Endocrinol Metab.  2023 Oct;38(5):504-521. 10.3803/EnM.2023.501.

Nuclear Factor-Kappa B Regulation of Osteoclastogenesis and Osteoblastogenesis

Affiliations
  • 1Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
  • 2Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA

Abstract

Maintenance of skeletal integrity requires the coordinated activity of multinucleated bone-resorbing osteoclasts and bone-forming osteoblasts. Osteoclasts form resorption lacunae on bone surfaces in response to cytokines by fusion of precursor cells. Osteoblasts are derived from mesenchymal precursors and lay down new bone in resorption lacunae during bone remodeling. Nuclear factorkappa B (NF-κB) signaling regulates osteoclast and osteoblast formation and is activated in osteoclast precursors in response to the essential osteoclastogenic cytokine, receptor activator of NF-κB ligand (RANKL), which can also control osteoblast formation through RANK-RANKL reverse signaling in osteoblast precursors. RANKL and some pro-inflammatory cytokines, including tumor necrosis factor (TNF), activate NF-κB signaling to positively regulate osteoclast formation and functions. However, these cytokines also limit osteoclast and osteoblast formation through NF-κB signaling molecules, including TNF receptor-associated factors (TRAFs). TRAF6 mediates RANKL-induced osteoclast formation through canonical NF-κB signaling. In contrast, TRAF3 limits RANKL- and TNF-induced osteoclast formation, and it restricts transforming growth factor β (TGFβ)-induced inhibition of osteoblast formation in young and adult mice. During aging, neutrophils expressing TGFβ and C-C chemokine receptor type 5 (CCR5) increase in bone marrow of mice in response to increased NF-κB-induced CC motif chemokine ligand 5 (CCL5) expression by mesenchymal progenitor cells and injection of these neutrophils into young mice decreased bone mass. TGFβ causes degradation of TRAF3, resulting in decreased glycogen synthase kinase-3β/β-catenin-mediated osteoblast formation and age-related osteoporosis in mice. The CCR5 inhibitor, maraviroc, prevented accumulation of TGFβ+/CCR5+ neutrophils in bone marrow and increased bone mass by inhibiting bone resorption and increasing bone formation in aged mice. This paper updates current understanding of how NF-κB signaling is involved in the positive and negative regulation of cytokine-mediated osteoclast and osteoblast formation and activation with a focus on the role of TRAF3 signaling, which can be targeted therapeutically to enhance bone mass.

Keyword

Osteoclasts; Osteoblasts; Aging; Osteoporosis; TNF receptor-associated factor 6; TNF receptor-associated factor 3; Transforming growth factor-beta; Bone resorption; Neutrophils; Receptors, chemokine

Figure

  • Fig. 1. Canonical and non-canonical nuclear factor-kappa B (NF-κB) signaling induced by tumor necrosis factor (TNF) and receptor activator of NF-κB ligand (RANKL) in osteoclast precursors. RANKL and TNF induce canonical NF-κB signaling by recruiting TNF receptor-associated factor 6 (TRAF6) and TRAF2/5, respectively, to their receptors to activate a complex consisting of inhibitory NF-κB (IκB) kinase α (IKKα), IKKβ, and IKKγ (NF-κB essential modulator [NEMO]). This complex induces phosphorylation and degradation of IκB-α and the release of p65/p50 heterodimers, which translocate to the nucleus. p65/p50 induce expression of c-Fos and nuclear factor of activated T cells c1 (NFATc1), two other transcription factors necessary for osteoclast precursor differentiation, as well as the inhibitory κB protein, NF-κB p100. In unstimulated cells, p100 binds to RelB to prevent its translocation to the nucleus. RANKL induces the ubiquitination (Ub) and lysosomal degradation of TRAF3, releasing NF-κB-inducing kinase (NIK) to activate (phosphorylate) IKKα, which leads to proteasomal processing of p100 to p52. RelB:p52 heterodimers then go to the nucleus to induce target gene expression. TNF signaling does not degrade TRAF3, and thus NIK is degraded constitutively, leading to the accumulation of p100 in the cytoplasm of osteoclast precursors to limit their differentiation. TAK1, TGFβ-activated kinase-1; cIAP, complex inhibitor of apoptosis.

  • Fig. 2. Mouse models with genetically modified nuclear factor-kappa B (NF-κB) activation status in osteoblast lineage cells demonstrating that both canonical and non-canonical NF-κB signaling pathways are involved in osteoblast functions postnatally. Col2, collagen 2; IKK, inhibitory IκB kinase; OB, osteoblast; Bglap2, bone gamma-carboxyglutamate protein 2; Prx1, paired related homeobox 1; TRAF3, TNF receptor-associated factor 3; NIK, NF-κB-inducing kinase; MSC, mesenchymal stem cell; Osx, osterix.

  • Fig. 3. Tumor necrosis factor (TNF) receptor-associated factor 3 (TRAF3) functions in osteoclast and osteoblast precursors in bone resorption and bone formation. In aged mice, increased levels of receptor activator of nuclear factor-kappa B (NF-κB) ligand (RANKL) induce (1) TRAF3 ubiquitination and subsequent lysosomal degradation in osteoclast (OC) precursors to stimulate bone resorption through NF-κB signaling. As a result, (2) transforming growth factor β1 (TGFβ1) is released from bone matrix and activated in the acid environment in resorption lacunae. Activated TGFβ1 binds to its receptor complex on mesenchymal progenitor cells (MPCs) to which TRAF3 is recruited and subsequently ubiquitinated and degraded in lysosomes. As a result, (3) NF-κB signaling is activated and both RelA and RelB bind to the RANKL promoter to further promote RANKL production, enhancing bone resorption. In young and adult mice, TRAF3 inhibits glycogen synthase kinase-3β (GSK3β) activation in MPCs to prevent β-catenin degradation, allowing β-catenin accumulation and nuclear translocation to maintain osteoblast [OB] differentiation). In aged mice, TRAF3 degradation also leads to (4) increased NF-κB-induced secretion of CC motif chemokine ligand 5 (CCL5) by MPCs, which attracts increased numbers of TGFβ1/C-C chemokine receptor type 3 (CCR3)-expressing neutrophils (TCN) to the bone marrow where they release TGFβ, leading to further degradation of TRAF3 and (5) phosphorylation of GSK3β on T216. This phosphorylation mediates degradation of β-catenin, leading to (6) inhibition of OB precursor differentiation, increased production of osteoprotegerin (OPG) and decreased bone mass.


Reference

1. Yahara Y, Nguyen T, Ishikawa K, Kamei K, Alman BA. The origins and roles of osteoclasts in bone development, homeostasis and repair. Development. 2022; 149:dev199908.
Article
2. Jacome-Galarza CE, Percin GI, Muller JT, Mass E, Lazarov T, Eitler J, et al. Developmental origin, functional maintenance and genetic rescue of osteoclasts. Nature. 2019; 568:541–5.
Article
3. Yahara Y, Barrientos T, Tang YJ, Puviindran V, Nadesan P, Zhang H, et al. Erythromyeloid progenitors give rise to a population of osteoclasts that contribute to bone homeostasis and repair. Nat Cell Biol. 2020; 22:49–59.
Article
4. Tsukasaki M, Huynh NC, Okamoto K, Muro R, Terashima A, Kurikawa Y, et al. Stepwise cell fate decision pathways during osteoclastogenesis at single-cell resolution. Nat Metab. 2020; 2:1382–90.
Article
5. McDonald MM, Khoo WH, Ng PY, Xiao Y, Zamerli J, Thatcher P, et al. Osteoclasts recycle via osteomorphs during RANKL-stimulated bone resorption. Cell. 2021; 184:1330–47.
Article
6. Hughes DE, Boyce BF. Apoptosis in bone physiology and disease. Mol Pathol. 1997; 50:132–7.
Article
7. Boyce BF. Advances in the regulation of osteoclasts and osteoclast functions. J Dent Res. 2013; 92:860–7.
Article
8. Yu W, Zhong L, Yao L, Wei Y, Gui T, Li Z, et al. Bone marrow adipogenic lineage precursors promote osteoclastogenesis in bone remodeling and pathologic bone loss. J Clin Invest. 2021; 131:e140214.
Article
9. Sims NA, Martin TJ. Osteoclasts provide coupling signals to osteoblast lineage cells through multiple mechanisms. Annu Rev Physiol. 2020; 82:507–29.
Article
10. Zhao C, Irie N, Takada Y, Shimoda K, Miyamoto T, Nishiwaki T, et al. Bidirectional ephrinB2-EphB4 signaling controls bone homeostasis. Cell Metab. 2006; 4:111–21.
Article
11. Hayashi M, Nakashima T, Taniguchi M, Kodama T, Kumanogoh A, Takayanagi H. Osteoprotection by semaphorin 3A. Nature. 2012; 485:69–74.
Article
12. Ikebuchi Y, Aoki S, Honma M, Hayashi M, Sugamori Y, Khan M, et al. Coupling of bone resorption and formation by RANKL reverse signalling. Nature. 2018; 561:195–200.
Article
13. Karin M, Greten FR. NF-kappaB: linking inflammation and immunity to cancer development and progression. Nat Rev Immunol. 2005; 5:749–59.
Article
14. Courtois G, Gilmore TD. Mutations in the NF-kappaB signalingpathway: implications for human disease. Oncogene. 2006; 25:6831–43.
Article
15. Vallabhapurapu S, Karin M. Regulation and function of NF-kappaB transcription factors in the immune system. Annu Rev Immunol. 2009; 27:693–733.
Article
16. Boyce BF, Xing L. Biology of RANK, RANKL, and osteoprotegerin. Arthritis Res Ther. 2007; 9(Suppl 1):S1.
Article
17. Novack DV, Yin L, Hagen-Stapleton A, Schreiber RD, Goeddel DV, Ross FP, et al. The IkappaB function of NF-kappaB2 p100 controls stimulated osteoclastogenesis. J Exp Med. 2003; 198:771–81.
18. Hayden MS, Ghosh S. Shared principles in NF-kappaB signaling. Cell. 2008; 132:344–62.
19. Dobrzanski P, Ryseck RP, Bravo R. Specific inhibition of RelB/p52 transcriptional activity by the C-terminal domain of p100. Oncogene. 1995; 10:1003–7.
20. Liou HC, Nolan GP, Ghosh S, Fujita T, Baltimore D. The NF-kappa B p50 precursor, p105, contains an internal I kappa B-like inhibitor that preferentially inhibits p50. EMBO J. 1992; 11:3003–9.
Article
21. Palombella VJ, Rando OJ, Goldberg AL, Maniatis T. The ubiquitin-proteasome pathway is required for processing the NF-kappa B1 precursor protein and the activation of NF-kappa B. Cell. 1994; 78:773–85.
Article
22. Overgaard M, Borch J, Gerdes K. RelB and RelE of Escherichia coli form a tight complex that represses transcription via the ribbon-helix-helix motif in RelB. J Mol Biol. 2009; 394:183–96.
Article
23. Shibata W, Maeda S, Hikiba Y, Yanai A, Ohmae T, Sakamoto K, et al. Cutting edge: the IkappaB kinase (IKK) inhibitor, NEMO-binding domain peptide, blocks inflammatory injury in murine colitis. J Immunol. 2007; 179:2681–5.
24. Chaisson ML, Branstetter DG, Derry JM, Armstrong AP, Tometsko ME, Takeda K, et al. Osteoclast differentiation is impaired in the absence of inhibitor of kappa B kinase alpha. J Biol Chem. 2004; 279:54841–8.
25. Zarnegar BJ, Wang Y, Mahoney DJ, Dempsey PW, Cheung HH, He J, et al. Noncanonical NF-kappaB activation requires coordinated assembly of a regulatory complex of the adaptors cIAP1, cIAP2, TRAF2 and TRAF3 and the kinase NIK. Nat Immunol. 2008; 9:1371–8.
Article
26. Bonizzi G, Karin M. The two NF-kappaB activation pathways and their role in innate and adaptive immunity. Trends Immunol. 2004; 25:280–8.
27. Senftleben U, Cao Y, Xiao G, Greten FR, Krahn G, Bonizzi G, et al. Activation by IKKalpha of a second, evolutionary conserved, NF-kappa B signaling pathway. Science. 2001; 293:1495–9.
Article
28. Yilmaz ZB, Weih DS, Sivakumar V, Weih F. RelB is required for Peyer’s patch development: differential regulation of p52-RelB by lymphotoxin and TNF. EMBO J. 2003; 22:121–30.
Article
29. Fusco AJ, Savinova OV, Talwar R, Kearns JD, Hoffmann A, Ghosh G. Stabilization of RelB requires multidomain interactions with p100/p52. J Biol Chem. 2008; 283:12324–32.
Article
30. Chang J, Wang Z, Tang E, Fan Z, McCauley L, Franceschi R, et al. Inhibition of osteoblastic bone formation by nuclear factor-kappaB. Nat Med. 2009; 15:682–9.
Article
31. Yao Z, Li Y, Yin X, Dong Y, Xing L, Boyce BF. NF-κB RelB negatively regulates osteoblast differentiation and bone formation. J Bone Miner Res. 2014; 29:866–77.
Article
32. Xing L, Chen D, Boyce BF. Mice deficient in NF-κB p50 and p52 or RANK have defective growth plate formation and post-natal dwarfism. Bone Res. 2013; 1:336–45.
Article
33. Beg AA, Sha WC, Bronson RT, Ghosh S, Baltimore D. Embryonic lethality and liver degeneration in mice lacking the RelA component of NF-kappa B. Nature. 1995; 376:167–70.
Article
34. Franzoso G, Carlson L, Xing L, Poljak L, Shores EW, Brown KD, et al. Requirement for NF-kappaB in osteoclast and Bcell development. Genes Dev. 1997; 11:3482–96.
35. Iotsova V, Caamano J, Loy J, Yang Y, Lewin A, Bravo R. Osteopetrosis in mice lacking NF-kappaB1 and NF-kappaB2. Nat Med. 1997; 3:1285–9.
36. Lacey DL, Timms E, Tan HL, Kelley MJ, Dunstan CR, Burgess T, et al. Osteoprotegerin ligand is a cytokine that regulates osteoclast differentiation and activation. Cell. 1998; 93:165–76.
Article
37. Yasuda H, Shima N, Nakagawa N, Yamaguchi K, Kinosaki M, Mochizuki S, et al. Osteoclast differentiation factor is a ligand for osteoprotegerin/osteoclastogenesis-inhibitory factor and is identical to TRANCE/RANKL. Proc Natl Acad Sci U S A. 1998; 95:3597–602.
Article
38. Li J, Sarosi I, Yan XQ, Morony S, Capparelli C, Tan HL, et al. RANK is the intrinsic hematopoietic cell surface receptor that controls osteoclastogenesis and regulation of bone mass and calcium metabolism. Proc Natl Acad Sci U S A. 2000; 97:1566–71.
Article
39. Xing L, Bushnell TP, Carlson L, Tai Z, Tondravi M, Siebenlist U, et al. NF-kappaB p50 and p52 expression is not required for RANK-expressing osteoclast progenitor formation but is essential for RANK- and cytokine-mediated osteoclastogenesis. J Bone Miner Res. 2002; 17:1200–10.
40. Xing L, Carlson L, Story B, Tai Z, Keng P, Siebenlist U, et al. Expression of either NF-kappaB p50 or p52 in osteoclast precursors is required for IL-1-induced bone resorption. J Bone Miner Res. 2003; 18:260–9.
41. Fuller K, Owens JM, Jagger CJ, Wilson A, Moss R, Chambers TJ. Macrophage colony-stimulating factor stimulates survival and chemotactic behavior in isolated osteoclasts. J Exp Med. 1993; 178:1733–44.
Article
42. Tanaka S, Takahashi N, Udagawa N, Tamura T, Akatsu T, Stanley ER, et al. Macrophage colony-stimulating factor is indispensable for both proliferation and differentiation of osteoclast progenitors. J Clin Invest. 1993; 91:257–63.
Article
43. Vaira S, Alhawagri M, Anwisye I, Kitaura H, Faccio R, Novack DV. RelA/p65 promotes osteoclast differentiation by blocking a RANKL-induced apoptotic JNK pathway in mice. J Clin Invest. 2008; 118:2088–97.
Article
44. Vaira S, Johnson T, Hirbe AC, Alhawagri M, Anwisye I, Sammut B, et al. RelB is the NF-kappaB subunit downstream of NIK responsible for osteoclast differentiation. Proc Natl Acad Sci U S A. 2008; 105:3897–902.
45. Novack DV. Unique personalities within the NF-κB family: distinct functions for p65 and RelB in the osteoclast. Adv Exp Med Biol. 2011; 691:163–7.
Article
46. Asagiri M, Sato K, Usami T, Ochi S, Nishina H, Yoshida H, et al. Autoamplification of NFATc1 expression determines its essential role in bone homeostasis. J Exp Med. 2005; 202:1261–9.
Article
47. Takayanagi H, Kim S, Koga T, Nishina H, Isshiki M, Yoshida H, et al. Induction and activation of the transcription factor NFATc1 (NFAT2) integrate RANKL signaling in terminal differentiation of osteoclasts. Dev Cell. 2002; 3:889–901.
Article
48. Zhao B, Ivashkiv LB. Negative regulation of osteoclastogenesis and bone resorption by cytokines and transcriptional repressors. Arthritis Res Ther. 2011; 13:234.
Article
49. Yamashita T, Yao Z, Li F, Zhang Q, Badell IR, Schwarz EM, et al. NF-kappaB p50 and p52 regulate receptor activator of NF-kappaB ligand (RANKL) and tumor necrosis factor-induced osteoclast precursor differentiation by activating c-Fos and NFATc1. J Biol Chem. 2007; 282:18245–53.
50. Miyauchi Y, Ninomiya K, Miyamoto H, Sakamoto A, Iwasaki R, Hoshi H, et al. The Blimp1-Bcl6 axis is critical to regulate osteoclast differentiation and bone homeostasis. J Exp Med. 2010; 207:751–62.
Article
51. Darnay BG, Besse A, Poblenz AT, Lamothe B, Jacoby JJ. TRAFs in RANK signaling. Adv Exp Med Biol. 2007; 597:152–9.
Article
52. Otero JE, Dai S, Foglia D, Alhawagri M, Vacher J, Pasparakis M, et al. Defective osteoclastogenesis by IKKbeta-null precursors is a result of receptor activator of NF-kappaB ligand (RANKL)-induced JNK-dependent apoptosis and impaired differentiation. J Biol Chem. 2008; 283:24546–53.
53. Ruocco MG, Maeda S, Park JM, Lawrence T, Hsu LC, Cao Y, et al. IκB kinase (IKK)β, but not IKKα, is a critical mediator of osteoclast survival and is required for inflammation-induced bone loss. J Exp Med. 2005; 201:1677–87.
Article
54. Otero JE, Dai S, Alhawagri MA, Darwech I, Abu-Amer Y. IKKbeta activation is sufficient for RANK-independent osteoclast differentiation and osteolysis. J Bone Miner Res. 2010; 25:1282–94.
Article
55. Jimi E, Aoki K, Saito H, D’Acquisto F, May MJ, Nakamura I, et al. Selective inhibition of NF-kappa B blocks osteoclastogenesis and prevents inflammatory bone destruction in vivo. Nat Med. 2004; 10:617–24.
Article
56. Aya K, Alhawagri M, Hagen-Stapleton A, Kitaura H, Kanagawa O, Novack DV. NF-(kappa)B-inducing kinase controls lymphocyte and osteoclast activities in inflammatory arthritis. J Clin Invest. 2005; 115:1848–54.
57. Soysa NS, Alles N, Weih D, Lovas A, Mian AH, Shimokawa H, et al. The pivotal role of the alternative NF-kappaB pathway in maintenance of basal bone homeostasis and osteoclastogenesis. J Bone Miner Res. 2010; 25:809–18.
58. Zhao C, Xiu Y, Ashton J, Xing L, Morita Y, Jordan CT, et al. Noncanonical NF-κB signaling regulates hematopoietic stem cell self-renewal and microenvironment interactions. Stem Cells. 2012; 30:709–18.
Article
59. Weih F, Carrasco D, Durham SK, Barton DS, Rizzo CA, Ryseck RP, et al. Multiorgan inflammation and hematopoietic abnormalities in mice with a targeted disruption of RelB, a member of the NF-kappa B/Rel family. Cell. 1995; 80:331–40.
Article
60. Burkly L, Hession C, Ogata L, Reilly C, Marconi LA, Olson D, et al. Expression of relB is required for the development of thymic medulla and dendritic cells. Nature. 1995; 373:531–6.
Article
61. Xiu Y, Xu H, Zhao C, Li J, Morita Y, Yao Z, et al. Chloroquine reduces osteoclastogenesis in murine osteoporosis by preventing TRAF3 degradation. J Clin Invest. 2014; 124:297–310.
Article
62. Yao Z, Xing L, Boyce BF. NF-kappaB p100 limits TNF-induced bone resorption in mice by a TRAF3-dependent mechanism. J Clin Invest. 2009; 119:3024–34.
63. Xiao Y, Jin J, Chang M, Chang JH, Hu H, Zhou X, et al. Peli1 promotes microglia-mediated CNS inflammation by regulating Traf3 degradation. Nat Med. 2013; 19:595–602.
Article
64. Nakhaei P, Mesplede T, Solis M, Sun Q, Zhao T, Yang L, et al. The E3 ubiquitin ligase Triad3A negatively regulates the RIG-I/MAVS signaling pathway by targeting TRAF3 for degradation. PLoS Pathog. 2009; 5:e1000650.
Article
65. Hu H, Brittain GC, Chang JH, Puebla-Osorio N, Jin J, Zal A, et al. OTUD7B controls non-canonical NF-κB activation through deubiquitination of TRAF3. Nature. 2013; 494:371–4.
Article
66. Gonzalez-Noriega A, Grubb JH, Talkad V, Sly WS. Chloroquine inhibits lysosomal enzyme pinocytosis and enhances lysosomal enzyme secretion by impairing receptor recycling. J Cell Biol. 1980; 85:839–52.
Article
67. Ben-Zvi I, Kivity S, Langevitz P, Shoenfeld Y. Hydroxychloroquine: from malaria to autoimmunity. Clin Rev Allergy Immunol. 2012; 42:145–53.
Article
68. Miller AV, Ranatunga SK. Immunotherapies in rheumatologic disorders. Med Clin North Am. 2012; 96:475–96.
Article
69. He JQ, Zarnegar B, Oganesyan G, Saha SK, Yamazaki S, Doyle SE, et al. Rescue of TRAF3-null mice by p100 NFkappa B deficiency. J Exp Med. 2006; 203:2413–8.
70. Sun S, Tao J, Sedghizadeh PP, Cherian P, Junka AF, Sodagar E, et al. Bisphosphonates for delivering drugs to bone. Br J Pharmacol. 2021; 178:2008–25.
Article
71. Yang G, Zhu L, Hou N, Lan Y, Wu XM, Zhou B, et al. Osteogenic fate of hypertrophic chondrocytes. Cell Res. 2014; 24:1266–9.
Article
72. Yang L, Tsang KY, Tang HC, Chan D, Cheah KS. Hypertrophic chondrocytes can become osteoblasts and osteocytes in endochondral bone formation. Proc Natl Acad Sci U S A. 2014; 111:12097–102.
Article
73. Maes C, Kobayashi T, Selig MK, Torrekens S, Roth SI, Mackem S, et al. Osteoblast precursors, but not mature osteoblasts, move into developing and fractured bones along with invading blood vessels. Dev Cell. 2010; 19:329–44.
Article
74. Mizoguchi T, Pinho S, Ahmed J, Kunisaki Y, Hanoun M, Mendelson A, et al. Osterix marks distinct waves of primitive and definitive stromal progenitors during bone marrow development. Dev Cell. 2014; 29:340–9.
Article
75. Zhou BO, Yue R, Murphy MM, Peyer JG, Morrison SJ. Leptin-receptor-expressing mesenchymal stromal cells represent the main source of bone formed by adult bone marrow. Cell Stem Cell. 2014; 15:154–68.
Article
76. Pineault KM, Song JY, Kozloff KM, Lucas D, Wellik DM. Hox11 expressing regional skeletal stem cells are progenitors for osteoblasts, chondrocytes and adipocytes throughout life. Nat Commun. 2019; 10:3168.
Article
77. Israel A. The IKK complex, a central regulator of NF-kappaB activation. Cold Spring Harb Perspect Biol. 2010; 2:a000158.
78. Gilbert L, He X, Farmer P, Rubin J, Drissi H, van Wijnen AJ, et al. Expression of the osteoblast differentiation factor RUNX2 (Cbfa1/AML3/Pebp2alpha A) is inhibited by tumor necrosis factor-alpha. J Biol Chem. 2002; 277:2695–701.
79. Gilbert LC, Rubin J, Nanes MS. The p55 TNF receptor mediates TNF inhibition of osteoblast differentiation independently of apoptosis. Am J Physiol Endocrinol Metab. 2005; 288:E1011–8.
Article
80. Li Y, Li A, Strait K, Zhang H, Nanes MS, Weitzmann MN. Endogenous TNFalpha lowers maximum peak bone mass and inhibits osteoblastic Smad activation through NF-kappaB. J Bone Miner Res. 2007; 22:646–55.
81. Yamazaki M, Fukushima H, Shin M, Katagiri T, Doi T, Takahashi T, et al. Tumor necrosis factor alpha represses bone morphogenetic protein (BMP) signaling by interfering with the DNA binding of Smads through the activation of NF-kappaB. J Biol Chem. 2009; 284:35987–95.
82. Alles N, Soysa NS, Hayashi J, Khan M, Shimoda A, Shimokawa H, et al. Suppression of NF-kappaB increases bone formation and ameliorates osteopenia in ovariectomized mice. Endocrinology. 2010; 151:4626–34.
83. Cho HH, Shin KK, Kim YJ, Song JS, Kim JM, Bae YC, et al. NF-kappaB activation stimulates osteogenic differentiation of mesenchymal stem cells derived from human adipose tissue by increasing TAZ expression. J Cell Physiol. 2010; 223:168–77.
84. Moro GE, Minoli I, Fulconis F, Clementi M, Raiha NC. Growth and metabolic responses in low-birth-weight infants fed human milk fortified with human milk protein or with a bovine milk protein preparation. J Pediatr Gastroenterol Nutr. 1991; 13:150–4.
Article
85. Swarnkar G, Zhang K, Mbalaviele G, Long F, Abu-Amer Y. Constitutive activation of IKK2/NF-κB impairs osteogenesis and skeletal development. PLoS One. 2014; 9:e91421.
Article
86. Tata PR, Mou H, Pardo-Saganta A, Zhao R, Prabhu M, Law BM, et al. Dedifferentiation of committed epithelial cells into stem cells in vivo. Nature. 2013; 503:218–23.
Article
87. Park D, Spencer JA, Koh BI, Kobayashi T, Fujisaki J, Clemens TL, et al. Endogenous bone marrow MSCs are dynamic, fate-restricted participants in bone maintenance and regeneration. Cell Stem Cell. 2012; 10:259–72.
Article
88. Geurtzen K, Knopf F, Wehner D, Huitema LF, Schulte-Merker S, Weidinger G. Mature osteoblasts dedifferentiate in response to traumatic bone injury in the zebrafish fin and skull. Development. 2014; 141:2225–34.
Article
89. Blum N, Begemann G. Retinoic acid signaling spatially restricts osteoblasts and controls ray-interray organization during zebrafish fin regeneration. Development. 2015; 142:2888–93.
Article
90. Mishra R, Sehring I, Cederlund M, Mulaw M, Weidinger G. NF-κB signaling negatively regulates osteoblast dedifferentiation during zebrafish bone regeneration. Dev Cell. 2020; 52:167–82.
Article
91. Sun SC. The non-canonical NF-κB pathway in immunity and inflammation. Nat Rev Immunol. 2017; 17:545–58.
Article
92. Seo Y, Fukushima H, Maruyama T, Kuroishi KN, Osawa K, Nagano K, et al. Accumulation of p100, a precursor of NF-κB2, enhances osteoblastic differentiation in vitro and bone formation in vivo in aly/aly mice. Mol Endocrinol. 2012; 26:414–22.
93. Davis JL, Cox L, Shao C, Lyu C, Liu S, Aurora R, et al. Conditional activation of NF-κB inducing kinase (NIK) in the osteolineage enhances both basal and loading-induced bone formation. J Bone Miner Res. 2019; 34:2087–100.
Article
94. Davis JL, Thaler R, Cox L, Ricci B, Zannit HM, Wan F, et al. Constitutive activation of NF-κB inducing kinase (NIK) in the mesenchymal lineage using Osterix (Sp7)- or Fibroblast-specific protein 1 (S100a4)-Cre drives spontaneous soft tissue sarcoma. PLoS One. 2021; 16:e0254426.
Article
95. Davis JL, Pokhrel NK, Cox L, Rohatgi N, Faccio R, Veis DJ. Conditional loss of IKKα in Osterix+cells has no effect on bone but leads to age-related loss of peripheral fat. Sci Rep. 2022; 12:4915.
Article
96. Khosla S, Oursler MJ, Monroe DG. Estrogen and the skeleton. Trends Endocrinol Metab. 2012; 23:576–81.
Article
97. Biswas DK, Singh S, Shi Q, Pardee AB, Iglehart JD. Crossroads of estrogen receptor and NF-kappaB signaling. Sci STKE. 2005; 2005:pe27.
98. Yin L, Wu L, Wesche H, Arthur CD, White JM, Goeddel DV, et al. Defective lymphotoxin-beta receptor-induced NF-kappaB transcriptional activity in NIK-deficient mice. Science. 2001; 291:2162–5.
Article
99. Zarei A, Yang C, Gibbs J, Davis JL, Ballard A, Zeng R, et al. Manipulation of the alternative NF-κB pathway in mice has sexually dimorphic effects on bone. JBMR Plus. 2018; 3:14–22.
Article
100. Sun SC. Non-canonical NF-κB signaling pathway. Cell Res. 2011; 21:71–85.
Article
101. Jimi E. The role of BMP signaling and NF-κB signaling on osteoblastic differentiation, cancer development, and vascular diseases: is the activation of NF-κB a friend or foe of BMP function? Vitam Horm. 2015; 99:145–70.
102. Li X, Bai XZ. NF-kappaB modulates activation of the BMP-2 gene by trichostatin A. Mol Biol (Mosk). 2008; 42:990–6.
103. Tarapore RS, Lim J, Tian C, Pacios S, Xiao W, Reid D, et al. NF-κB has a direct role in inhibiting Bmp- and Wnt-induced matrix protein expression. J Bone Miner Res. 2016; 31:52–64.
Article
104. Urata M, Kokabu S, Matsubara T, Sugiyama G, Nakatomi C, Takeuchi H, et al. A peptide that blocks the interaction of NF-κB p65 subunit with Smad4 enhances BMP2-induced osteogenesis. J Cell Physiol. 2018; 233:7356–66.
Article
105. Baron R, Rawadi G. Targeting the Wnt/beta-catenin pathway to regulate bone formation in the adult skeleton. Endocrinology. 2007; 148:2635–43.
Article
106. Houschyar KS, Tapking C, Borrelli MR, Popp D, Duscher D, Maan ZN, et al. Wnt pathway in bone repair and regeneration: what do we know so far. Front Cell Dev Biol. 2019; 6:170.
107. Eferl R, Hoebertz A, Schilling AF, Rath M, Karreth F, Kenner L, et al. The Fos-related antigen Fra-1 is an activator of bone matrix formation. EMBO J. 2004; 23:2789–99.
Article
108. Lin X, Patil S, Gao YG, Qian A. The bone extracellular matrix in bone formation and regeneration. Front Pharmacol. 2020; 11:757.
Article
109. Li J, Ayoub A, Xiu Y, Yin X, Sanders JO, Mesfin A, et al. TGFβ-induced degradation of TRAF3 in mesenchymal progenitor cells causes age-related osteoporosis. Nat Commun. 2019; 10:2795.
Article
110. Ginaldi L, Di Benedetto MC, De Martinis M. Osteoporosis, inflammation and ageing. Immun Ageing. 2005; 2:14.
Article
111. Bliuc D, Nguyen ND, Milch VE, Nguyen TV, Eisman JA, Center JR. Mortality risk associated with low-trauma osteoporotic fracture and subsequent fracture in men and women. JAMA. 2009; 301:513–21.
Article
112. Pietschmann P, Mechtcheriakova D, Meshcheryakova A, Foger-Samwald U, Ellinger I. Immunology of osteoporosis: a mini-review. Gerontology. 2016; 62:128–37.
Article
113. Tang Y, Wu X, Lei W, Pang L, Wan C, Shi Z, et al. TGF-beta1-induced migration of bone mesenchymal stem cells couples bone resorption with formation. Nat Med. 2009; 15:757–65.
Article
114. ten Berge D, Brouwer A, Korving J, Martin JF, Meijlink F. Prx1 and Prx2 in skeletogenesis: roles in the craniofacial region, inner ear and limbs. Development. 1998; 125:3831–42.
115. Martin JF, Bradley A, Olson EN. The paired-like homeo box gene MHox is required for early events of skeletogenesis in multiple lineages. Genes Dev. 1995; 9:1237–49.
Article
116. Ng AH, Omelon S, Variola F, Allo B, Willett TL, Alman BA, et al. Adynamic bone decreases bone toughness during aging by affecting mineral and matrix. J Bone Miner Res. 2016; 31:369–79.
Article
117. Demontiero O, Vidal C, Duque G. Aging and bone loss: new insights for the clinician. Ther Adv Musculoskelet Dis. 2012; 4:61–76.
Article
118. Crane JL, Cao X. Bone marrow mesenchymal stem cells and TGF-β signaling in bone remodeling. J Clin Invest. 2014; 124:466–72.
Article
119. Lian N, Lin T, Liu W, Wang W, Li L, Sun S, et al. Transforming growth factor β suppresses osteoblast differentiation via the vimentin activating transcription factor 4 (ATF4) axis. J Biol Chem. 2012; 287:35975–84.
Article
120. Li J, Yao Z, Liu X, Duan R, Yi X, Ayoub A, et al. TGFβ1+ CCR5+ neutrophil subset increases in bone marrow and causes age-related osteoporosis in male mice. Nat Commun. 2023; 14:159.
121. Day TF, Guo X, Garrett-Beal L, Yang Y. Wnt/beta-catenin signaling in mesenchymal progenitors controls osteoblast and chondrocyte differentiation during vertebrate skeletogenesis. Dev Cell. 2005; 8:739–50.
122. Hill TP, Spater D, Taketo MM, Birchmeier W, Hartmann C. Canonical Wnt/beta-catenin signaling prevents osteoblasts from differentiating into chondrocytes. Dev Cell. 2005; 8:727–38.
123. Glass DA 2nd, Bialek P, Ahn JD, Starbuck M, Patel MS, Clevers H, et al. Canonical Wnt signaling in differentiated osteoblasts controls osteoclast differentiation. Dev Cell. 2005; 8:751–64.
Article
124. Udagawa N, Takahashi N, Jimi E, Matsuzaki K, Tsurukai T, Itoh K, et al. Osteoblasts/stromal cells stimulate osteoclast activation through expression of osteoclast differentiation factor/RANKL but not macrophage colony-stimulating factor: receptor activator of NF-kappa B ligand. Bone. 1999; 25:517–23.
Article
125. Bishop KA, Coy HM, Nerenz RD, Meyer MB, Pike JW. Mouse Rankl expression is regulated in T cells by c-Fos through a cluster of distal regulatory enhancers designated the T cell control region. J Biol Chem. 2011; 286:20880–91.
Article
126. Pang WW, Price EA, Sahoo D, Beerman I, Maloney WJ, Rossi DJ, et al. Human bone marrow hematopoietic stem cells are increased in frequency and myeloid-biased with age. Proc Natl Acad Sci U S A. 2011; 108:20012–7.
Article
127. Nam JS, Terabe M, Mamura M, Kang MJ, Chae H, Stuelten C, et al. An anti-transforming growth factor beta antibody suppresses metastasis via cooperative effects on multiple cell compartments. Cancer Res. 2008; 68:3835–43.
128. Chen F, Yang W, Huang X, Cao AT, Bilotta AJ, Xiao Y, et al. Neutrophils promote amphiregulin production in intestinal epithelial cells through TGF-β and contribute to intestinal homeostasis. J Immunol. 2018; 201:2492–501.
Article
129. Chu HW, Trudeau JB, Balzar S, Wenzel SE. Peripheral blood and airway tissue expression of transforming growth factor beta by neutrophils in asthmatic subjects and normal control subjects. J Allergy Clin Immunol. 2000; 106:1115–23.
130. Martin-Blondel G, Brassat D, Bauer J, Lassmann H, Liblau RS. CCR5 blockade for neuroinflammatory diseases: beyond control of HIV. Nat Rev Neurol. 2016; 12:95–105.
Article
131. Sun SC. The noncanonical NF-κB pathway. Immunol Rev. 2012; 246:125–40.
Article
132. Baud V, Liu ZG, Bennett B, Suzuki N, Xia Y, Karin M. Signaling by proinflammatory cytokines: oligomerization of TRAF2 and TRAF6 is sufficient for JNK and IKK activation and target gene induction via an amino-terminal effector domain. Genes Dev. 1999; 13:1297–308.
Article
133. Shi JH, Sun SC. Tumor necrosis factor receptor-associated factor regulation of nuclear factor κB and mitogen-activated protein kinase pathways. Front Immunol. 2018; 9:1849.
Article
134. Bishop GA. TRAF3 as a powerful and multitalented regulator of lymphocyte functions. J Leukoc Biol. 2016; 100:919–26.
Article
135. Nagel I, Bug S, Tonnies H, Ammerpohl O, Richter J, Vater I, et al. Biallelic inactivation of TRAF3 in a subset of B-cell lymphomas with interstitial del(14)(q24.1q32.33). Leukemia. 2009; 23:2153–5.
Article
136. Keats JJ, Fonseca R, Chesi M, Schop R, Baker A, Chng WJ, et al. Promiscuous mutations activate the noncanonical NF-kappaB pathway in multiple myeloma. Cancer Cell. 2007; 12:131–44.
Article
137. Lalani AI, Luo C, Han Y, Xie P. TRAF3: a novel tumor suppressor gene in macrophages. Macrophage (Houst). 2015; 2:e1009.
138. Burkiewicz JS, Scarpace SL, Bruce SP. Denosumab in osteoporosis and oncology. Ann Pharmacother. 2009; 43:1445–55.
Article
Full Text Links
  • ENM
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr