Abstract

Activation of p38 MAPK is a key pathway for cell proliferation and differentiation in breast cancer and thyroid cells. The sodium/iodide symporter (NIS) concentrates iodide in the thyroid and lactating breast. All-trans-retinoic acid (tRA) markedly induces NIS activity in some breast cancer cell lines and promotes uptake of β-emitting radioiodide (131)I sufficient for targeted cytotoxicity. To identify a signal transduction pathway that selectively stimulates NIS expression, we investigated regulation by the Rac1-p38 signaling pathway in MCF-7 breast cancer cells and compared it with regulation in FRTL-5 rat thyroid cells. Loss of function experiments with pharmacologic inhibitors and small interfering RNA, as well as RT-PCR analysis of p38 isoforms, demonstrated the requirement of Rac1, MAPK kinase 3B, and p38β for the full expression of NIS in MCF-7 cells. In contrast, p38α was critical for NIS expression in FRTL-5 cells. Treatment with tRA or overexpression of Rac1 induced the phosphorylation of p38 isoforms, including p38β. A dominant negative mutant of Rac1 abolished tRA-induced phosphorylation in MCF-7 cells. Overexpression of p38β or Rac1 significantly enhanced (1.9- and 3.9-fold, respectively), the tRA-stimulated NIS expression in MCF-7 cells. This study demonstrates differential regulation of NIS by distinct p38 isoforms in breast cancer cells and thyroid cells. Targeting isoform-selective activation of p38 may enhance NIS induction, resulting in higher efficacy of (131)I concentration and treatment of breast cancer.

Highlights

  • Induction of the iodide transporter in cancer cells confers targeted cytotoxicity with radioiodide

  • These data indicate that p38 ␣ and/or ␤ play a major role in mediating trans-retinoic acid (tRA) induction of NIS expression and iodide uptake in MCF-7 cells

  • To investigate if Rac1 plays an important role in NIS expression in MCF-7 cells, we added a pharmacological Rac1 inhibitor, NSC23766. tRA-induced iodide uptake was significantly decreased by NSC23766 (ϳ66% reduction compared with tRA only), and the inhibitory effect was enhanced by the addition of the p38 inhibitor SB203580 (Fig. 5A)

Read more

Summary

Background

Induction of the iodide transporter in cancer cells confers targeted cytotoxicity with radioiodide. Conclusion: p38 isoform-specific stimulation may induce iodide uptake sufficient for radioiodide therapy in breast cancer. All-trans-retinoic acid (tRA) markedly induces NIS activity in some breast cancer cell lines and promotes uptake of ␤-emitting radioiodide 131I sufficient for targeted cytotoxicity. To identify a signal transduction pathway that selectively stimulates NIS expression, we investigated regulation by the Rac1-p38 signaling pathway in MCF-7 breast cancer cells and compared it with regulation in FRTL-5 rat thyroid cells. Stimulation of p38 MAPK activity is required for TSH-induced NIS expression in thyroid cells [21] as well as tRA-induced NIS expression in MCF-7 cells [17]. The Rac1-p38 pathway is upregulated by TSH-cAMP stimulation in thyroid cells [21] and by tRA treatment in MCF-7 breast cancer cells [18]. The effect of tRA treatment on the NIS-selective p38 signal transduction pathway was investigated

EXPERIMENTAL PROCEDURES
RESULTS
DISCUSSION
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