Oncotarget

Research Papers:

Autocrine and paracrine STIP1 signaling promote osteolytic bone metastasis in renal cell carcinoma

Jiang Wang, Hongbo You, Jun Qi, Caihong Yang, Ye Ren and Hao Cheng _

PDF  |  HTML  |  Supplementary Files  |  How to cite

Oncotarget. 2017; 8:17012-17026. https://doi.org/10.18632/oncotarget.15222

Metrics: PDF 2535 views  |   HTML 2324 views  |   ?  


Abstract

Jiang Wang1, Hongbo You1, Jun Qi1, Caihong Yang1, Ye Ren1, Hao Cheng1

1Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R.China

Correspondence to:

Ye Ren, email: [email protected]

Hao Cheng, email: [email protected]

Keywords: RCC bone metastasis, stress-induced phosphoprotein 1(STIP1), STIP1-ALK2-SMAD1/5 signaling, STIP1-prion-ERK1/2 signaling, tumor microenvironment

Received: May 07, 2016     Accepted: January 04, 2017     Published: February 09, 2017

ABSTRACT

Bone metastases are responsible for some of the most devastating complications of renal cell carcinoma (RCC). However, pro-metastatic factors leading to the highly osteolytic characteristics of RCC bone metastasis have barely been explored. We previously developed novel bone-seeking RCC cell lines by the in vivo selection strategy and performed a comparative proteome analysis on their total cell lysate. Here, we focused on STIP1 (stress-induced phosphoprotein 1), the high up-regulated protein in the bone-seeking cells, and explored its clinical relevance and functions in RCC bone metastasis. We observed high levels of both intracellular and extracellular STIP1 protein in bone metastatic tissue samples. Elevated STIP1 mRNA in the primary RCC tumors remarkably correlated with worse clinical outcomes. Furthermore, both human recombinant STIP1 protein and anti-STIP1 neutralizing antibody were used in the functional studies. We found that 1) STIP1 protein on the extracellular surface of tumor cells promoted the proliferation and migration/invasion of RCC tumor cells through the autocrine STIP1-ALK2-SMAD1/5 pathway; and 2) STIP1 protein secreted into the extracellular tumor stromal area, promoted the differentiation of osteoclasts through the paracrine STIP1-PrPc-ERK1/2 pathway. Increased cathepsin K (CTSK), the key enzyme secreted by osteoclasts to degrade collagen and other matrix proteins during bone resorption was further detected in the differentiated osteoclasts. These results provide evidence of the great potential of STIP1 as a novel biomarker and therapeutic target in RCC bone metastasis.


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 15222