首页 | 本学科首页   官方微博 | 高级检索  
   检索      


c-Fms Signaling Mediates Neurofibromatosis Type-1 Osteoclast Gain-In-Functions
Authors:Yongzheng He  Steven D Rhodes  Shi Chen  Xiaohua Wu  Jin Yuan  Xianlin Yang  Li Jiang  Xianqi Li  Naoyuki Takahashi  Mingjiang Xu  Khalid S Mohammad  Theresa A Guise  Feng-Chun Yang
Institution:1. Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, United States of America.; 2. Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America.; 3. Division of Hard Tissue Research, Matsumoto Dental University, Shiojiri, Nagano, Japan.; 4. Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, United States of America.; University of Toronto, Canada,
Abstract:Skeletal abnormalities including osteoporosis and osteopenia occur frequently in both pediatric and adult neurofibromatosis type 1 (NF1) patients. NF1 (Nf1) haploinsufficient osteoclasts and osteoclast progenitors derived from both NF1 patients and Nf1+/− mice exhibit increased differentiation, migration, and bone resorptive capacity in vitro, mediated by hyperactivation of p21Ras in response to limiting concentrations of macrophage-colony stimulating factor (M-CSF). Here, we show that M-CSF binding to its receptor, c-Fms, results in increased c-Fms activation in Nf1+/ osteoclast progenitors, mediating multiple gain-in-functions through the downstream effectors Erk1/2 and p90RSK. PLX3397, a potent and selective c-Fms inhibitor, attenuated M-CSF mediated Nf1+/− osteoclast migration by 50%, adhesion by 70%, and pit formation by 60%. In vivo, we administered PLX3397 to Nf1 +/ osteoporotic mice induced by ovariectomy (OVX) and evaluated changes in bone mass and skeletal architecture. We found that PLX3397 prevented bone loss in Nf1+/−-OVX mice by reducing osteoclast differentiation and bone resorptive activity in vivo. Collectively, these results implicate the M-CSF/c-Fms signaling axis as a critical pathway underlying the aberrant functioning of Nf1 haploinsufficient osteoclasts and may provide a potential therapeutic target for treating NF1 associated osteoporosis and osteopenia.
Keywords:
设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号