Abstract

BackgroundTriple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer, with poor prognosis and limited treatment options. Hypoxia is a key hallmark of TNBC. Metabolic adaptation promotes progression of TNBC cells that are located within the hypoxic tumor regions. However, it is not well understood regarding the precise molecular mechanisms underlying the regulation of metabolic adaptions by hypoxia.MethodsRNA sequencing was performed to analyze the gene expression profiles in MDA-MB-231 cell line (20% O2 and 1% O2). Expressions of Slc6a8, which encodes the creatine transporter protein, were detected in breast cancer cells and tissues by quantitative real-time PCR. Immunohistochemistry was performed to detect SLC6A8 protein abundances in tumor tissues. Clinicopathologic correlation and overall survival were evaluated by chi-square test and Kaplan-Meier analysis, respectively. Cell viability assay and flow cytometry analysis with Annexin V/PI double staining were performed to investigate the impact of SLC6A8-mediated uptake of creatine on viability of hypoxic TNBC cells. TNBC orthotopic mouse model was used to evaluate the effects of creatine in vivo.ResultsSLC6A8 was aberrantly upregulated in TNBC cells in hypoxia. SLC6A8 was drastically overexpressed in TNBC tissues and its level was tightly associated with advanced TNM stage, higher histological grade and worse overall survival of TNBC patients. We found that SLC6A8 was transcriptionally upregulated by p65/NF-κB and mediated accumulation of intracellular creatine in hypoxia. SLC6A8-mediated accumulation of creatine promoted survival and suppressed apoptosis via maintaining redox homeostasis in hypoxic TNBC cells. Furthermore, creatine was required to facilitate tumor growth in xenograft mouse models. Mechanistically, intracellular creatine bolstered cell antioxidant defense by reducing mitochondrial activity and oxygen consumption rates to reduce accumulation of intracellular reactive oxygen species, ultimately activating AKT-ERK signaling, the activation of which protected the viability of hypoxic TNBC cells via mediating the upregulation of Ki-67 and Bcl-2, and the downregulation of Bax and cleaved Caspase-3.ConclusionsOur study indicates that SLC6A8-mediated creatine accumulation plays an important role in promoting TNBC progression, and may provide a potential therapeutic strategy option for treatment of SLC6A8 high expressed TNBC.

Highlights

  • Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer, with poor prognosis and limited treatment options

  • Our study indicates that Solute carrier family 6 member 8 (SLC6A8)-mediated creatine accumulation plays an important role in promoting TNBC progression, and may provide a potential therapeutic strategy option for treatment of SLC6A8 high expressed TNBC

  • SLC6A8 is upregulated in response to hypoxia in TNBC cells As a common existence in solid tumors, hypoxia is tightly associated with tumor progression and often leads to enhanced local invasiveness, altered metabolism, unregulated angiogenesis, incipient metastases and spread of cancer stem cells [22]

Read more

Summary

Introduction

Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer, with poor prognosis and limited treatment options. Metabolic adaptation promotes progression of TNBC cells that are located within the hypoxic tumor regions. It is not well understood regarding the precise molecular mechanisms underlying the regulation of metabolic adaptions by hypoxia. Breast cancers devoid of expression of estrogen receptor alpha (ERα), progesterone receptor (PR) and amplification of human epidermal growth factor receptor 2 (HER2) are classified as triple-negative breast cancer (TNBC), which constitutes about 15% of all breast cancers and is the most aggressive subtype of breast cancer [2]. Ample evidence suggests that altered metabolic profile is induced by hypoxia to gain an undeniable survival advantage for cancer cells, limiting patient prognosis [3]. Identification of the molecular mechanisms mediating metabolic alteration may be useful in developing novel TNBC therapy regimen

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