1. Academic Validation
  2. RhoA enhances osteosarcoma resistance to MPPa-PDT via the Hippo/YAP signaling pathway

RhoA enhances osteosarcoma resistance to MPPa-PDT via the Hippo/YAP signaling pathway

  • Cell Biosci. 2021 Oct 9;11(1):179. doi: 10.1186/s13578-021-00690-6.
Fangbiao Zhan 1 2 Tao He 1 3 Zhiyu Chen 1 Qiang Zuo 1 Yang Wang 1 Qiaochu Li 1 Shenxi Zhong 1 Yunsheng Ou 4
Affiliations

Affiliations

  • 1 Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing, 400016, China.
  • 2 Department of Orthopedics, Chongqing University Three Gorges Hospital, Wanzhou, Chongqing, 404000, China.
  • 3 Department of Orthopaedic, Chongqing General Hospital, University of Chinese Academy of Sciences, Chongqing, 401147, China.
  • 4 Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing, 400016, China. ouyunsheng2001@163.com.
Abstract

Background: Osteosarcoma (OS) is the most prevalent primary bone malignancy affecting adolescents, yet the emergence of chemoradiotherapeutic resistance has limited efforts to cure affected patients to date. Pyropheophorbide-α methyl ester-mediated photodynamic therapy (MPPa-PDT) is a recently developed, minimally invasive treatment for OS that is similarly constrained by such therapeutic resistance. This study sought to explore the mechanistic basis for RhoA-activated YAP1 (YAP)-mediated resistance in OS.

Methods: The relationship between YAP expression levels and patient prognosis was analyzed, and YAP levels in OS cell lines were quantified. Immunofluorescent staining was used to assess YAP nuclear translocation. OS cell lines (HOS and MG63) in which RhoA and YAP were knocked down or overexpressed were generated using lentiviral vectors. CCK-8 assays were used to examine OS cell viability, while the apoptotic death of these cells was monitored via Hoechst staining, Western blotting, and flow cytometry. Tumor-bearing nude mice were additionally used to assess the relationship between lentivirus-mediated alterations in RhoA expression and MPPa-PDT treatment outcomes. TUNEL and immunohistochemical staining approaches were leveraged to assess apoptotic cell death in tissue samples.

Results: OS patients exhibited higher levels of YAP expression, and these were correlated with a poor prognosis. MPPa-PDT induced Apoptosis in OS cells, and such MPPa-PDT-induced Apoptosis was enhanced following YAP knockdown whereas it was suppressed by YAP overexpression. RhoA and YAP expression levels were positively correlated in OS patients, and both active and total RhoA protein levels rose in OS cells following MPPa-PDT treatment. When RhoA was knocked down, levels of unphosphorylated YAP and downstream target genes were significantly reduced, while RhoA/ROCK2/LIMK2 pathway phosphorylation was suppressed, whereas RhoA overexpression resulted in the opposite phenotype. MPPa-PDT treatment was linked to an increase in HMGCR protein levels, and the inhibition of RhoA or HMGCR was sufficient to suppress RhoA activity and to decrease the protein levels of YAP and its downstream targets. Mevalonate administration partially reversed these reductions in the expression of YAP and YAP target genes. RhoA knockdown significantly enhanced the apoptotic death of OS cells in vitro and in vivo following MPPa-PDT treatment, whereas RhoA overexpression had the opposite effect.

Conclusions: These results suggest that the mevalonate pathway activates RhoA, which in turn activates YAP and promotes OS cell resistance to MPPa-PDT therapy. Targeting the RhoA/ROCK2/LIMK2/YAP pathway can significantly improve the efficacy of MPPa-PDT treatment for OS.

Keywords

Apoptosis; MPPa-PDT; Mevalonate; Osteosarcoma; RhoA; YAP.

Figures
Products