Peroxisome proliferator-activated receptor-delta induces cell proliferation by a cyclin E1-dependent mechanism and is up-regulated in thyroid tumors

Cancer Res. 2008 Aug 15;68(16):6578-86. doi: 10.1158/0008-5472.CAN-08-0855.

Abstract

Peroxisome proliferator-activated receptors (PPARs) are lipid-sensing nuclear receptors that have been implicated in multiple physiologic processes including cancer. Here, we determine that PPARdelta induces cell proliferation through a novel cyclin E1-dependent mechanism and is up-regulated in many human thyroid tumors. The expression of PPARdelta was induced coordinately with proliferation in primary human thyroid cells by the activation of serum, thyroid-stimulating hormone/cyclic AMP, or epidermal growth factor/mitogen-activated protein kinase mitogenic signaling pathways. Engineered overexpression of PPARdelta increased thyroid cell number, the incorporation of bromodeoxyuridine, and the phosphorylation of retinoblastoma protein by 40% to 45% in just 2 days, one usual cell population doubling. The synthetic PPARdelta agonist GW501516 augmented these PPARdelta proliferation effects in a dose-dependent manner. Overexpression of PPARdelta increased cyclin E1 protein by 9-fold, whereas knockdown of PPARdelta by small inhibitory RNA reduced both cyclin E1 protein and cell proliferation by 2-fold. Induction of proliferation by PPARdelta was abrogated by knockdown of cyclin E1 by small inhibitory RNA in primary thyroid cells and by knockout of cyclin E1 in mouse embryo fibroblasts, confirming a cyclin E1 dependence for this PPARdelta pathway. In addition, the mean expression of native PPARdelta was increased by 2-fold to 5-fold (P < 0.0001) and correlated with that of the in situ proliferation marker Ki67 (R = 0.8571; P = 0.02381) in six different classes of benign and malignant human thyroid tumors. Our experiments identify a PPARdelta mechanism that induces cell proliferation through cyclin E1 and is regulated by growth factor and lipid signals. The data argue for systematic investigation of PPARdelta antagonists as antineoplastic agents and implicate altered PPARdelta-cyclin E1 signaling in thyroid and other carcinomas.

Publication types

  • Research Support, N.I.H., Extramural
  • Research Support, Non-U.S. Gov't

MeSH terms

  • Adenoma / metabolism
  • Adenoma / pathology
  • Adenoma, Oxyphilic / metabolism
  • Adenoma, Oxyphilic / secondary
  • Carcinoma / metabolism
  • Carcinoma / secondary
  • Carcinoma, Papillary / metabolism
  • Carcinoma, Papillary / secondary
  • Cell Differentiation
  • Cell Proliferation*
  • Cells, Cultured
  • Cyclic AMP / pharmacology
  • Cyclin E / metabolism*
  • Epithelial Cells / metabolism
  • Humans
  • Immunoblotting
  • Immunoenzyme Techniques
  • Oncogene Proteins / metabolism*
  • PPAR delta / antagonists & inhibitors
  • PPAR delta / genetics
  • PPAR delta / metabolism*
  • Phosphorylation
  • RNA, Messenger / genetics
  • RNA, Messenger / metabolism
  • RNA, Small Interfering / pharmacology
  • Retinoblastoma Protein / metabolism
  • Reverse Transcriptase Polymerase Chain Reaction
  • Signal Transduction
  • Thiazoles / pharmacology
  • Thyroid Gland / metabolism
  • Thyroid Neoplasms / metabolism*
  • Thyroid Neoplasms / pathology*
  • Thyrotropin / pharmacology
  • Tissue Array Analysis

Substances

  • CCNE1 protein, human
  • Cyclin E
  • GW 501516
  • Oncogene Proteins
  • PPAR delta
  • RNA, Messenger
  • RNA, Small Interfering
  • Retinoblastoma Protein
  • Thiazoles
  • Thyrotropin
  • Cyclic AMP