Abstract B70: Developing a new and potent anti-K-RAS strategy by inhibiting SIAH E3 ligase, the most downstream “gatekeeper” in the oncogenic K-RAS signal pathway, to block well-established pancreatic tumor growth

Author(s):  
Vasilena Zheleva ◽  
Minglei Bian ◽  
Xiaofei Gao ◽  
Justin J. Odanga ◽  
Monicah M. Njogu ◽  
...  
2010 ◽  
Vol 48 (08) ◽  
Author(s):  
N Azoitei ◽  
GV Pusapati ◽  
A Kleger ◽  
C Brunner ◽  
F Genze ◽  
...  

2012 ◽  
Vol 38 (12) ◽  
pp. 1121-1131
Author(s):  
Xiao-Hui WANG ◽  
Ya-Min ZHENG ◽  
Ye-Qing CUI ◽  
Shuang LIU ◽  
Hai-Chen SUN ◽  
...  

2021 ◽  
Author(s):  
I Pozios ◽  
E Günzler ◽  
N Hering ◽  
M Arndt ◽  
C Kamphues ◽  
...  

2010 ◽  
Vol 78 (2) ◽  
pp. 226-236 ◽  
Author(s):  
Indira Jutooru ◽  
Gayathri Chadalapaka ◽  
Maen Abdelrahim ◽  
Md Riyaz Basha ◽  
Ismael Samudio ◽  
...  

Oncotarget ◽  
2015 ◽  
Vol 6 (14) ◽  
pp. 12174-12187 ◽  
Author(s):  
Jianguo Hu ◽  
Ying Meng ◽  
Tinghe Yu ◽  
Lina Hu ◽  
Ming Mao

2008 ◽  
Vol 68 (11) ◽  
pp. 4360-4368 ◽  
Author(s):  
Hongkui Jin ◽  
Renhui Yang ◽  
Zhong Zheng ◽  
Mally Romero ◽  
Jed Ross ◽  
...  

Cancers ◽  
2020 ◽  
Vol 12 (6) ◽  
pp. 1412
Author(s):  
Chang Woo Han ◽  
Mi Suk Jeong ◽  
Sung Chul Ha ◽  
Se Bok Jang

Kirsten-RAS (KRAS) has been the target of drugs because it is the most mutated gene in human cancers. Because of the low affinity of drugs for KRAS mutations, it was difficult to target these tumor genes directly. We found a direct interaction between KRAS G12V and tumor suppressor novel H-REV107 peptide with high binding affinity. We report the first crystal structure of an oncogenic mutant, KRAS G12V-H-REV107. This peptide was shown to interact with KRAS G12V in the guanosine diphosphate (GDP)-bound inactive state and to form a stable complex, blocking the activation function of KRAS. We showed that the peptide acted as an inhibitor of mutant KRAS targets by [α-32P] guanosine triphosphate (GTP) binding assay. The H-REV107 peptide inhibited pancreatic cancer and colon cancer cell lines in cell proliferation assay. Specially, the H-REV107 peptide can suppress pancreatic tumor growth by reduction of tumor volume and weight in xenotransplantation mouse models. Overall, the results presented herein will facilitate development of novel drugs for inhibition of KRAS mutations in cancer patients.


Author(s):  
Mu-Su Pan ◽  
Hui Wang ◽  
Kamar Hasan Ansari ◽  
Xin-Ping Li ◽  
Wei Sun ◽  
...  

Abstract Background Cancer-associated fibroblasts (CAFs) and vasculogenic mimicry (VM) play important roles in the occurrence and development of tumors. However, the relationship between CAFs and VM formation, especially in gallbladder cancer (GBC) has not been clarified. In this study, we investigated whether gallbladder CAFs (GCAFs) can promote VM formation and tumor growth and explored the underlying molecular mechanism. Methods A co-culture system of human GBC cells and fibroblasts or HUVECs was established. VM formation, proliferation, invasion, migration, tube formation assays, CD31-PAS double staining, optic/electron microscopy and tumor xenograft assay were used to detect VM formation and malignant phenotypes of 3-D co-culture matrices in vitro, as well as the VM formation and tumor growth of xenografts in vivo, respectively. Microarray analysis was used to analyze gene expression profile in GCAFs/NFs and VM (+)/VM (−) in vitro. QRT-PCR, western blotting, IHC and CIF were used to detected NOX4 expression in GCAFs/NFs, 3-D culture/co-culture matrices in vitro, the xenografts in vivo and human gallbladder tissue/stroma samples. The correlation between NOX4 expression and clinicopathological and prognostic factors of GBC patients was analyzed. And, the underlying molecular mechanism of GCAFs promoting VM formation and tumor growth in GBC was explored. Results GCAFs promote VM formation and tumor growth in GBC; and the finding was confirmed by facts that GCAFs induced proliferation, invasion, migration and tube formation of GBC cells in vitro, and promoted VM formation and tumor growth of xenografts in vivo. NOX4 is highly expressed in GBC and its stroma, which is the key gene for VM formation, and is correlated with tumor aggression and survival of GBC patients. The GBC patients with high NOX4 expression in tumor cells and stroma have a poor prognosis. The underlying molecular mechanism may be related to the upregulation of NOX4 expression through paracrine IL-6 mediated IL-6/JAK/STAT3 signaling pathway. Conclusions GCAFs promote VM formation and tumor growth in GBC via upregulating NOX4 expression through the activation of IL-6-JAK-STAT3 signal pathway. NOX4, as a VM-related gene in GBC, is overexpressed in GBC cells and GCAFs, which is related to aggression and unfavorable prognosis of GBC patients.


2020 ◽  
Vol 401 ◽  
pp. 115118
Author(s):  
Ye Yao ◽  
Qing-yu Yao ◽  
Jun-sheng Xue ◽  
Xiu-yun Tian ◽  
Qi-ming An ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document