Saikosaponin-d: A potential chemotherapeutics in castration resistant prostate cancer by suppressing cancer metastases and cancer stem cell phenotypes

2016 ◽  
Vol 474 (4) ◽  
pp. 722-729 ◽  
Author(s):  
Di Zhong ◽  
Hui-jian Zhang ◽  
Yao-dong Jiang ◽  
Peng Wu ◽  
Huan Qi ◽  
...  
2018 ◽  
Vol 78 (8) ◽  
pp. 2026-2039 ◽  
Author(s):  
Younghun Jung ◽  
Frank C. Cackowski ◽  
Kenji Yumoto ◽  
Ann M. Decker ◽  
Jingcheng Wang ◽  
...  

2022 ◽  
Vol 11 ◽  
Author(s):  
Qimei Lin ◽  
Jiasong Cao ◽  
Xiaoling Du ◽  
Kuo Yang ◽  
Yongmei Shen ◽  
...  

Treatment of patients with castration-resistant prostate cancer (CRPC) remains a major clinical challenge. We previously showed that estrogenic effects contribute to CRPC progression and are primarily caused by the increased endogenous estradiol produced via highly expressed aromatase. However, the mechanism of aromatase upregulation and its role in CRPC are poorly described. In this study, we report that HeyL is aberrantly upregulated in CRPC tissues, and its expression is positively correlated with aromatase levels. HeyL overexpression increased endogenous estradiol levels and estrogen receptor-α (ERα) transcriptional activity by upregulating CYP19A1 expression, which encodes aromatase, enhancing prostate cancer stem cell (PCSC) properties in PC3 cells. Mechanistically, HeyL bound to the CYP19A1 promoter and activated its transcription. HeyL overexpression significantly promoted bicalutamide resistance in LNCaP cells, which was reversed by the aromatase inhibitor letrozole. In PC3 cells, the HeyL-aromatase axis promoted the PCSC phenotype by upregulating autophagy-related genes, while the autophagy inhibitor chloroquine (CQ) suppressed the aromatase-induced PCSC phenotype. The activated HeyL-aromatase axis promoted PCSC autophagy via ERα-mediated estrogenic effects. Taken together, our results indicated that the HeyL-aromatase axis could increase endogenous estradiol levels and activate ERα to suppress PCSC apoptosis by promoting autophagy, which enhances the understanding of how endogenous estrogenic effects influence CRPC development.


2021 ◽  
Vol 81 (22) ◽  
pp. 5777-5777
Author(s):  
Younghun Jung ◽  
Frank C. Cackowski ◽  
Kenji Yumoto ◽  
Ann M. Decker ◽  
Jingcheng Wang ◽  
...  

2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Meixia Che ◽  
Aashi Chaturvedi ◽  
Sarah A. Munro ◽  
Samuel P. Pitzen ◽  
Alex Ling ◽  
...  

AbstractEndocrine therapies for prostate cancer inhibit the androgen receptor (AR) transcription factor. In most cases, AR activity resumes during therapy and drives progression to castration-resistant prostate cancer (CRPC). However, therapy can also promote lineage plasticity and select for AR-independent phenotypes that are uniformly lethal. Here, we demonstrate the stem cell transcription factor Krüppel-like factor 5 (KLF5) is low or absent in prostate cancers prior to endocrine therapy, but induced in a subset of CRPC, including CRPC displaying lineage plasticity. KLF5 and AR physically interact on chromatin and drive opposing transcriptional programs, with KLF5 promoting cellular migration, anchorage-independent growth, and basal epithelial cell phenotypes. We identify ERBB2 as a point of transcriptional convergence displaying activation by KLF5 and repression by AR. ERBB2 inhibitors preferentially block KLF5-driven oncogenic phenotypes. These findings implicate KLF5 as an oncogene that can be upregulated in CRPC to oppose AR activities and promote lineage plasticity.


2012 ◽  
Vol 18 (8) ◽  
pp. 2352-2359 ◽  
Author(s):  
Jonathan Shamash ◽  
Jimmy Jacob ◽  
Samir Agrawal ◽  
Thomas Powles ◽  
Katherine Mutsvangwa ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document