Abstract

Triple-negative breast cancer (TNBC) comprises ∼20% of all breast cancers and is the most aggressive mammary cancer subtype. Devoid of the estrogen and progesterone receptors, along with the receptor tyrosine kinase ERB2 (HER2), that define most mammary cancers, there are no targeted therapies for patients with TNBC. This, combined with a high metastatic rate and a lower 5-year survival rate than for other breast cancer phenotypes, means there is significant unmet need for new therapeutic strategies. Herein, the anti-neoplastic effects of the electrophilic fatty acid nitroalkene derivative, 10-nitro-octadec-9-enoic acid (nitro-oleic acid, NO2-OA), were investigated in multiple preclinical models of TNBC. NO2-OA reduced TNBC cell growth and viability in vitro, attenuated TNFα-induced TNBC cell migration and invasion, and inhibited the tumor growth of MDA-MB-231 TNBC cell xenografts in the mammary fat pads of female nude mice. The up-regulation of these aggressive tumor cell growth, migration, and invasion phenotypes is mediated in part by the constitutive activation of pro-inflammatory nuclear factor κB (NF-κB) signaling in TNBC. NO2-OA inhibited TNFα-induced NF-κB transcriptional activity in human TNBC cells and suppressed downstream NF-κB target gene expression, including the metastasis-related proteins intercellular adhesion molecule-1 and urokinase-type plasminogen activator. The mechanisms accounting for NF-κB signaling inhibition by NO2-OA in TNBC cells were multifaceted, as NO2-OA (a) inhibited the inhibitor of NF-κB subunit kinase β phosphorylation and downstream inhibitor of NF-κB degradation, (b) alkylated the NF-κB RelA protein to prevent DNA binding, and (c) promoted RelA polyubiquitination and proteasomal degradation. Comparisons with non-tumorigenic human breast epithelial MCF-10A and MCF7 cells revealed that NO2-OA more selectively inhibited TNBC function. This was attributed to more facile mechanisms for maintaining redox homeostasis in normal breast epithelium, including a more favorable thiol/disulfide balance, greater extents of multidrug resistance protein-1 (MRP1) expression, and greater MRP1-mediated efflux of NO2-OA-glutathione conjugates. These observations reveal that electrophilic fatty acid nitroalkenes react with more alkylation-sensitive targets in TNBC cells to inhibit growth and viability.

Highlights

  • Triple-negative breast cancer (TNBC) comprises ϳ20% of all breast cancers and is the most aggressive mammary cancer subtype

  • This study reports the inhibition of TNBC (MDA-MB-231 and MDA-MB468) cell proliferation, invasion, and metastasis by a synthetic homolog of an endogenous electrophilic NO2-FA found in species ranging from plants to humans (10-nitro-octadec-9-enoic acid, termed nitro-oleic acid and NO2-OA)

  • To examine whether NO2-OA preferentially inhibited TNBC cell growth, Hoechst 33258 was used for counting non-tumorigenic breast epithelial cells (MCF-10A), an ERϩ breast cancer cell line (MCF7), and two TNBC cell lines (MDA-MB-231 and MDAMB-468)

Read more

Summary

Edited by Alex Toker

Triple-negative breast cancer (TNBC) comprises ϳ20% of all breast cancers and is the most aggressive mammary cancer subtype. To more facile mechanisms for maintaining redox homeostasis in normal breast epithelium, including a more favorable thiol/ disulfide balance, greater extents of multidrug resistance protein-1 (MRP1) expression, and greater MRP1-mediated efflux of NO2-OA– glutathione conjugates These observations reveal that electrophilic fatty acid nitroalkenes react with more alkylation-sensitive targets in TNBC cells to inhibit growth and viability. Through transient post-translational modification (PTM) reactions with hyperreactive protein thiols, NO2-FA modulate signaling pathways involved in cell proliferation and inflammatory responses This occurs as a result of the alkylation of functionally significant Cys residues in transcriptional regulatory proteins, including the Kelch-like ECH-associated protein-1 (Keap1) regulator of nuclear factor (erythroid-derived-2)-like 2 (Nrf2) signaling, the nuclear lipid receptor peroxisome proliferator–activated receptor ␥ (PPAR␥), and NF-␬B [25,26,27]. These findings reveal that electrophilic NO2-FA can mediate chemotherapeutic actions in treating TNBC and possibly other inflammation-related cancers

Results
Discussion
Cell culture and reagents
Cell growth assay
Cell migration analysis
Cell invasion assay
Western blotting
GSH and GSSG extraction and analysis
Statistical analysis
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