Loss of Kelch-like ECH-associated protein 1 function in prostate cancer cells causes chemoresistance and radioresistance and promotes tumor growth

P Zhang, A Singh, S Yegnasubramanian, D Esopi… - Molecular cancer …, 2010 - AACR
P Zhang, A Singh, S Yegnasubramanian, D Esopi, P Kombairaju, M Bodas, H Wu, SG Bova
Molecular cancer therapeutics, 2010AACR
Loss-of-function mutations in the nuclear factor erythroid-2–related factor 2 (Nrf2) inhibitor
Kelch-like ECH-associated protein 1 (Keap1) result in increased Nrf2 activity in non–small
cell lung cancer and confer therapeutic resistance. We detected point mutations in Keap1
gene, leading to nonconservative amino acid substitutions in prostate cancer cells. We
found novel transcriptional and posttranscriptional mechanisms of Keap1 inactivation, such
as promoter CpG island hypermethylation and aberrant splicing of Keap1, in DU-145 cells …
Abstract
Loss-of-function mutations in the nuclear factor erythroid-2–related factor 2 (Nrf2) inhibitor Kelch-like ECH-associated protein 1 (Keap1) result in increased Nrf2 activity in non–small cell lung cancer and confer therapeutic resistance. We detected point mutations in Keap1 gene, leading to nonconservative amino acid substitutions in prostate cancer cells. We found novel transcriptional and posttranscriptional mechanisms of Keap1 inactivation, such as promoter CpG island hypermethylation and aberrant splicing of Keap1, in DU-145 cells. Very low levels of Keap1 mRNA were detected in DU-145 cells, which significantly increased by treatment with DNA methyltransferase inhibitor 5-aza-deoxycytidine. The loss of Keap1 function led to an enhanced activity of Nrf2 and its downstream electrophile/drug detoxification pathway. Inhibition of Nrf2 expression in DU-145 cells by RNA interference attenuated the expression of glutathione, thioredoxin, and the drug efflux pathways involved in counteracting electrophiles, oxidative stress, and detoxification of a broad spectrum of drugs. DU-145 cells constitutively expressing Nrf2 short hairpin RNA had lower levels of total glutathione and higher levels of intracellular reactive oxygen species. Attenuation of Nrf2 function in DU-145 cells enhanced sensitivity to chemotherapeutic drugs and radiation-induced cell death. In addition, inhibition of Nrf2 greatly suppressed in vitro and in vivo tumor growth of DU-145 prostate cancer cells. Thus, targeting the Nrf2 pathway in prostate cancer cells may provide a novel strategy to enhance chemotherapy and radiotherapy responsiveness and ameliorate the growth and tumorigenicity, leading to improved clinical outcomes. Mol Cancer Ther; 9(2); 336–46
AACR