scholarly journals An oxanthroquinone derivative that disrupts RAS plasma membrane localization inhibits cancer cell growth

2018 ◽  
Vol 293 (35) ◽  
pp. 13696-13706 ◽  
Author(s):  
Lingxiao Tan ◽  
Kwang-Jin Cho ◽  
Pratik Neupane ◽  
Robert J. Capon ◽  
John F. Hancock
2019 ◽  
Vol 19 (23) ◽  
pp. 2114-2127 ◽  
Author(s):  
Na Ye ◽  
Qingfeng Xu ◽  
Wanwan Li ◽  
Pingyuan Wang ◽  
Jia Zhou

: The Ras proteins play an important role in cell growth, differentiation, proliferation and survival by regulating diverse signaling pathways. Oncogenic mutant K-Ras is the most frequently mutated class of Ras superfamily that is highly prevalent in many human cancers. Despite intensive efforts to combat various K-Ras-mutant-driven cancers, no effective K-Ras-specific inhibitors have yet been approved for clinical use to date. Since K-Ras proteins must be associated to the plasma membrane for their function, targeting K-Ras plasma membrane localization represents a logical and potentially tractable therapeutic approach. Here, we summarize the recent advances in the development of K-Ras plasma membrane localization inhibitors including natural product-based inhibitors achieved from high throughput screening, fragment-based drug design, virtual screening, and drug repurposing as well as hit-to-lead optimizations.


2018 ◽  
Vol 25 (4) ◽  
pp. 453-469 ◽  
Author(s):  
Mark A White ◽  
Efrosini Tsouko ◽  
Chenchu Lin ◽  
Kimal Rajapakshe ◽  
Jeffrey M Spencer ◽  
...  

Despite altered metabolism being an accepted hallmark of cancer, it is still not completely understood which signaling pathways regulate these processes. Given the central role of androgen receptor (AR) signaling in prostate cancer, we hypothesized that AR could promote prostate cancer cell growth in part through increasing glucose uptake via the expression of distinct glucose transporters. Here, we determined that AR directly increased the expression ofSLC2A12, the gene that encodes the glucose transporter GLUT12. In support of these findings, gene signatures of AR activity correlated withSLC2A12expression in multiple clinical cohorts. Functionally, GLUT12 was required for maximal androgen-mediated glucose uptake and cell growth in LNCaP and VCaP cells. Knockdown of GLUT12 also decreased the growth of C4-2, 22Rv1 and AR-negative PC-3 cells. This latter observation corresponded with a significant reduction in glucose uptake, indicating that additional signaling mechanisms could augment GLUT12 function in an AR-independent manner. Interestingly, GLUT12 trafficking to the plasma membrane was modulated by calcium/calmodulin-dependent protein kinase kinase 2 (CaMKK2)-5′-AMP-activated protein kinase (AMPK) signaling, a pathway we previously demonstrated to be a downstream effector of AR. Inhibition of CaMKK2-AMPK signaling decreased GLUT12 translocation to the plasma membrane by inhibiting the phosphorylation of TBC1D4, a known regulator of glucose transport. Further, AR increasedTBC1D4expression. Correspondingly, expression ofTBC1D4correlated with AR activity in prostate cancer patient samples. Taken together, these data demonstrate that prostate cancer cells can increase the functional levels of GLUT12 through multiple mechanisms to promote glucose uptake and subsequent cell growth.


2006 ◽  
Vol 175 (4S) ◽  
pp. 257-257
Author(s):  
Jennifer Sung ◽  
Qinghua Xia ◽  
Wasim Chowdhury ◽  
Shabana Shabbeer ◽  
Michael Carducci ◽  
...  

Planta Medica ◽  
2012 ◽  
Vol 78 (11) ◽  
Author(s):  
F Epifano ◽  
S Genovese ◽  
P Lullo ◽  
S Fiorito ◽  
G Trivisonno ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document