis in ovariectomized mice, J. Bone Miner. Res 26 (7) (2011) 1472–1483. [DOI] [PubMed] [Google Scholar]
  • [143].Ries WL, Seeds MC, Key LL, Interleukin-2 stimulates osteoclastic activity; increased acid production and radioactive calcium release, J. Periodontal Res 24 (4) (1989) 242–246. [DOI] [PubMed] [Google Scholar]
  • [144].Ashcroft A, Cruickshank S, Croucher P, Perry M, Rollinson S, Lippitt J, Child J, Dunstan C, Felsburg P, Morgan G, Colonic dendritic cells, intestinal inflammation, and T cell-mediated bone destruction are modulated by recombinant osteoprotegerin, Immunity 19 (6) (2003) 849–861. [DOI] [PubMed] [Google Scholar]
  • [145].Palmqvist P, Persson E, Conaway HH, Lerner UH, IL-6, leukemia inhibitory factor, and oncostatin M stimulate bone resorption and regulate the expression of receptor activator of NF-κΒ ligand, osteoprotegerin, and receptor activator of NF-κB in mouse calvariae, J. Immunol 169 (6) (2002) 3353–3362. [DOI] [PubMed] [Google Scholar]
  • [146].Kudo O, Sabokbar A, Pocock A, Itonaga I, Fujikawa Y, Athanasou N, Interleukin-6 and interleukin-11 support human osteoclast formation by a RANKL-independent mechanism, Bone 32 (1) (2003) 1–7. [DOI] [PubMed] [Google Scholar]
  • [147].Yoshitake F, Itoh S, Narita H, Ishihara K, Ebisu S, Interleukin-6 directly inhibits osteoclast differentiation by suppressing receptor activator of NF-κB signaling pathways, J. Biol. Chem 283 (17) (2008) 11535–11540. [DOI] [PubMed] [Google Scholar]
  • [148].FDA, in: FDA (Ed.), https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/125276s114lbl.pdf, 2010. [Google Scholar]
  • [149].Chen Y-M, Chen H-H, Huang W-N, Liao T-L, Chen J-P, Chao W-C, Lin C-T, Hung W-T, Hsieh C-W, Hsieh T-Y, Tocilizumab potentially prevents bone loss in patients with anticitrullinated protein antibody-positive rheumatoid arthritis, PLoS One 12 (11) (2017), e0188454. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [150].Chen L, Wei XQ, Evans B, Jiang W, Aeschlimann D, IL-23 promotes osteoclast formation by up-regulation of receptor activator of NF-kappaB (RANK) expression in myeloid precursor cells, Eur. J. Immunol 38 (10) (2008) 2845–2854. [DOI] [PubMed] [Google Scholar]
  • [151].Shin HS, Sarin R, Dixit N, Wu J, Gershwin E, Bowman EP, Adamopoulos IE, Crosstalk among IL-23 and DNAX activating protein of 12 kDa-dependent pathways promotes osteoclastogenesis, J. Immunol 194 (1) (2015) 316–324. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [152].Iwakura Y, Ishigame H, The IL-23/IL-17 axis in inflammation, J. Clin. Invest 116 (5) (2006) 1218–1222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [153].Yago T, Nanke Y, Kawamoto M, Furuya T, Kobashigawa T, Kamatani N, Kotake S, IL-23 induces human osteoclastogenesis via IL-17 in vitro, and anti-IL-23 antibody attenuates collagen-induced arthritis in rats, Arthritis Res. Ther 9 (5) (2007) R96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [154].Quinn JM, Sims NA, Saleh H, Mirosa D, Thompson K, Bouralexis S, Walker EC, Martin TJ, Gillespie MT, IL-23 inhibits osteoclastogenesis indirectly through lymphocytes and is required for the maintenance of bone mass in mice, J. Immunol 181 (8) (2008) 5720–5729. [DOI] [PubMed] [Google Scholar]
  • [155].Kamiya S, Nakamura C, Fukawa T, Ono K, Ohwaki T, Yoshimoto T, Wada S, Effects of IL-23 and IL-27 on osteoblasts and osteoclasts: inhibitory effects on osteoclast differentiation, J. Bone Miner. Metab 25 (5) (2007) 277–285. [DOI] [PubMed] [Google Scholar]
  • [156].Kavanaugh A, Ritchlin C, Rahman P, Puig L, Gottlieb AB, Li S, Wang Y, Noonan L, Brodmerkel C, Song M, Mendelsohn AM, McInnes IB, P. Summit, G. Study, Ustekinumab, an anti-IL-12/23 p40 monoclonal antibody, inhibits radiographic progression in patients with active psoriatic arthritis: results of an integrated analysis of radiographic data from the phase 3, multicentre, randomised, double-blind, placebo-controlled PSUMMIT-1 and PSUMMIT-2 trials, Ann. Rheum. Dis 73 (6) (2014) 1000–1006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [157].Bommireddy R, Engle SJ, Ormsby I, Boivin GP, Babcock GF, Doetschman T, Elimination of both CD4+ and CD8+ T cells but not B cells eliminates inflammation and prolongs the survival of TGFβ1-deficient mice, Cell. Immunol 232 (1–2) (2004) 96–104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [158].Crane JL, Cao X, Bone marrow mesenchymal stem cells and TGF-β signaling in bone remodeling, J. Clin. Invest 124 (2) (2014) 466–472. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [159].Kasagi S, Chen W, TGF-beta1 on osteoimmunology and the bone component cells, Cell Biosci. 3 (1) (2013) 1–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [160].Wu M, Chen G, Li YP, TGF-beta and BMP signaling in osteoblast, skeletal development, and bone formation, homeostasis and disease, Bone Res. 4 (2016) 16009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [161].Quinn JM, Itoh K, Udagawa N, Hausler K, Yasuda H, Shima N, Mizuno A, Higashio K, Takahashi N, Suda T, Martin TJ, Gillespie MT, Transforming growth factor beta affects osteoclast differentiation via direct and indirect actions, J. Bone Miner. Res 16 (10) (2001) 1787–1794. [DOI] [PubMed] [Google Scholar]
  • [162].Yasui T, Kadono Y, Nakamura M, Oshima Y, Matsumoto T, Masuda H, Hirose J, Omata Y, Yasuda H, Imamura T, Nakamura K, Tanaka S, Regulation of RANKL-induced osteoclastogenesis by TGF-beta through molecular interaction between Smad3 and Traf6, J. Bone Miner. Res 26 (7) (2011) 1447–1456. [DOI] [PubMed] [Google Scholar]
  • [163].Lee B, Oh Y, Jo S, Kim T-H, Ji JD, A dual role of TGF-β in human osteoclast differentiation mediated by Smad1 versus Smad3 signaling, Immunol. Lett 206 (2019) 33–40. [DOI] [PubMed] [Google Scholar]
  • [164].Houde N, Chamoux E, Bisson M, Roux S, Transforming growth factor-beta1 (TGF-beta1) induces human osteoclast apoptosis by up-regulating Bim, J. Biol. Chem 284 (35) (2009) 23397–23404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [165].Kaneda T, Nojima T, Nakagawa M, Ogasawara A, Kaneko H, Sato T, Mano H, Kumegawa M, Hakeda Y, Endogenous production of TGF-beta is essential for osteoclastogenesis induced by a combination of receptor activator of NF-kappa B ligand and macrophage-colony-stimulating factor, J. Immunol 165 (8) (2000) 4254–4263. [DOI] [PubMed] [Google Scholar]
  • [166].Karst M, Gorny G, Galvin RJ, Oursler MJ, Roles of stromal cell RANKL, OPG, and M-CSF expression in biphasic TGF-beta regulation of osteoclast differentiation, J. Cell. Physiol 200 (1) (2004) 99–106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [167].Xia Y, Inoue K, Du Y, Baker SJ, Premkumar Reddy E, Greenblatt MB, Zhao B, TGFbeta reprograms TNF stimulation of macrophages towards a non-canonical pathway driving inflammatory osteoclastogenesis, Nat. Commun 13 (1) (2022) 3920. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [168].Zhen G, Wen C, Jia X, Li Y, Crane JL, Mears SC, Askin FB, Frassica FJ, Chang W, Yao J, Inhibition of TGF-β signaling in mesenchymal stem cells of subchondral bone attenuates osteoarthritis, Nat. Med 19 (6) (2013) 704–712. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES