Abstract

Prostate cancer (PCA) kills thousands of men every year, demanding additional approaches to better understand and target this malignancy. Recently, critical role of aberrant lipogenesis is highlighted in prostate carcinogenesis, offering a unique opportunity to target it to reduce PCA. Here, we evaluated efficacy and associated mechanisms of silibinin in inhibiting lipid metabolism in PCA cells. At physiologically achievable levels in human, silibinin strongly reduced lipid and cholesterol accumulation specifically in human PCA cells but not in non-neoplastic prostate epithelial PWR-1E cells. Silibinin also decreased nuclear protein levels of sterol regulatory element binding protein 1 and 2 (SREBP1/2) and their target genes only in PCA cells. Mechanistically, silibinin activated AMPK, thereby increasing SREBP1 phosphorylation and inhibiting its nuclear translocation; AMPK inhibition reversed silibinin-mediated decrease in nuclear SREBP1 and lipid accumulation. Additionally, specific SREBP inhibitor fatostatin and stable overexpression of SREBP1 further confirmed the central role of SREBP1 in silibinin-mediated inhibition of PCA cell proliferation and lipid accumulation and cell cycle arrest. Importantly, silibinin also inhibited synthetic androgen R1881-induced lipid accumulation and completely abrogated the development of androgen-independent LNCaP cell clones via targeting SREBP1/2. Together, these mechanistic studies suggest that silibinin would be effective against PCA by targeting critical aberrant lipogenesis.

Highlights

  • Dysregulated metabolism is accepted as one of the hallmarks of cancer [1], and metabolic alterations supporting high growth rate and energy requirements are essential for the sustained tumor growth and progression

  • Since lipid synthesis in prostate cancer (PCA) cells is controlled by androgens, and under low androgen conditions, lipogenesis regulators play an important role in androgen biosynthesis [27, 28], we examined silibinin effect on androgeninduced lipid accumulation as well as lipogenesis regulators (SREBP1/2) expression under low androgen conditions

  • In order to understand how PCA cells are unique in terms of their metabolic profile, we first evaluated a series of prostate/PCA cell lines for their glucose and fat uptake rates as well as endogenous lipid levels

Read more

Summary

Introduction

Dysregulated metabolism is accepted as one of the hallmarks of cancer [1], and metabolic alterations supporting high growth rate and energy requirements are essential for the sustained tumor growth and progression. During androgen deprivation therapy, lipids (cholesterol) play an important role in the de novo synthesis of androgens by PCA cells, providing them self-sufficiency in androgen receptor (AR) signaling and hormonerefractory progression [10, 11]. This unique dependence of PCA cells on lipids for their growth and progression provides an excellent opportunity to reduce PCA burden via inhibiting lipogenesis and associated molecular regulators using non-toxic small molecules. In the present study, employing both PCA and non-neoplastic prostate epithelial cells, for the first time we examined the detailed effect of silibinin on cellular lipid levels as well as molecular regulators of lipogenesis

Methods
Results
Conclusion
Full Text
Paper version not known

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

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.