C/EBPα regulates osteoclast lineage commitment - PubMed (original) (raw)

C/EBPα regulates osteoclast lineage commitment

Wei Chen et al. Proc Natl Acad Sci U S A. 2013.

Abstract

Despite recent insights gained from the effects of targeted deletion of the Finkel-Biskis-Jinkins osteosarcoma oncogene (c-fos), Spleen focus-forming virus (SFFV) proviral integration 1 (PU.1), microphthalmia-associated transcription factor, NF-κB, and nuclear factor of activated cells cytoplasmic 1 (NFATc1) transcription factor genes, the mechanism underlying transcription factors specifying osteoclast (OC) lineage commitment from monocyte/macrophage remains unclear. To characterize the mechanism by which transcription factors regulate OC lineage commitment, we mapped the critical cis-regulatory element in the promoter of cathepsin K (Ctsk), which is expressed specifically in OCs, and found that CCAAT/enhancer binding protein α (C/EBPα) is the critical cis-regulatory element binding protein. Our results indicate that C/EBPα is highly expressed in pre- OCs and OCs. The combined presence of macrophage colony-stimulating factor and receptor activator of NF-κB ligand significantly induces high C/EBPα expression. Furthermore, C/EBPα(-/-) newborn mice exhibited impaired osteoclastogenesis, and a severe osteopetrotic phenotype, but unaffected monocyte/macrophage development. Impaired osteoclastogenesis of C/EBPα(-/-) mouse bone marrow cells can be rescued by c-fos overexpression. Ectopic expression of C/EBPα in mouse bone marrow cells and monocyte/macrophage cells, in the absence of receptor activator of NF-κB ligand, induces expression of receptor activator of NF-κB, c-fos, Nfatc1, and Ctsk, and it reprograms monocyte/macrophage cells to OC-like cells. Our results demonstrate that C/EBPα directly up-regulates c-fos expression. C/EBPα(+/-) mice exhibit an increase in bone density compared with C/EBPα(+/+) controls. These discoveries establish C/EBPα as the key transcriptional regulator of OC lineage commitment, providing a unique therapeutic target for diseases of excessive bone resorption, such as osteoporosis and arthritis.

PubMed Disclaimer

Conflict of interest statement

The authors declare no conflict of interest.

Figures

Fig. 1.

Fig. 1.

Identification of Ctsk CCRE and CCRE DNA binding protein as C/EBPα. (A) Activity of deletion mutants of the pCCAT construct was measured in RANKL-induced RAW64.7 cells. Schematic representation of each reporter construct is shown. Values are relative to the activity obtained with the largest promoter fragment (pCCAT-1474). CAT activities are normalized with the cotransfected β-gal activity. Experimental data are reported as mean ± SD of triplicate independent samples (n = 5, repeated three times). (B and C) Summary of site-specific mutagenesis of the Ctsk promoter binding site from −116 to −34 and characterization of the CCRE as a C/EBPα site. (D) Gel mobility shift experiment using a 32P-labeled −53 to −30 WT Ctsk promoter oligonucleotide probe in nuclear extracts prepared from RANKL-induced RAW264.7 cells, with and without 100-fold molar excess of unlabeled WT or mutant oligonucleotides as described on the left. NP, nuclear protein. (E) DNase I footprint assays identify a binding site between −34 to −51 in the −137 to −31 mouse Ctsk promoter. The protected region is indicated by a shaded box, which contains a C/EBPα site. DNase I digestion of the naked DNA incubated with (lane 1) and without (lane 2) RANKL-induced RAW264.7 cell nuclear extracts is shown. G + A Maxam–Gilbert reaction of the −137 to −31 promoter fragment is shown in lane 3.

Fig. 2.

Fig. 2.

C/EBPα is expressed in pre-OCs and OCs, and it is induced by RANKL. (A) Northern blot hybridization using a _C/EBPα_-specific probe in RANKL-induced and uninduced C/EBPα +/+ MBM, GCTs of bone, hSCs, rat osteoblast (ROB), a human macrophage cell line (U-937 cells), and C/EBPα +/+ mouse tissues. (B) Time-course Western blot analysis of C/EBPα expression in C/EBPα +/+ MBM cultured with M-CSF (20 ng/mL) alone for 2 d and then stimulated with M-CSF (10 ng/mL)/RANKL (10 ng/mL) for 0–120 h. (C) TRAP staining and immunostaining for Ctsk, C/EBPα, C/EBPβ, and C/EBPδ in GCTs of bone and C/EBPα+/+ tibiae sections. Normal serum is shown as a control. (D) Immunofluorescence staining of C/EBPα (green) and Ctsk (red) in C/EBPα+/+ tibiae indicates significant overlap, visualized as a yellow area in the merged image (n > 9). Inset is the magnified image of the boxed area.

Fig. 3.

Fig. 3.

Increased bone mass and defective osteoclastogenesis in _C/EBPα_−/− mice in vivo and in vitro. (A) Radiographic analysis of femora and tibiae indicates osteopetrosis in _C/EBPα_−/− newborn mice compared with C/EBPα+/+ newborn mice (n = 5, repeated three times). (B) Three-dimensional microcomputed tomography images of C/EBPα+/+ and _C/EBPα_−/− newborn mice femora (n = 5, repeated three times). (C) Quantification of bone volume/tissue volume and bone mineral density of C/EBPα+/+ and _C/EBPα_−/− newborn mice femora. *P < 0.05; ***_P_ < 0.001. (_D_) Histological sections of _C/EBPα_−/− newborn mice tibiae have an extended growth plates (arrows) and a dramatic reduction in TRAP+ OCs compared with _C/EBPα_+/+ controls (_n_ > 50). Boxed areas (Upper) are magnified (Lower). The histological sections of the same area of _C/EBPα_−/− newborn mice also show dramatically decreased TRAP+ OCs. (E) TRAP stain of RANKL-induced C/EBPα+/+ and _C/EBPα_−/− MBM. (F) TRAP stain of cocultures with MOBs and MBM obtained from _C/EBPα_−/− and C/EBPα+/+ mice.

Fig. 4.

Fig. 4.

C/EBPα KO reduces the expression of key OC regulators and marker genes. Western blot (A) and quantification of expression (B) of PU.1, Ctsk, c-Fos, and Nfatc1 in C_/EBPα_+/+ and C/EBPα_−/− MBM cells cultured with M-CSF (20 ng/mL) alone for 2 d and then stimulated with M-CSF (10 ng/mL)/RANKL (10 ng/mL) for 5 d to generate OC-like cells. *P < 0.05; **P < 0.01. (C) Time-course Western blot analysis of IκBα and p38 and their phosphorylated forms (p-IκBα and p-p38) in C/EBPα_+/+ and _C/EBPα_−/− MBM cells cultured with M-CSF (20 ng/mL) alone for 2 d and then stimulated with M-CSF (10 ng/mL)/RANKL (10 ng/mL) for 0–60 min.

Fig. 5.

Fig. 5.

_C/EBPα_−/− mice exhibit a dramatic increase in trabecular bone number, and _C/EBPα_−/− impaired osteoclastogenesis is rescued by ectopic expression of c-fos. (A) Compared with C/EBPα+/+ controls, _C/EBPα_−/− tibiae have extended growth plates (arrows) and a dramatic increase in mineralized tissue (bright blue regions), which indicates osteopetrosis (n = 3, repeated three times). (B) Histomorphometric analysis of tibiae from C/EBPα+/+ and _C/EBPα_−/− mice. *P < 0.05; **P < 0.01. N.S., not significant. (C and D) Impaired OC differentiation in C/EBPα_−/− MBM cells cultured with M-CSF/RANKL can be rescued by ectopic expression of c-fos using pBMN–_c-fos compared with control virus (pBMN-GFP). ***P < 0.001.

Fig. 6.

Fig. 6.

C/EBPα induces osteoclastogenesis in the absence of RANKL and activates the c-fos promoter. (A) Western blot and quantification of forced expression of C/EBPα in C_/EBPα_+/+ MBM stimulated with 20 ng/mL M-CSF for 24 h and then transduced with pBMN-C/EBPα for 96 h. Compared with the control group, C/EBPα expression increased significantly 96 h after cells were transduced with pBMN-C/EBPα. (B) GFP expression and TRAP expression in C_/EBPα_+/+ MBM in which C/EBPα is overexpressed (pBMN-C/EBPα) compared with the control (pBMN-GFP) after 96 h of stimulation with M-CSF (20 ng/mL) alone. (C) qPCR analysis in MBM cultured in M-CSF alone and transduced with pBMN-C_/EBPα_ (+) or with the vector control pBMN-GFP for 96 h. *P < 0.05; **P < 0.01; ***P < 0.001. (D) ChIP analysis of C/EBPα binding to the c-fosA, c-fosB, or upstream promoter region [c-fos (−5 Kb) as a negative control] in MBM stimulated with RANKL/M-CSF to generate mature OCs. (E) Consensus sequence alignment of putative C/EBPα binding sites of mouse, rat, and human c-fos promoter regions. C/EBPα activates c-Fos expression. (F) Truncated c-fos promoters were induced by C/EBPα. Luciferase reporter assays with truncated c-fos promoters indicated the presence of two C/EBPα binding sites (GL3B, control vector).

Fig. 7.

Fig. 7.

Ectopic expression of C/EBPα reprograms the monocyte/macrophage cell line to OC-like cells in the absence of RANKL. (A) Western blot of C/EBPα expression in RAW264.7 cells (not stimulated with M-CSF/RANKL) transfected with pcDNA3.1-C_/EBPα_ (+) or with the vector control pcDNA3.1 (−). (B) TRAP and Ctsk immunostaining in uninduced RAW264.7 cells in which C/EBPα is overexpressed [pcDNA3.1-C_/EBPα_ (+)] compared with the control [pcDNA3.1 (−)]. Semi-qPCR (C) and qPCR (D) analysis (25 cycles, Hprt served as a loading control) of genes important for monocytes/macrophages and OCs in uninduced RAW264.7 cells transfected with pcDNA3.1-C_/EBPα_ (+) or with the vector control pcDNA3.1 (−).*P < 0.05; **P < 0.01.

References

    1. Valledor AF, Borràs FE, Cullell-Young M, Celada A. Transcription factors that regulate monocyte/macrophage differentiation. J Leukoc Biol. 1998;63(4):405–417. - PubMed
    1. Zhao B, Ivashkiv LB. Negative regulation of osteoclastogenesis and bone resorption by cytokines and transcriptional repressors. Arthritis Res Ther. 2011;13(4):234. - PMC - PubMed
    1. Takayanagi H, et al. Induction and activation of the transcription factor NFATc1 (NFAT2) integrate RANKL signaling in terminal differentiation of osteoclasts. Dev Cell. 2002;3(6):889–901. - PubMed
    1. Iotsova V, et al. Osteopetrosis in mice lacking NF-kappaB1 and NF-kappaB2. Nat Med. 1997;3(11):1285–1289. - PubMed
    1. Grigoriadis AE, et al. c-Fos: A key regulator of osteoclast-macrophage lineage determination and bone remodeling. Science. 1994;266(5184):443–448. - PubMed

Publication types

MeSH terms

Substances

Grants and funding

LinkOut - more resources