Bone development is tightly regulated by bone forming cells, osteoblasts, and bone resorbing cells, osteoclasts. Therefore, understanding the mechanisms governing osteoclastogenesis is crucial for addressing bone loss pathologies. Differentiation of osteoclasts is governed by RANK ligand which activates several signal transduction pathways, including MAP kinases and NF-?B pathways. Proximal activation entails recruitment of TRAF6 and other key proteins including TGF-?-activated kinase-1 (TAK1) to the receptor RANK. TRAF6, TAK1 and other signaling partners undergo extensive post-translational modifications aimed at stabilizing RANK signaling and enabling precise regulation and execution of proper down stream signals, primarily NF-?B activation. Precise regulation of NF-?B activity is crucial to maintain normal osteoclast activity and bone homeostasis. Conversely, abnormal activity of this transcription factor causes deleterious inflammatory osteolysis. In fact, we discovered recently that constitutive activation of IKK2 is sufficient to induce RANKL-independent osteoclastogenesis in vitro. More convincingly, we reported that knock-in of constitutively active IKK2 causes severe bone loss in mice. Given that IKK2 phosphorylation and activation is governed by TAK1, a MAP kinase heavily implicated in poly-ubiquitination and stabilization of RANK-TRAF6 complexes and down-stream signaling, we decided to investigate its molecular role in osteoclastogenesis. Thus, we generated mice harboring myeloid-specific deletion of TAK1. These mice displayed all hallmarks of osteopetrosis primarily defective osteoclastogenesis. Mechanistically, we observed that Tak1-null precursors fail to generate osteoclasts. More importantly, we discovered diminished expression of key osteoclastogenic proteins including TRAF6, NEMO and Notch-NICD. This phenomenon was associated with accumulation of NUMBL, a previously described neuron protein. Consistent with these observations, we established that exogenous expression of NUMBL induces degradation of TRAF6, NEMO, NOTCH1-NICD, and inhibits osteoclastogenesis in vitro. Inhibition of NUMBL using a dominant negative PTB-phosphotyrosine-binding of NUMBL and shRNAs knockdown of NUMBL enhanced expression of TRAF6 and NEMO and did not inhibit osteoclastogenesis in wild-type cells. In addition, inhibition of NUMBL using a dominant negative PTB of NUMBL and exogenous expression of NOTCH1-NICD restored osteoclastogenesis in TAK1-null cells. Based on these observations we hypothesize that """"""""NUMBL is a repressor of osteoclastogenesis and its expression is regulated by TAK1. Deletion of TAK1 leads to accumulation of NUMBL protein which induces degradation of TRAF6, NEMO and NICD proteins, and subsequently blocks osteoclastogenesis."""""""" To test this hypothesis, we propose to investigate the following specific aims: 1) Determine the mechanism by which TAK1 regulates NUMBL expression. 2) Determine the mechanism by which TAK1 deletion regulates and impedes expression of TRAF6, NEMO and NICD. 3) Determine the effect of genetic ablation of NUMBL on the osteopetrotic phenotype of TAK1-null mice.

Agency
National Institute of Health (NIH)
Institute
National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS)
Type
Research Project (R01)
Project #
5R01AR054326-07
Application #
8635282
Study Section
Skeletal Biology Structure and Regeneration Study Section (SBSR)
Program Officer
Chen, Faye H
Project Start
2006-12-01
Project End
2018-03-31
Budget Start
2014-04-01
Budget End
2015-03-31
Support Year
7
Fiscal Year
2014
Total Cost
$323,000
Indirect Cost
$110,500
Name
Washington University
Department
Orthopedics
Type
Schools of Medicine
DUNS #
068552207
City
Saint Louis
State
MO
Country
United States
Zip Code
63130
Rohatgi, Nidhi; Zou, Wei; Collins, Patrick L et al. (2018) ASXL1 impairs osteoclast formation by epigenetic regulation of NFATc1. Blood Adv 2:2467-2477
Wang, Chun; Hockerman, Susan; Jacobsen, E Jon et al. (2018) Selective inhibition of the p38? MAPK-MK2 axis inhibits inflammatory cues including inflammasome priming signals. J Exp Med 215:1315-1325
Wang, Cuicui; Abu-Amer, Yousef; O'Keefe, Regis J et al. (2018) Loss of Dnmt3b in Chondrocytes Leads to Delayed Endochondral Ossification and Fracture Repair. J Bone Miner Res 33:283-297
Swarnkar, Gaurav; Chen, Tim Hung-Po; Arra, Manoj et al. (2017) NUMBL Interacts with TAK1, TRAF6 and NEMO to Negatively Regulate NF-?B Signaling During Osteoclastogenesis. Sci Rep 7:12600
Wang, Chun; Xu, Can-Xin; Alippe, Yael et al. (2017) Chronic inflammation triggered by the NLRP3 inflammasome in myeloid cells promotes growth plate dysplasia by mesenchymal cells. Sci Rep 7:4880
Alippe, Yael; Wang, Chun; Ricci, Biancamaria et al. (2017) Bone matrix components activate the NLRP3 inflammasome and promote osteoclast differentiation. Sci Rep 7:6630
Zou, Wei; Rohatgi, Nidhi; Chen, Timothy Hung-Po et al. (2016) PPAR-? regulates pharmacological but not physiological or pathological osteoclast formation. Nat Med 22:1203-1205
Wang, C; Qu, C; Alippe, Y et al. (2016) Poly-ADP-ribosylation-mediated degradation of ARTD1 by the NLRP3 inflammasome is a prerequisite for osteoclast maturation. Cell Death Dis 7:e2153
Swarnkar, Gaurav; Shim, Kyuhwan; Nasir, Amjad M et al. (2016) Myeloid Deletion of Nemo Causes Osteopetrosis in Mice Owing to Upregulation of Transcriptional Repressors. Sci Rep 6:29896
Swarnkar, Gaurav; Karuppaiah, Kannan; Mbalaviele, Gabriel et al. (2015) Osteopetrosis in TAK1-deficient mice owing to defective NF-?B and NOTCH signaling. Proc Natl Acad Sci U S A 112:154-9

Showing the most recent 10 out of 27 publications