American Association for Cancer Research
Browse
15417786mcr180047-sup-195545_2_supp_4850192_p4vr3c.xlsx (11.79 kB)

Table S1: Primer sets used for cloning of N and C-V5 aromatase mutants from Aromatase Acetylation Patterns and Altered Activity in Response to Sirtuin Inhibition

Download (11.79 kB)
dataset
posted on 2023-04-03, 16:42 authored by Deborah Molehin, Isabel Castro-Piedras, Monica Sharma, Souad R. Sennoune, Daphne Arena, Pulak R. Manna, Kevin Pruitt

Supplementary Table 1: Primer sets used in cloning of aromatase into pcDNA3.1+ and those used to amplify V5-tagged aromatase in a pool of RNA and their expected DNA product size.

Funding

National Institutes of Health

History

ARTICLE ABSTRACT

Aromatase, a cytochrome P450 member, is a key enzyme involved in estrogen biosynthesis and is dysregulated in the majority of breast cancers. Studies have shown that lysine deacetylase inhibitors (KDI) decrease aromatase expression in cancer cells, yet many unknowns remain regarding the mechanism by which this occurs. However, advances have been made to clarify factors involved in the transcriptional regulation of the aromatase gene (CYP19A1). Yet, despite aromatase being a primary target for breast cancer therapy, its posttranslational regulation has been virtually unexplored. Acetylation is a posttranslational modification (PTM) known to alter the activity and stability of many oncoproteins, and given the role of KDIs in regulating aromatase expression, we postulate that aromatase acetylation acts as a novel posttranslational regulatory mechanism that impacts aromatase expression and/or activity in breast cancer. Liquid chromatography–tandem mass spectrometry (LC-MS/MS) analysis revealed that aromatase is basally acetylated on several lysine residues (108, 169, 242, 262, 334, 352, and 354) in MCF-7 cells, and treatment with a SIRT-1 inhibitor induced additional acetylation (376, 390, 440, and 448). These acetylated lysine residues are in regions critical for aromatase activity. Site-directed mutagenesis and overexpression studies demonstrated that K108R/Q or K440R/Q mutations significantly altered aromatase activity in breast cancer cells without altering its subcellular localization.Implications: These findings demonstrate a novel posttranslational regulation of aromatase and uncover novel anticancer effects of deacetylase inhibitors, thus providing new insight for ongoing development of deacetylase inhibitors as cancer therapeutics. Mol Cancer Res; 16(10); 1530–42. ©2018 AACR.

Usage metrics

    Molecular Cancer Research

    Licence

    Exports

    RefWorks
    BibTeX
    Ref. manager
    Endnote
    DataCite
    NLM
    DC