Abstract

Background: The present study was designed to explore the underlying role of hypoxia-inducible factor 1α (HIF-1α) in reactive oxygen species (ROS) formation and apoptosis in osteosarcoma (OS) cells induced by hypoxia. Methods: In OS cells, ROS accumulated and apoptosis increased within 24 h after exposure to low HIF-1α expression levels. A co-expression analysis showed that HIF was positively correlated with Forkhead box class O1 (FoxO1) expression and negatively correlated with CYP-related genes from the National Center for Biotechnology Information’s Gene Expression Omnibus (NCBI GEO) datasets. Hypoxia also considerably increased HIF-1α and FoxO1 expression. Moreover, the promoter region of FoxO1 was directly regulated by HIF-1α. We inhibited HIF-1α via siRNA and found that the ROS accumulation and apoptosis induced by hypoxia in OS cells decreased. In this study, a murine xenograft model of BALB-c nude mice was adopted to test tumour growth and measure the efficacy of 2-ME + As2O3 treatment. Results: Ad interim knockdown of HIF-1α also inhibited manganese-dependent superoxide dismutase (MnSOD), catalase and sestrin 3 (Sesn3) expression in OS cells. Furthermore, hypoxia-induced ROS formation and apoptosis in OS cells were associated with CYP450 protein interference and were ablated by HIF-1α silencing via siRNA. Conclusions: Our data reveal that HIF-1α inhibits ROS accumulation by directly regulating FoxO1 in OS cells, which induces MnSOD, catalase and Sesn3 interference, thus resulting in anti-oxidation effects. The combination of an HIF-1α inhibitor (2-mercaptoethanol,2-ME) and ROS inducer (arsenous oxide, As2O3) can prohibit proliferation and migration and promote apoptosis in MG63 cells in vitro while inhibiting tumour growth in vivo.

Highlights

  • Osteosarcoma (OS) is the most common primary bone cancer and is one of the leading causes of mortalities related to cancer in paediatric patients [1]

  • OS cell apoptosis induced through hypoxia injury is a significant cellular biological event in this disease [23]

  • The underlying mechanism of hypoxia injury mediated by OS cells has not been completely understood

Read more

Summary

Introduction

Osteosarcoma (OS) is the most common primary bone cancer and is one of the leading causes of mortalities related to cancer in paediatric patients [1]. FoxO factors are involved in regulating the genes associated with cellular cycle progression and inducing apoptosis [10]. The genome within the mitochondria encodes 12 proteins closely associated with biogenetic activities [19] The expression of these genes is regulated by a network of transcription factors, such as Saccharomyces cerevisiae and mitochondrial transcription specificity factors, in up- and downstream signalling pathways. We present a comprehensive analysis of the transcriptional response to HIF-1α, revealing the repression of numerous nuclear-encoded mitochondrial genes through the regulation of FoxO1 function. Regulation of mitochondrial structure and function could be an important role for HIF-1α factors in regulating ROS production, and these processes can affect cellular adaptation to hypoxia. In an in vivo drug-sensitivity test, the combination of 2ME and As2 O3 achieved anti-tumour effects without obvious adverse reactions

Microarray Data
Osteosarcoma Specimens
Immunohistochemistry
Cell Lines and Culture Conditions
Western Blot Analysis
Plasmid Construction and Cell Transfection
Cell Proliferation Assay
Xenograft Transplantation
2.10. Statistical Analysis
HIF-1α and FoxO1 was were
HIF-1α Targeted FoxO1 Directly to Promote Its Expression in OS Cells
The ROS Metabolism Regulation Pathway Is Involved in HIF-1α-Silenced OS Cells
Discussion
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