Abstract P2-06-02: Pparg deacetylation by SIRT1 determines breast tumor lipid synthesis and growth

2013 
Peroxisome proliferator-activated receptorg (Pparγ) is a member of the nuclear receptor (NR) superfamily, which regulates diverse biological functions including lipogenesis and differentiation, anti-inflammation, insulin sensitivity, cellular proliferation, and autophagy. Independent lines of evidence support a role for Pparγ as either a collaborative oncogene or as a tumor suppressor. Heterozygous mutations of Pparγ have been detected in 4/55 patients with colon cancer and a chromosomal translocation between PAX8 and Pparγ in follicular thyroid cancer appeared to serve as a dominant inhibitor of endogenous Pparγ expression. Pparγ agonists reduced tumorigenesis in several in vivo models. In contrast, several studies suggest Pparγ may enhance tumor growth. Pparγ ligands increased polyp numbers in the Apc mouse model of familial adenomatosis. Pparγ and its ligands inhibit breast tumor growth; however, constitutively active Pparγ collaborated in mammary oncogenesis with polyoma middle T antigen or oncogenic ErbB2. Pparγ activation involves post-translational modifications including phosphorylation and sumoylation upon growth factor or ligand stimulus. Mutation of the Pparγ1 sumoylation site at K77 and K365 demonstrated that K77 may either reduce Pparγ-dependent gene induction and enhance repression or reduce repression, depending upon the synthetic reporter gene used. Lysine residues of nuclear receptors also serve as substrates for acetylation and Pparγ binds co-activators and co-repressors with intrinsic or associated histone acetylase or deacetylase activity including NCoR, SMRT, SIRT1, and p300. Initially characterized for the ERα, AR and, subsequently, the orphan nuclear receptor steroidogenic factor 1 (SF-1), acetylation occurs at a conserved lysine motif shared amongst evolutionarily related nuclear receptors. Several nuclear receptors and co-integrators involved in lipid metabolism are regulated by acetylation including p300, PGC1α, FXR, LXR and RAR. Both TSA- and NAD-sensitive HDACs (e.g. SIRT1) regulate Pparγ function and SIRT1 inhibits Pparγ-dependent adipocyte differentiation. Whether Pparγ is acetylated in cancer cells and how Pparγ exerts it9s crucial, though controversial, function in tumorigenesis have not been established. Pparγ induces gene transcription through binding specific NR half-sites and through non-canonical binding sequences (such as CREB/AP-1 sites). Transcriptional repression involves Pparγ sumoylation at lysine 77 (K77). Herein, Pparγ was shown to be acetylated at nine distinct lysine residues. SIRT1 bound and deacetylated Pparγ at K154/155. ChIP-Seq analysis for genome-wide DNA binding demonstrated the acetylation site was required for binding NR half-sites, but was not required for non-canonical site binding. Breast tumor growth, de novo lipid synthesis, induction of autophagy and evasion of apoptosis was promoted by K154/155 and inhibited by K77 in vivo. Pparγ acetylation induced a gene signature that was increased in breast cancer, associated with a reduction in SIRT1 abundance and poor outcome. The Pparγ acetylation site determines binding to autophagy and apoptosis signaling to regulate breast tumor lipid metabolism and growth. Citation Information: Cancer Res 2013;73(24 Suppl): Abstract nr P2-06-02.
    • Correction
    • Source
    • Cite
    • Save
    • Machine Reading By IdeaReader
    0
    References
    2
    Citations
    NaN
    KQI
    []