info@biomedres.us   +1 (502) 904-2126   One Westbrook Corporate Center, Suite 300, Westchester, IL 60154, USA   Site Map
ISSN: 2574 -1241

Impact Factor : 0.548

  Submit Manuscript

Review ArticleOpen Access

The Place of Osteoimmunology in the Pathogenesis of Rheumatic Joint and Bone Diseases Volume 50- Issue 1

Momcheva I*, Kazmin I and Staykov D

  • University Multiprofile Hospital for Active Treatment Burgas, Bulgaria

Received: March 10, 2023;   Published: April 27, 2023

*Corresponding author: Momcheva I, University Multiprofile Hospital for Active Treatment Burgas, Bulgaria

DOI: 10.26717/BJSTR.2023.50.007894

Abstract PDF

ABSTRACT

The bone and immune cells all share the same microenvironment, interact with each other, share common signaling pathways, collaborate, performing the functions of an “osteoimmune system”. The same cytokines may have different and often opposite effects depending on the specific environment in which they function, the maturation stage of the target cells and/or the influence of other cytokines. Through osteoimmunology, the intimate mechanisms in the pathogenesis of many rheumatological diseases such as Rheumatoid arthritis, axSpA, autoinflammatory diseases, osteonecrosis, osteoarthrosis, osteoporosis are clarified. Osteoimmunology is a conceptual framework for decoding the complex language through which the immune system and bone communicate. This review summarizes the data accumulated to date on the interactions between the immune and bone systems of the human organism and reveals the bidirectionality of these interactions and their role in the pathogenesis of rheumatic joint and bone diseases.

Keywords: Osteoimmunology; Cytokines; Rheumatic Joint and Bone Diseases; Signal Pathways

Introduction: the «Embrace» of the Immune and Bone Systems

The skeletal system and the adaptive immune system developed simultaneously during the evolution of vertebrates. The implication is that certain components of the skeletal system are essential for the proper operation of the immune system (Boehm 2012). About 385 million years ago, the aquatic vertebrates moved to land and the skeletal system evolved to maintain the motor activity in terrestrial conditions. The adaptive immune system first appears in cartilaginous fish. The nuclear factor receptor activator ligand - κB (RANKL) is known from cartilaginous fish (OrthoDB database). The osteoclasts first appear in bony fish. Active bone metabolism, regulated by calciotropic hormones such as vitamin D and parathyroid hormone, has been known since the advent of amphibians. Immunoglobulin classes and lymph nodes are found only in terrestrial animals. The bone marrow serves as a site for communication and collaboration between bone and immune cells, working together to carry out crucial tasks such as strengthening the body, regulating mineral metabolism, and facilitating hematopoiesis (the production of blood cells) (Morrison and Scadden 2014). The concept of osteoimmunology originated from initial research demonstrating that immune cells secrete factors that activate osteoclasts (Horton et al. 1972; Mundy et al. 1974). Similar to other multifunctional cytokines in the TNF superfamily, the influence of RANKL extends beyond its role in regulating bone remodeling. It also plays a part in immune responses and the formation of immune organs. The RANKL/RANK pathway plays a critical role in the formation of important immune organs in mammals, including the thymus and lymph nodes. Additionally, the bone marrow houses hematopoietic stem cells (HSCs) [1]. The bone marrow contains hematopoietic stem cells (HSCs), myeloid and lymphoid progenitors, as well as mature immune cells, neutrophils, macrophages and T lymphocytes. Bone and immune cells share the same microenvironment and interact with each other, cooperating, performing the functions of an «osteoimmune system». In 2000, the term «osteoimmunology» was proposed by Arron and Choi to emphasize the T-lymphocyte-controlled regulation of osteoclastogenesis in the context of rheumatoid arthritis. Subsequently, it has become evident that the immune and skeletal systems are subject to shared regulation by a variety of cytokines, chemokines, transcription factors, and signaling molecules. In addition, the evidence suggests that bone cells reciprocally regulate immune cells and hematopoiesis [1].

Accumulated scientific evidence on the communication between immune and bone cells has led to a revised understanding of bone remodeling. This new model posits that the phases of bone resorption and formation, which are in a constant state of balance, are subject to precise immunological regulation [2]. The skeletal and immune systems are intricately interconnected through intricate networks that work together to maintain homeostasis. Bone is a constantly changing tissue comprised of bone proteins that are infused with mineral crystals and interwoven with bone cells such as osteocytes (OCs), osteoblasts (OBs), and osteoclasts (OCLs). The osteoblast, which derives from the mesenchymal stem cell (MSC), has the potential to differentiate into other cell types such as chondrocytes, bone marrow stromal cells, and adipocytes [3]. Osteoclasts (OCLs) are specialized myeloid cells with multiple nuclei that are responsible for breaking down mineralized bone tissue through the secretion of lysosomal enzymes like tartrate-resistant acid phosphatase (TRAP) and cathepsin K [4]. OCL originates from a progenitor cell in the bone marrow, which allows differentiation to «professional» antigen-presenting cell (APC), i.e. dendritic cell and macrophage. Based on that data, OCL can also be considered a specialized immune cell. Under normal circumstances, osteoblasts (OBs), osteoclasts (OCLs), and osteocytes (OCs) engage in continuous communication with one another to maintain optimal bone quality and quantity in a homeostatic manner. Osteoblasts (OBs) secrete various signaling molecules such as macrophage colony-stimulating factor (MCSF), RANKL, and other stimulatory factors to direct the differentiation of myeloid lineage progenitor cells into osteoclasts (OCLs) [5]. The receptor activator of nuclear factor Kappa B (RANK) and its ligand (RANKL), along with the nuclear factor of activated T-cell (NFATc1), are essential for the communication between osteoclasts (OCLs) and osteoblasts (OBs). The expressed on the osteoclast precursor RANK interacts with RANKL and forms the RANK-RANKL complex, which activates the Wnt signaling pathway, leading to the stimulation of maturation, differentiation and activation of osteoclasts and inhibits their apoptosis. The activated T cells produce RANKL, which activates OCL, which is not followed by activation of OB, i.e. bone formation did not follow witch is the so-called pathological remodeling.

Other potential inducers of RANKL are the pro-inflammatory cytokines: TNFα, IL 1, 6, 17, VEGF (vascular endothelial growth factor). This association between pro-inflammatory cytokines and osteoclast formation explains why cytokine-targeted therapy delays structural bone damage in IMIDs. It is worth noting that RANKL and RANK also have a significant impact on the development of the lactating mammary gland during pregnancy, highlighting their pleiotropic effects [6]. Additionally, osteocytes (OCs), which were once believed to solely regulate bone remodeling, are now known to regulate immune cells and form the «endosteal niche.» Both osteoblasts (OBs) and osteoclasts (OCLs) play a role in the formation of the endosteal niche, which mobilizes hematopoietic stem cells (HSCs) [7]. Studies have shown that RANKL, which is produced by osteocytes (OCs), contributes to increased osteoclastogenesis and bone loss, as observed in estrogen-deficient conditions. Additionally, there is evidence supporting a link between OCs and the immune system, as in vivo ablation of OCs has been found to result in severe lymphopenia, according to Sato and colleagues. The conversation the relationship between the immune system and bones works in both directions, indicating that immune cells can also be impacted by bone cells. OCL has been shown to regulate the HSC niche directly and indirectly through OB. First, OCLs can increase the mobilization of HSCs by secreting cathepsin K, an important protein for their function. As a result, HSCs enter the circulation. As is known, the differentiation of OCL strictly depends on the RANKL / RANK path [8,9]. Studies have demonstrated that OCL (osteoclasts) can control the HSC (hematopoietic stem cell) niche both directly and indirectly by interacting with the interaction between RANKL and RANK, which are expressed by OCL precursors, leads to the recruitment of TNFR-associated factors (TRAFs). These TRAFs then trigger the differentiation of OCL by promoting the translocation of NF-kB to activated B and T cells. RANKL is also produced by activated T lymphocytes. The significance of RANKL in the context of immunology is highlighted by the fact that mice deficient in RANKL not only exhibit a bone phenotype characterized by osteopetrosis due to the absence of osteoclasts, but also display immunological abnormalities such as impaired lymphocyte development and lack of lymph node organogenesis.

Shared Signal Pathways

Cytokines and transcription factors, which act as mediators, are involved in both inflammation and bone metabolism. RANKL is expressed by immune system cells, particularly activated T and B lymp. TNF-α, IL-1, IL-6, and IL-17 are inflammatory cytokines that play a critical role in acute and chronic inflammation. These cytokines are potent stimulators of bone resorption. T-lymphocytes present in the bone marrow are important immune cells that play a crucial role in regulating bone remodeling. Inflammatory cytokines produced by activated T lymphocytes induce bone resorption during inflammatory diseases or conditions characterized by lowgrade systemic inflammation. T helper (Th17) cells are involved in stimulating bone resorption and have a significant impact on bone loss observed in inflammatory diseases such as psoriasis, rheumatoid arthritis, periodontitis, Crohn’s disease, and chronic ulcerative colitis. Th17 cells promote osteoclastogenesis by producing cytokines such as IL-17, RANKL, TNF-α, IL-1, and IL-6, in addition to low levels of IFN-γ. IL-17 triggers the release of RANKL from both osteoblasts (OBs) and osteoclasts (OCs), thereby activating RANK signaling in osteoclasts (OCLs). Conversely, T regulatory (Treg) cells inhibit osteoclastogenesis and promote bone formation. The osteoclastassociated receptor (OSCAR) not only facilitates interactions between osteoblasts (OB) and osteoclasts (OCL), but it also plays a role in regulating both adaptive and innate immunity [10]. OSCAR, initially identified as a regulator of osteoclast differentiation and an immunomodulatory mediator in bone, is believed to be involved in cellular activation and inflammation in atherosclerosis [11]. TNF-α promotes the expression of OSCAR and other receptors on the surface of monocytoid peripheral blood cells that play a crucial role in the differentiation of osteoclasts (OCLs) [12]. Cytokines have inherent pleiotropic functions, and it is not surprising that the same cytokines may have different and even opposing effects depending on various factors such as the specific environment in which they operate, the stage of maturation of target cells, and the influence of other cytokines. It is seen that not only do immune system cells regulate bone remodeling, but also bone cells are able to affect the immune system [13-15].

Key Cellular and Humoral Participants in the Cross Communication Between the Immune System and the Bones

T cells are a key component of the adaptive immunity. The links between T cells and the bone biology are numerous: essentially all T cell subtypes are able to affect bone cells (mostly OCLs). However, the role of Th17 and T-reg cells is particularly important. Th17 cells are thought to be the majority of osteoclastogenesis-inducing T cells. These cells stimulate the expression of macrophage-colonystimulating factor (M-CSF) and RANKL in osteoblasts and stromal cells, resulting in the production of RANKL and TNF-α. Furthermore, they increase the expression of RANK in osteoclast precursor cells [16]. These characteristics make them potent inducers of osteoclastogenesis, which is why they have already been described as «players» in the bone lesions in RA [17] and multiple myeloma [18]. The role of T-reg cells in inhibiting osteoclastogenesis is well established. This is achieved through both soluble factor-mediated mechanisms as well as contact-mediated mechanisms [19]. Dendritic cells (DCs) are a type of antigen-presenting cells that play a crucial role in directing cell-mediated immunity towards appropriate targets with speed and accuracy while preventing autoimmunity [20]. Historically, the role of dendritic cells (DCs) in bone biology has been considered to be indirect, primarily through their interaction with T cells [21]. Later studies have shown that dendritic cells not only present antigens to T cells but also play a role in regulating the activity and balance of T cell subtypes through cytokine signaling [22]. The common myeloid origin of DCs and OCLs should not be overlooked. Neutrophils are also involved in bone biology, particularly in inflammation-induced bone loss [23]. Neutrophils are typically the first type of immune cell to migrate to the site of bone damage, where they release chemokines, cytokines, and other small molecules that act as immunomodulatory factors. The secretion of chemokines CCL2 and CCL20 by neutrophils attracts Th17 cells, which contributes to bone loss. However, the absence of neutrophils can also result in bone loss due to the activation of IL-17-mediated local inflammation [24].

Activated neutrophils express RANKL at the site of inflammation, which leads to their active involvement in osteoclastogenesis. This process can increase juxta-articular osteoporosis associated with rheumatoid arthritis [25]. In summary, however, the role of neutrophils in osteoimmunology is not fully understood, the general consensus is that activated neutrophils are inducers of osteoclastogenesis, directly and indirectly. B-cells: B-cell development is regulated by various factors, including RANKL, OPG, IL-7, and CXCL12, which are secreted by bone marrow stromal cells and osteoblasts [26]. New research indicates that B cells not only rely on RANKL for their development, but they also produce RANKL and use it as an autocrine signaling molecule [27]. Natural killer (NK) cells: Natural killer (NK) cells participate in the regulation of bone homeostasis along with other lymphocytes. NK cells play a role in regulating bone homeostasis and are also involved in the pathogenesis of bone damage in rheumatoid arthritis by inducing osteoblast cell death [28]. NK cells have a dual role in RA-induced bone loss as they can induce OB cell death, making them a potential therapeutic target for reducing bone loss [29]. However, NK cells are also necessary for delaying RA progression, which raises doubts about the effectiveness of anti-NK therapy in RA. Inflammation and inflammatory factors: IFN-γ, produced by various immune system cells such as T and B cells, NK cells, monocytes/ macrophages, and dendritic cells, plays a crucial role in both innate and adaptive immunity as well as in the regulation of inflammation [30,31]. In bones, IFN-γ affects both OB and OCL. IFN-γ has a positive effect on OBs, which usually produce low levels of this cytokine. This is because IFN-γ can activate genes that are involved in osteoblast differentiation. IFN-γ has been found to have an inhibitory effect on the differentiation of osteoclasts (OCLs) by counteracting the effects of M-CSF on OCL precursors [31]. It does so by reducing the expression of c-fms receptor, which leads to a reduced number of pre-OCLs that are positive for RANK [32]. In addition, IFN- γ induces osteoclast apoptosis. The importance of IFN type I in bone homeostasis was underscored by the observation that mice deficient in type I-IFNreceptor component (IFNAR1) spontaneously developed osteopenia accompanied by enhanced osteoclastogenesis [33].

Both types of interferons (IFNs) inhibit osteoclastogenesis through the activation of the signal transducer and activator of transcription 1 (STAT1) in the skeletal system [34]. In the skeletal system, both types of IFN inhibit osteoclastogenesis by STAT1. INF-γ also inhibits the effect of PTH and IL-1 on stimulating OCL formation in bone marrow cultures [35]. INF-γ inhibits RANK signaling but does not directly inhibit bone resorption from mature OCLs [36]. It has been reported that INF-γ can also stimulate bone resorption by enhancing the production of RANKL and TNF-α in T lymphocytes [37]. Inflammatory cytokines, which are produced mainly by macrophages such as IL-1, TNF and IL-6, stimulate osteoclastogenesis. They are called osteoclastogenic cytokines because of their resorptive effect on the bone [38]. IL-1 stimulates TRAF6 (and therefore activates NF- κB and MAPKs) and synergizes with RANKL to induce mature OCL. Activated T cells expressing RANKL have the potential to induce osteoclast differentiation by directly affecting osteoclast progenitor cells. T cells also secrete various anti-inflammatory cytokines such as IL-4, so the effects of T cells on the osteoclastogenesis depend on the balance between pro- and anti-inflammatory factors produced by them. For example, CD4 + T helper cell subgroups Th1 and Th2 produce IFN-γ and IL-4 with an anti-osteoclastogenic effect. Recently, researchers have been considering the role of T cells in non-inflammatory metabolic bone diseases and postmenopausal osteoporosis. TNF and other inflammatory cytokines not only cause local inflammation but also play a direct and indirect role in activating OCLs. Such osteoimmunological pleiotropy may explain the efficacy of RANKL-specific antibody not only in postmenopausal osteoporosis but in rheumatoid arthritis and also its preventive effect on the bone metastases [39]. TNF-α stimulates the formation of OCL and enhances the bone resorption in vivo [40-42]. The ability of TNF-α to stimulate OCL formation from osteoclast precursor is dependent on IL-1 [43], while TNF-α-induced osteolysis is dependent on M-CSF [44].

The results of studies with RANK-deficient mouse cell cultures show that TNF-α directly stimulates the formation of OCL, independent of RANK [45]. TNF-α also inhibits osteoblast differentiation and collagen synthesis [46-48]. In addition, TNF-α is highly proapoptotic to osteoblasts [49], possibly by signaling Fas-Fas ligand (FasL). Fas ligand (FasL), also known as CD95L or CD178, is a protein that is found on the surface of cells and belongs to the TNF family. When FasL binds to its receptor, it can induce a process called apoptosis, which is a type of programmed cell death. The normal bone development does not seem to be affected by TNF-α, as the absence of abnormal bone phenotypes has been observed in TNF receptor 1 and TNF receptor 2. However, TNF-α can affect bone in inflammatory conditions. IL-7, which plays roles in B- and T-cell lymphopoiesis, also regulates the bone homeostasis [50]. The mechanisms by which IL-7 affects bone cells are controversial. The systemic application of IL-7 stimulates the creation of OCLs by boosting the production of cytokines that promote osteoclastogenesis in T cells [51]. IL-8 is a chemokine of the CXC family, which is produced by osteoclasts. It has been found to stimulate the process of osteoclastogenesis and bone resorption, and this effect is not dependent on the RANKL pathway [52,53]. Activated T and B lymphocytes produce IL-10, which has a direct inhibitory effect on the differentiation of osteoclasts and osteoblasts. IL-17 and IL-23 belong to a group of six cytokines called the IL-17 family, which are critical for the adaptive immune system’s response [54]. They are produced by a specific subset of CD4+ T lymphocytes called Th17, and they have a strong ability to promote the formation of osteoclasts that rely on IL-17 [55]. IL-17A promotes the development of osteoclasts in cultures that include both hematopoietic cells and osteoblasts, by inducing the production of prostaglandins and increasing the expression of RANKL [56]. IL-23 is linked to IL-12 in its effect, as both cytokines are critical for the differentiation and proliferation of Th17 cells, along with TGF-β and IL-6 [57]. IL-18, which belongs to the IL-1 superfamily, is found to be elevated in inflamed areas such as those affected by rheumatoid arthritis. While osteoblasts produce IL-18, it can inhibit the formation of osteoclasts through different mechanisms. This includes an increase in the expression of GM-CSF in T cells. IL-18 has been shown to stimulate the production of INF-γ in OB cultures in vitro.

The inhibitory effect of IFN-γ on osteoclastogenesis is further enhanced by the presence of IL-12. Studies have demonstrated that IL-18 can increase the production of osteoprotegerin (OPG). The effects of IL-18 on osteoclastogenesis are indirect and mediated through its effects on T lymphocytes [58]. IL-13 and IL-4 decrease bone resorption that is stimulated by IL-1 by reducing the levels of prostaglandin and the activity of cyclooxygenase-2. Toll-like receptor stimulators (TLRs): TLRs (Toll-like receptors) play a crucial role in initiating innate immune responses and are prominently expressed on cells responsible for presenting antigens, such as macrophages and dendritic cells [59]. Since macrophages, dendritic cells, and OCLs have common progenitors, it is not surprising that TLRs are also found in the bone cells. Direct TLR signaling on OCL precursors, including TLR4, inhibits RANKL-mediated osteoclastogenesis [60], which seems controversial as bacterial infections cause inflammation by inducing proinflammatory cytokines in response to TLR ligands [60]. Although TLR stimulation inhibits osteoclast differentiation, TLR ligand treated OCL precursors retain pronounced phagocytic activity. Therefore, TLR stimulation of OCL precursors is likely to differentiate them into non-immune cells, such as mature OCLs. Cathepsin K is a cysteine protease identified in OCLs. Cathepsin K inhibition was recently reported to suppress not only osteoclastic bone resorption but also the autoimmune inflammation. Further studies revealed a role for cathepsin K in regulating Th17 differentiation by mediating TLR9 activation of dendritic cells and the production of cytokines such as IL-6 and IL-23 [61]. This is another example of a molecule originally found in the bones that was later shown to play a role in regulating the immune system. Cathepsin K is a key osteoclast collagenase. The signal for OCL migration to the future site of resorption are the microfractures, as a result of which OCs matrix metalloproteases enter the extracellular matrix of the bone. Cytokines such as RANKL, OPG, M-CSF and TGFα are released under the action of these metalloproteases. Specific products of matrix protein proteolysis by metalloproteases serve as signals for OCLs attachment. After the attachment, OCLs change their functional activity and switch to bone resorption. The primary enzyme involved in bone resorption is cathepsin K. Cathepsin K is expressed in pre-OCL, epithelium of bronchi, bile ducts and thyroid gland, chondrocytes and smooth muscle cells of the arteries affected by atherosclerosis.

Cathepsin K is able to cleave many proteins: elastin, gelatin, osteopontin, osteonectin, collagen, aggrecan. A characteristic difference between cathepsin K and other proteases that only cleave collagen telopeptides is that it is also capable of cleaving 3-helix collagen [62]. In the field of osteoimmunology discovering of new molecules continues in recent years. This is the case with another bone mass regulator that has been discussed in the last few years: LipoCaliN-2 (Lcn2). The protein, known as lipocalin, is connected with neutrophil gelatinase (NGAL) as it has the ability to attach to and stabilize the MMP9 factor, which is responsible for the movement of neutrophils out of the bloodstream and into tissues (extravasation). Furthermore, Lcn2 is upregulated during inflammation, and its function in inflammatory disorders still needs to be investigated. This molecule’s role in innate immunity is clear. In vitro mechanical overburden resulted in the upregulation of Lcn2 in osteoblasts, as demonstrated in a study conducted in 2009 [63]. It was unexpected that removing the production of Lcn2 genetically actually leads to a decrease in bone mass instead of an increase, as it was previously believed that Lcn2 was detrimental to bone health. This paradoxical effect is thought to be caused by Lcn2 overexpression damaging osteoblasts and interfering with their energy balance, ultimately leading to osteoblast dysfunction [64]. Osteoimmunology clarifies the intimate mechanisms in the pathogenesis of various diseases such as rheumatoid arthritis [65], ax SpA, autoinflammatory diseases, osteonecrosis, recovery from bone fractures, myelodysplasia, leukemia, neoplasms, metabolic syndrome. The field of osteoimmunology provides a novel perspective for investigating bone-related disorders like osteoporosis (OP) and osteoarthritis (OA), which have not conventionally been recognized as inflammatory conditions [66]. Understanding the intricate communication between the immune [67-73] system and bones is crucial for identifying potential therapeutic targets in various rheumatic and non-rheumatic diseases that involve [74-82] similar mediators and signaling pathways [83-92]. This review summarizes the currently accumulated data on the mutual «embrace» of the immune and skeletal systems of the human body [93-102]. And as it is with any hug, there is a two-way interaction [103-116]. All this again tells us that when studying human health in depth, we must always strive to reveal the integrity and interconnectedness of the processes in the human body in health and disease [117-130].

References

  1. Masayuki Tsukasaki, Hiroshi Takayanagi* Nature Reviews | Immunology, Osteoimmunology: evolving concepts in bone–immune interactions in health and disease.
  2. Teitelbaum S L (2013) Bone resorption by osteoclasts. Science, 2000, 289(5484): 1504-1508. Greenblatt M B, Shim J H, Osteoimmunology: a brief introduction. Immune Netw 13(4): 111-115.
  3. Kazuo Okamoto, Hiroshi Takayanagi, Department of Osteoimmunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo: 113-0033 Japan,Osteoimmunology.
  4. Panwar P, S e K, Guido R V, Bueno R V, Delaisse J M, et al. (2016) A novel approach to inhibit bone resorption: exosite inhibitors against cathepsin K. Br J Pharmacol 173(2): 396-410.
  5. Teitelbaum S L (2000)Bone resorption by osteoclasts. Science 289(5484): 1504-1508.
  6. Fata JE, Kong YY, Li J, Sasaki T, Irie-Sasaki J, et al. (2000) The osteoclast differentiation factor osteoprotegerin-ligand is essential for mammary gland development. Cell 103: 41-
  7. Marco Ponzetti, Nadia Rucci* (2019) Updates on Osteoimmunology: What’s New on the Cross-Talk Between Bone and Immune System. Frontiers in Endocrinology frontiersin.org 1(10): 236
  8. Lacey DL, Timms E, Tan HL, Kelley MJ, Dunstan CR, et al. (1998) Osteoprotegerin ligand is a cytokine that regulates osteoclast differentiation and activation. Cell 93: 165-1
  9. Simonet WS, Lacey DL, Dunstan CR, Kelley M, Chang MS, et al. (1997) Osteoprotegerin: a novl secreted protein involved in the regulation of bone density. Cell 89: 309-3
  10. Barrow A D, Raynal N, Andersen T L, Slatter D A, Bihan D, et al. (2011) OSCAR is a collagen receptor that costimulates osteoclastogenesis in DAP12-deficient humans and mice. J Clin Invest 121(9): 3505-3516.
  11. Merck E, de Saint-Vis B, Scuiller M, Claude Gaillard, Christophe Caux, et al. (2005) "Fc receptor gamma-chain activation via hOSCAR induces survival and maturation of dendritic cells and modulates Toll-like receptor responses". Blood 105(9): 3623-36
  12. Nemeth K, Schoppet M, Al-Fakhri N, Helas S, Jessberger R, et al. (2011) The role of osteoclast-associated receptor in osteoimmunology. J Immunol 186(1): 13-
  13. Liu W, Zhang X (2015) Receptor activator of nuclear factor-κB ligand (RANKL)/RANK/osteoprotegerin system in bone and other tissues (review). Mol Med Rep 11(5): 3212-3218.
  14. Charles J F, Aliprantis A O (2014) Osteoclasts: more than ‘bone eaters’. Trends Mol Med 20(8): 449-459.
  15. Dallas S L, Prideaux M, Bonewald L F (2013) The osteocyte: an endocrine cell and more. Endocr Rev 34(5): 658-690.
  16. Adamopoulos IE, Chao CC, Geissler R, Laface D, BlumenscheinW, et al. (2010) Interleukin-17A upregulates receptor activator of NF-kappaB on osteoclast precursors. Arthritis Res Ther 12: ar2936
  17. Maddur MS, Miossec P, Kaveri SV, Bayry J (2012) Th17 cells: biology, pathogenesis of autoimmune and inflammatory diseases, and therapeutic strategies. Am Jpathol 181: 8-
  18. Noonan K, Marchionni L, Anderson J, Pardoll D, Roodman GD, et al. (2010) A novel role of IL-17-producing lymphocytes in mediating lytic bone disease in multiple myeloma. Blood 116: 3554-35
  19. Mori G, D’Amelio P, Faccio R, Brunetti G (2013) The Interplay between the bone and the immune system. Clin Dev Immunol
  20. Steinman RM, Banchereau J (2007) Taking dendritic cells into medicine. Nature 449: 419-4
  21. Thomas R, MacDonald KP, Pettit AR, Cavanagh LL, Padmanabha J, et al. (1999) Dendritic cells and the pathogenesis of rheumatoid arthritis. J Leukoc Biol 66: 286-2
  22. Santiago-Schwarz F, Anand P, Liu S, Carsons SE (2001) Dendritic cells (DCs) in rheumatoid arthritis (RA): progenitor cells and soluble factors contained in RA synovial fluid yield a subset of myeloid DCs that preferentially activate Th1 inflammatory-type responses. J Immunol 167: 1758-17
  23. Hajishengallis G, Moutsopoulos NM, Hajishengallis E, Chavakis T (2016) Immune and regulatory functions of neutrophils in inflammatory bone loss. Semin Immunol 28: 146-1
  24. Moutsopoulos NM, Konkel J, Sarmadi M, Eskan MA, Wild T, et al. (2014) Defective neutrophil recruitment in leukocyte adhesion deficiency type I disease causes local IL-17-driven inflammatory bone loss. Sci Transl Med 6:
  25. Poubelle PE, Chakravarti A, Fernandes MJ, Doiron K, Marceau AA, et al. Differential expression of RANK, RANK-L, and osteoprotegerin by synovial fluid neutrophils from patients with rheumatoid arthritis and by healthy human blood neutrophils. Arthritis Res Ther 9:
  26. Nagasawa T (2006) Microenvironmental niches in the bone marrow required for Bcell development. Nat Rev Immunol 6: 107-1
  27. Onal M, Xiong J, Chen X, Thosten JD, Almeida JD, et al. (2012) Receptor activator of nuclear factor kB ligand (RANKL) protein expression by B lymphocytes contributes to ovariectomy-induced bone loss. J Biol Chem 287: 29851-298
  28. Cappariello A, Maurizi A, Veeriah V, Teti A (2014) The great beauty of osteoclast. Arch Biochem Biophys 561: 13-
  29. Lubberts E, KoendersMI, van der BergWB (2005) The role of T-cell interleukin-17 in conducting destructive arthritis: lessons fromanimal models. Arthritis Res Ther 7: 29-
  30. Schroder K, Hertzog PJ, Ravasi T, HumeDA (2004) Interferon-gamma: an overview of signals, mechanisms and functions. J Leukoc Biol 75: 163-1
  31. Tang M, Tian L, Luo G, Yu X Interferon-gamma-mediated osteoimmunology. Front Immunol 9:
  32. Ji JD, Park-Min KH, Shen Z, Fajardo RJ, Goldring SR, et al. (2009) Inhibition of RANK expression and osteoclastogenesis by TLRs and IFNgamma in human osteoclast precursors. J Immunol 183: 7223-72
  33. Takayanagi H, Sunhwa Kim, Koichi Matsuo, Hiroshi Suzuki, Tomohiko Suzuki, et al. (2002) RANKL maintains bone homeostasis through c-Fos-dependent induction of interferon-β. Nature 416: 744-749.
  34. Takayanagi H, Kim S, Koga T, Taniguchi T (2005) Stat1-mediated cytoplasmic attenuation in osteoimmunology. J Cell Biochem 94: 232-240.
  35. Hayman AR, Jones SJ, Boyde A, Foster D, Colledge WH, et al. (1996) Mice lacking tartrate-resistant acid phosphatase (Acp 5) have disrupted endochondral ossification and mild osteopetrosis. Development 122: 3151-
  36. Hikosaka Y, Nitta T, Ohigashi I, Yano K, Ishimaru N, et al. (2008) The cytokine RANKL produced by positively selected thymocytes fosters medullary thymic epithelial cells that express autoimmune regulator. Immunity 29: 438-
  37. Hino K, Ikeya M, Horigome K, Matsumoto Y, Ebise H, et al. (2015) Neofunction of ACVR1 in fibrodysplasia ossificans progressiva. Proc Natl Acad Sci USA 112: 15438-
  38. Boyle W J, Simonet WS, Lacey DL (2003) Osteoclast differentiation and activation. Nature 423: 337-342.
  39. Hiroshi Takayanagi (2007) Osteoimmunology: shared mechanisms and crosstalk between the immune and bone systems. 7(4): 292-304.
  40. Abe E, Marians RC, Yu W, Wu XB, Ando T, et al. (2003) TSH is a negative regulator of skeletal remodeling. Cell 115: 51-
  41. Abram CL, Roberge GL, Hu Y, Lowell CA (2014) Comparative analysis of the efficiency and specificity of myeloid-Cre deleting strains using ROSA-EYFP reporter mice. J Immunol Methods 408: 89-
  42. Adams GB, Chabner KT, Alley IR, Olson DP, Szczepiorkowski ZM, et al. (2006) Stem cell engraftment at the endosteal niche is specified by the calcium-sensing receptor. Nature 439: 599-
  43. Begg SK, Radley JM, Pollard JW, Chisholm OT, Stanley ER, et al. (1993) Delayed hematopoietic development in osteopetrotic (op/op) mice. J Exp Med 177: 237-
  44. Begovich AB, Carlton VE, Honigberg LA, Schrodi SJ, Chokkalingam AP, et al. (2004) A missense single-nucleotide polymorphism in a gene encoding a protein tyrosine phosphatase PTPN22 is associated with rheumatoid arthritis. Am J Hum Genet 75: 330-
  45. Behar O, Golden JA, Mashimo H, Schoen FJ, Fishman MC, et al. (1996) Semaphorin III is needed for normal patterning and growth of nerves, bones and heart. Nature 383: 525-
  46. Bennett CN, Longo KA, Wright WS, Suva LJ, Lane TF, et al. (2005) Regulation of osteoblastogenesis and bone mass by Wnt10b. Proc Natl Acad Sci USA 102: 3324-3329.
  47. Bennett CN, Ouyang H, Ma YL, Zeng Q, Gerin I, et al. (2007) Wnt10b increases postnatal bone formation by enhancing osteoblast differentiation. J Bone Miner Res 22: 1924-1932.
  48. Beral V, Bull D, Reeves G (2005) Million Women Study Collaborators. Endometrial cancer and hormone-replacement therapy in the Million Women Study. Lancet 365: 1543-
  49. Bernstein CN, Leslie WD, Leboff MS (2003) AGA technical review on osteoporosis in gastrointestinal diseases. Gastroenterology 124: 795-
  50. Elefteriou F (2008) Regulation of bone remodeling by the central and peripheral nervous system. Arch Biochem Biophys 473: 231-
  51. Ellmeier W, Jung S, Sunshine MJ, Hatam F, Xu Y, et al. (2000) Severe B cell deficiency in mice lacking the tec kinase family members Tec and Btk. J Exp Med 192: 1611-
  52. Fornier MN (2010) Denosumab: second chapter in controlling bone metastases or a new book. J Clin Oncol 28: 5127-5131.
  53. Franchimont N, Reenaers C, Lambert C, Belaiche J, Bours V, et al. (2004) Increased expression of receptor activator of NF-B ligand (RANKL), its receptor RANK and its decoy receptor osteoprotegerin in the colon of Crohn’s disease patients. Clin Exp Immunol 138: 491-498.
  54. Guerrini MM, Okamoto K, Komatsu N, Sawa S, Danks L, et al. (2015) Inhibition of the TNF family cytokine RANKL prevents autoimmune inflammation in the central nervous system. Immunity 43: 1174-
  55. Guerrini MM, Sobacchi C, Cassani B, Abinun M, Kilic SS, et al. (2008) Human osteoclast-poor osteopetrosis with hypogammaglobulinemia due to TNFRSF11A (RANK) mutations. Am J Hum Genet 83: 64-76.
  56. Guggenbuhl P, Grosbois B, Chalès G (2008) Gaucher disease. Joint Bone Spine 75: 116-
  57. Harre U, Georgess D, Bang H, Bozec A, Axmann R, et al. (2012) Induction of osteoclastogenesis and bone loss by human autoantibodies against citrullinated vimentin. J Clin Invest 122: 1791-
  58. Seoung-Hoon Lee, Tae-Soo Kim, Yongwon Choi, Joseph Lorenzo (2017) Osteoimmunology: cytokines and the skeletal system Physiol Rev 97(10).
  59. Jostins L, Ripke S, Weersma RK, Duerr RH, McGovern DP, et (2012) International IBD Genetics Consortium (IIBDGC). Host-microbe interactions have shaped the genetic architecture of inflammatory bowel disease. Nature 491: 119-124.
  60. Joyce-Shaikh B, Bigler ME, Chao CC, Murphy EE, Blumenschein WM, et al. (2010) Myeloid DAP12-associating lectin (MDL)-1 regulates synovial inflammation and bone erosion associated with autoimmune arthritis. J Exp Med 207: 579-
  61. Asagiri M, Hirai T, Kunigami T, Kamano S, Gober HJ, et al. (2008) Cathepsin K-dependent toll-like receptor 9 signaling revealed in experimental arthritis. Science 319: 624-
  62. Воропаева АА, Фаламеева ОВ, Садовой МА, Алексеева НА (2017) РОЛЬ КАТЕПСИНА К В РЕМОДЕЛИРОВАНИИ КОСТНОЙ ТКАНИ И МЕХАНИЗМЫ РЕГУЛЯЦИИ ЕГО АКТИВНОСТИ // Международный журнал прикладных и фундаментальных исследований. –№ 12-1. – С: 200-204.
  63. Capulli M, Rufo A, Teti A, Rucci N (2009) Global transcriptome analysis inmouse calvarial osteoblasts highlights sets of genes regulated by modelled microgravity and identifies a “mechanoresponsive osteoblast gene signature”. J Cell Biochem 15: 240-2
  64. Capulli M, Ponzetti M, Maurizi A, Gemini-Piperni S, Berger T, et al. (2018) A complex role for Lipocalin 2 in bone metabolism: global ablation in mice induces osteopenia caused by an altered energy metabolism. J Bone Miner Res 33: 1141-11
  65. Hristova (2019) Rheumatoid arthritis – the nature of the disease and assessment of its activity, GENERAL MEDICINE XXI 2: 73-76.
  66. Takayanagi H (2015) Osteoimmunology in 2014: Two-faced immunology-from osteogenesis to bone resorption. Nat Rev Rheumatol 11(2): 74-76.
  67. Sućur A, Katavić V, Kelava T, Jajić Z, Kovačić N, et al. (2014) Induction of osteoclast progenitors in inflammatory conditions: key to bone destruction in arthritis. Int Orthop 38(9): 1893-1903.
  68. Liu W, Zhang X (2015) Receptor activator of nuclear factor-κB ligand (RANKL)/RANK/osteoprotegerin system in bone and other tissues (review). Mol Med Rep 11(5): 3212-3218. 2.
  69. M Ma, Z Tan, W Li, H Zhang, Y Liu, et al. (2022) Osteoimmunology mechanism of osteonecrosis of the femoral head. Infographic Bone Joint Res 11(1): 29-
  70. Brown J P, Roux C, Ho P R, Bolognese M A, Hall J, et al. (2014) Denosumab significantly increases bone mineral density and reduces bone turnover compared with monthly oral ibandronate and risedronate in postmenopausal women who remained at higher risk for fracture despite previous suboptimal treatment with an oral bisphosphonate. Osteoporos Int 25(7): 1953-1961.
  71. Lories R J, Luyten F P (2009) Osteoimmunology: Wnt antagonists: for better or worse?. Nat Rev Rheumatol 5(8): 420-421.
  72. Deal C (2012) Bone loss in rheumatoid arthritis: systemic, periarticular and focal. Curr Rheumatol Rep 14(3): 231-237.
  73. Klontzas M E, Kenanidis E I, MacFarlane R J, Michail T, Potoupnis M E, et al. (2016) Investigational drugs for fracture healing: preclinical & clinical data. Expert Opin Investig Drugs 25(5): 585-596.
  74. DiSpirito J R, Mathis D (2015) Immunological contributions to adipose tissue homeostasis. Semin Immunol 27(5): 315-321.
  75. Madeira E, Mafort T T, Madeira M, Guedes E P, Moreira R O, et al. (2014) Lean mass as a predictor of bone density and microarchitecture in adult obese individuals with metabolic syndrome. Bone 59: 89-92.
  76. Sun K, Liu J, Lu N, Sun H, Ning G, et al. (2014) Association between metabolic syndrome and bone fractures: a metaanalysis of observational studies. BMC Endocr. Disord 14:
  77. Kishida K, Funahashi T, Shimomura I (2014) Adiponectin as a routine clinical biomarker. Best Pract Res Clin Endocrinol Metab 28(1): 119-130.
  78. Cildir G, Akıncılar S C, Tergaonkar V (2013) Chronic adipose tissue inflammation: all immune cells on the stage. Trends Mol Med 19(8): 487-500.
  79. Musso G, Paschetta E, Gambino R, Cassader M, Molinaro F, et al. (2013) Interactions among bone, liver, and adiposetissue predisposing to diabesity and fatty liver. Trends Mol Med 19(9): 522-535.
  80. M Ma, Z Tan, W Li, H Zhang, Y Liu, et al. (2022) Osteoimmunology mechanism of osteonecrosis of the femoral head. Infographic Bone Joint Res 11(1): 29-
  81. Calvi LM, G B Adams, K W Weibrecht, J M Weber, D P Olson, et al. (2003) Osteoblastic cells regulate the haematopoietic stem cell niche. Nature 425(6960): 841-846.
  82. Zhang J, Chao Niu, Ling Ye, Haiyang Huang, Xi He, et al. (2003) Identification of the haematopoietic stem cell niche and control of the niche size. Nature 425(6960): 836-
  83. Iwasaki H, Suda T (2009) Cancer stem cells and their niche. Cancer Sci 100(7): 1166-
  84. Dallas Jones, Laurie H Glimcher, Antonios O Aliprantis (2011) The Journal of Clinical Investigation, Osteoimmunology at the nexus of arthritis, osteoporosis, cancer, and infection 121(7): 2534-2542.
  85. Kawai T, Akira S (2010) The role of pattern-recognition receptors in innate immunity: update on Toll-like receptors. Nat Immunol 11(5): 373-
  86. Bar-Shavit Z (2008) Taking a toll on the bones: regulation of bone metabolism by innate immune regulators. Autoimmunity 41(3): 195-
  87. Delneste Y, Beauvillain C, Jeannin P (2007) "[Innate immunity: structure and function of TLRs]". Médecine/Sciences 23 (1): 67-
  88. Takeda K, Akira S (2005) "Toll-like receptors in innate immunity". International Immunology 17(1): 1-
  89. Medzhitov R, Preston-Hurlburt P, Janeway CA (1997) "A human homologue of the Drosophila Toll protein signals activation of adaptive immunity". Nature 388 (6640): 394- Bibcode: 1997Natur.388.394M.
  90. Sharma N, Akhade AS, Qadri A (2013) "Sphingosine-1-phosphate suppresses TLR-induced CXCL8 secretion from human T cells". Journal of Leukocyte Biology 93(4): 521-52
  91. Young AB, Cooley ID, Chauhan VS, Marriott I (2011) Causative agents of osteomyelitis induce death domain–containing TNF–related apoptosis–inducing ligand receptor expression on osteoblasts. Bone 48(4): 857-
  92. Alexander EH, Rivera FA, Marriott I, Anguita J, Bost KL, et al. (2003) Staphylococcus aureus – induced tumor necrosis factor - related apoptosis – inducing ligand expression mediates apoptosis and caspase-8 activation in infected osteoblasts. BMC Microbiol 3:
  93. Walker DG (1975) Bone resorption restored in osteopetrotic mice by transplantsof normal bone marrow and spleen cells. Science 190: 784-78
  94. Yasuda H, Shima N, Nakagawa N, Yamaguchi K, Kinosaki M, et al. (1998) Osteoclast differentiation factor is a ligand for osteoprotegerin/osteoclastogenesis-inhibitory factor and is identical to TRANCE/RANKL. Proc Natl Acad Sci USA 95: 3597-
  95. Danks L, Takayanagi H (2013) Immunology and bone. J Biochem 154: 29-
  96. Amarasekara DS, Yun H, Kim S, Lee N, Kim H, et al. Regulation of osteoclast differentiation by cytokine networks. Immune Netw. 18:
  97. Vidal C, Bermeo S, Li W, Huang D, Kremer R, et al. Interferon gamma inhibits adipogenesis in vitro and prevents marrow fat infiltration in oophorectomized mice. Stem Cells 30:
  98. Sato K, Suematsu A, Okamoto K, Yamaguchi A, Morishita Y, et al. (2006) Th17 functions as an osteoclastogenic helper T cell subset that links T cell activation and bone destruction. J Exp Med 203: 2673-26
  99. Pacifici R, Brown C, Puscheck E, Friedrich E, Slatopolsky E, et al. (1991) Effect of surgical menopause and estrogen replacement on cytokine release fromhuman blood mononuclear cells. Proc Natl Acad Sci USA 88: 5134-513
  100. Hughes DE, Dai A, Tiffee JC, Li HH, Mundy GR, et al. (1996) Estrogen promotes apoptosis of murine osteoclasts mediated by TGF-beta. Nat Med 2: 1132-113
  101. Piva R, Penolazzi L, Lambertini E, Giordano S, Gambari R, et al. (2005) Induction of apoptosis of human primary osteoclasts treated with a transcription factor decoy mimicking a promoter region of estrogen receptor alpha. Apoptosis 10: 1079-10
  102. Srivastava S, Toraldo G, Weitzmann MN, Cenci S, Ross FP, et al. (2001) Estrogen decreases osteoclast formation by down-regulating receptor activator of NF-kappa B ligand (RANKL) induced JNK activation. J Biol Chem 276: 8836-88
  103. Manolagas SC, O’Brien CA, Almeida M (2013) The role of estrogen and androgen receptors in bone health and disease. Nat Rev Endocrinol 9: 699-
  104. Eghbali-Fatourechi G, Khosla S, Sanyal A, Boyle WJ, Lacey DL, et al. (2003) Role of RANK ligand in mediating increased bone resorption in early postmenopausal women. J Clin Invest 111: 1221-12
  105. Breuil Y, Ticchioni M, Testa J, Roux CH, Ferrari P, et al. (2010) Immune changes in post-menopausal osteoporosis: the immunos study. Osteoporos Int 21: 805-8
  106. D’Amelio P, Grimaldi A, Di Bella S, Brianza SZM, Cristofaro MA, et al. (2008) Estrogen deficiency increases osteoclastogenesis up-regulating T cells activity: a key mechanism in osteoporosis. Bone 43: 92-
  107. Adeel S, Singh K, Vydareny KH, Kumari M, Shah E, et al. (2013) Bone loss in surgically ovariectomized premenopausal women is associated with T Lymphocyte activation and thymic hypertrophy. J Investig Med 61: 1178-11
  108. Richardson J, Hill AM, Johnston CJ, McGregor A, Norrish AR, et al. (2008) Fracture healing in HIV-positive populations. J Bone Joint Surg Br 90: 988-9
  109. El-Jawhari JJ, Jones E, Giannoudis PV (2016) The role of immune cells in bone haling; what we know, do not know and future perspectives. Injury 47: 2399-2
  110. Askalonov AA (1981) Changes in some indices of cellular immunity in patients with uncomplicated and complicated healing of bone fractures. J Hyg Epidemiol Microbiol Immunol 25: 307-3
  111. TimlinM, Toomey D, Condron C, Power C, Street J, et al. (2005) Fracture hematoma is a potent proinflammatory mediator of neutrophil function. J Trauma 58: 1223-122
  112. Nam D, Mau E, Wang Y, Wright D, Silkstone D, et al. (2012) T-lymphocytes enable osteoblast maturation via IL-17F during early phase of fracture repair. PLoS ONE 7:
  113. Krevvata M, Silva BC, Manavalan JS, Galan-Diez M, Kode A, et al. (2014) Inhibition of leukemia cell engraftment and disease progression in mice by osteoblasts. Blood 124: 2834-28
  114. Schofield R (1978) The relationship between the spleen colony-forming cell and the haemopoietic stem cell. Blood Cells 4: 7-
  115. Boulais PE, Frenette P S (2015) Making sense of hematopoietic stem cell niches. Blood 125: 2621–2629.
  116. Szade K, Gulati GS, Chan CKF, Kao KS, Miyanishi M, et al. (2018) Where hematopoietic stem cells live: The bone marrow niche. Antioxid. Redox Signal 29: 191-
  117. Cristine Donham, Betsabel Chicana, Alexander G Robling, Asmaa Mohamed, Sonny Elizaldi, et a (2021) Sclerostin Depletion Induces Inflammation in the Bone Marrow of Mice. Int J Mol Sci 22: 9111.
  118. Prism S Schneider, Emilie Sandman, Paul A (2018) Martineau, Osteoimmunology: Effects of Standard Orthopaedic Interventions on Inflammatory Response and Early Fracture Healing. J Am Acad Orthop Surg 0: 1-10.
  119. Bergenstock M, Min W, Simon AM, Sabatino C, O’Connor JP, et al. (2005) A comparison between the effects of acetaminophen and celecoxib on bone fracture healing in rats. J Orthop Trauma 19(10): 717-
  120. Chen MR, Dragoo JL (2013) The effect of nonsteroidal anti-inflammatory drugs on tissue healing. Knee Surg Sports Traumatol Arthrosc 21(3): 540-
  121. Takayanagi H, K Ogasawara, S Hida, T Chiba, S Murata, et al. (2000) T cell-mediated regulation of osteoclastogenesis by signalling cross-talk between RANKL and IFN-γ. Nature 408: 600-
  122. Bellido T, Ali AA, Gubrij I, Plotkin LI, Fu Q, et al. (2005) Chronic elevation of parathyroid hormone in mice reduces expression of sclerostin by osteocytes: a novel mechanism for hormonal control of osteoblastogenesis. Endocrinology 146: 4577-
  123. Janja Zupan, Matjaž Jeras, Janja Marc (2012) Osteoimmunology and the infl uence of pro-infl ammatory cytokines on osteoclasts 21(5).
  124. Hayden MS, Ghosh S (2008) Shared principles in NF- B signaling. Cell 132: 344-
  125. Hirose J, Masuda H, Tokuyama N, Omata Y, Matsumoto T, et al. (2014) Bone resorption is regulated by cell-autonomous negative feedback loop of Stat5-Dusp axis in the osteoclast. J Exp Med 211: 153-
  126. Kabashima K, Banks TA, Ansel KM, Lu TT, Ware CF, et al. (2005) Intrinsic lymphotoxin-receptor requirement for homeostasis of lymphoid tissue dendritic cells. Immunity 22: 439-
  127. Kazuo Okamoto, Tomoki Nakashima, Masahiro Shinohara, Takako Negishi-Koga, Noriko Komatsu, et al. (2017) OSTEOIMMUNOLOGY: THE CONCEPTUAL FRAMEWORK UNIFYING THE IMMUNE AND SKELETAL SYSTEMS. Physiol Rev 97: 1295-1349.
  128. Corona-Sanchez EG, Muñoz-Valle JF, Gonzalez-Lopez L, Sanchez-Hernandez JD, Vazquez-Del Mercado M, et al. (2012) -383 A/C tumor necrosis factor receptor 1 polymorphism and ankylosing spondylitis in Mexicans: a preliminary study. Rheumatol Int 32: 2565-
  129. Davidson SI, Liu Y, Danoy PA, Wu X, Thomas GP, et al. (2011) Association of STAT3 and TNFRSF1A with ankylosing spondylitis in Han Chinese. Ann Rheum Dis 70: 289-
  130. Karaderi T, Pointon JJ, Wordsworth TW, Harvey D, Appleton LH, et al. (2012) Evidence of genetic association between TNFRSF1A encoding the p55 tumour necrosis factor receptor, and ankylosing spondylitis in UK Caucasians. Clin ExpRheumatol 30: 110-