Abstract

Loss of ovarian function is closely related to estrogen (E2) deficiency, which is responsible for increased osteoclast (OC) differentiation and activity. We aimed to investigate the action mechanism of E2 to decrease bone resorption in OCs to protect from ovariectomy (OVX)-induced bone loss in mice. In vivo, tartrate-resistant acid phosphatase (TRAP) staining in femur and serum carboxy-terminal collagen crosslinks-1 (CTX-1) were analyzed upon E2 injection after OVX in mice. In vitro, OCs were analyzed by TRAP staining, actin ring formation, carboxymethylation, determination of reactive oxygen species (ROS) level, and immunoprecipitation coupled with Western blot. In vivo and in vitro, E2 decreased OC size more dramatically than OC number and Methyl-piperidino-pyrazole hydrate dihydrochloride (MPPD), an estrogen receptor alpha (ERα) antagonist, augmented the OC size. ERα was found in plasma membranes and E2/ERα signaling affected receptor activator of nuclear factor κB ligand (RANKL)-induced actin ring formation by rapidly decreasing a proto-oncogene tyrosine-protein kinase, cellular sarcoma (c-Src) (Y416) phosphorylation in OCs. E2 exposure decreased physical interactions between NADPH oxidase 1 (NOX1) and the oxidized form of c-Src homology 2 (SH2)-containing protein tyrosine phosphatase 2 (SHP2), leading to higher levels of reduced SHP2. ERα formed a complex with the reduced form of SHP2 and c-Src to decrease c-Src activation upon E2 exposure, which blocked a signal for actin ring formation by decreased Vav guanine nucleotide exchange factor 3 (Vav3) (p–Y) and Ras-related C3 botulinum toxin substrate 1 (Rac1) (GTP) activation in OCs. E2/ERα signals consistently inhibited bone resorption in vitro. In conclusion, our study suggests that E2-binding to ERα forms a complex with SHP2/c-Src to attenuate c-Src activation that was induced upon RANKL stimulation in a non-genomic manner, resulting in an impaired actin ring formation and reducing bone resorption.

Highlights

  • Within the skeleton, constant remodeling and repairing of old bones is required to ensure structural integrity

  • Our present findings suggest that E2 binding to estrogen receptor α (ERα) formed a complex with active

  • Dephosphorylation of cc-Src was followed by the blockade of Vav guanine nucleotide exchange factor 3 (Vav3) and related C3 botulinum toxin substrate 1 (Rac1) activation by RANKL stimulation

Read more

Summary

Introduction

Constant remodeling and repairing of old bones is required to ensure structural integrity. Osteoclasts (OCs) require two essential factors; macrophage-colony stimulating factor (M-CSF) and RANKL. M-CSF stimulates mainly OCs survival and proliferation as well as activation through cross-talking with RANKL [2]. A family member of tumor necrosis factor receptor, RANK expresses on OC precursor cells as a transmembrane signaling receptor for RANKL to result in expression of OC-specific genes, activation of bone resorption, and OC survival [3]. The degree of bone resorption reflects the number and matrix-degrading activity of OCs [4]. The number of OCs is regulated by OC differentiation as well as OC survival. While the functional activity of mature OCs in bone

Objectives
Methods
Results
Discussion
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call