FRI-452-Synergistic reduction of cancer proliferation and tumour growth in advanced murine hepatocellular carcinoma vis skewing and activation of innate and adoptive immunity

2019 ◽  
Vol 70 (1) ◽  
pp. e594-e595
Author(s):  
Muhammad Ashfaq-Khan ◽  
Misbah Aslam ◽  
Asif Qureshi ◽  
Yong Ook Kim ◽  
Tim Zimmermann ◽  
...  
2019 ◽  
Vol 70 (1) ◽  
pp. e849-e850
Author(s):  
Anais Vallet Pichard ◽  
Correas Jean Michel ◽  
Dorival Celine ◽  
Fabien Zoulim ◽  
Albert Tran ◽  
...  

2017 ◽  
Vol 8 (1) ◽  
Author(s):  
Sam Sheppard ◽  
Joana Guedes ◽  
Anna Mroz ◽  
Anastasia-Maria Zavitsanou ◽  
Hiromi Kudo ◽  
...  

Author(s):  
Ruwanthi Kolamunnage-Dona ◽  
Sarah Berhane ◽  
Harry Potts ◽  
Edward H. Williams ◽  
James Tanner ◽  
...  

2016 ◽  
Vol 40 (3-4) ◽  
pp. 757-769 ◽  
Author(s):  
Li Zhang ◽  
Guozhan Jia ◽  
Binya Shi ◽  
Guanqun Ge ◽  
Hongbin Duan ◽  
...  

Background: Protease serine 8 (PRSS8), a trypsin-like serine peptidase, has been shown to function as a tumour suppressor in various malignancies. The present study aimed to investigate the expression pattern, prognostic value and the biological role of PRSS8 in human hepatocellular carcinoma (HCC). Methods: PRSS8 expression in 106 HCC surgical specimens was examined by Real-time polymerase chain reaction (PCR) and immunohistochemistry, and its clinical significance was analysed. The role of PRSS8 in cell proliferation, apoptosis and invasion were examined in vitro and in vivo. Results: PRSS8 mRNA and protein expression were decreased in most HCC tumours from that in matched adjacent non-tumour tissues. Low intratumoral PRSS8 expression was significantly correlated with poor overall survival (OS) in patients with HCC (P = 0.001). PRSS8 expression was an independent prognostic factor for OS (hazard ratio [HR] = 1.704, P = 0.009). Furthermore, restoring PRSS8 expression in high metastatic HCCLM3 cells significantly inhibited cell proliferation and invasion. In contrast, silencing PRSS8 expression in non-metastatic HepG2 cells significantly enhanced cell growth and invasion. Moreover, our in vivo data revealed that attenuated PRSS8 expression in HepG2 cells greatly promoted tumour growth, while overexpression of PRSS8 remarkably inhibited tumour growth in an HCCLM3 xenograft model. Enhanced cell growth and invasion ability mediated by the loss of PRSS8 expression was associated with downregulation of PTEN, Bax and E-cadherin and an upregulation in Bcl-2, MMP9 and N-cadherin. Conclusions: Our data demonstrate that PRSS8 may serve as a tumour suppressor in HCC progression, and represent a valuable prognostic marker and potential therapeutic target for HCC.


2020 ◽  
Vol 2020 ◽  
pp. 1-19 ◽  
Author(s):  
Jie Qi ◽  
Xiangyuan Chen ◽  
Qichao Wu ◽  
Jing Wang ◽  
Hao Zhang ◽  
...  

Background. Hepatocellular carcinoma (HCC) is a life-threatening cancer, and the Kelch-like ECH-associated protein 1 (Keap1)/NF-E2-related factor 2 (Nrf2)/antioxidant response element (ARE) signalling pathway plays a crucial role in apoptosis resistance in cancer cells. Fasting is reported to mediate tumour growth reduction and apoptosis. SET8 is involved in cancer proliferation, invasiveness, and migration. However, whether SET8 participates in fasting-mediated apoptosis in HCC remains unclear. Methods. We used immunohistochemical staining to analyse the expression of SET8, Keap1, and Nrf2 in HCC tissues. Cell viability, apoptosis, and cellular reactive oxygen species (ROS) were assessed, and Western blot and qPCR analyses were used to examine the expression of Keap1/Nrf2 in HCC cells under fasting, SET8 overexpression, and PGC1α overexpression conditions. Mass spectrometry, coimmunoprecipitation, and confocal microscopy were used to determine whether PGC1α interacts with SET8. In vivo experiments were performed to verify the conclusions from the in vitro experiments. Results. Our data indicate that SET8 expression is associated with poor survival in HCC patients. Both in vitro and in vivo results demonstrated that fasting decreased cell viability and downregulated expression of SET8, Nrf2, and downstream effectors of Nrf2, while it upregulated Keap1 expression, mediated ROS accumulation, and induced HCC cell apoptosis. These results were similar to what is observed in SET8-deficient cells. Furthermore, SET8 was found to interact with PGC1α, and both PGC1α and H4K20me1, a downstream target of SET8, were found to be enriched at the Keap1 promoter region. These two factors were further determined to attenuate Keap1 promoter activity. Conclusions. The results of our study demonstrate that fasting induces HCC apoptosis by inhibiting SET8 expression and that SET8 interacts with PGC1α to activate the Nrf2/ARE signalling pathway by inhibiting Keap1 expression.


Oncotarget ◽  
2014 ◽  
Vol 5 (13) ◽  
pp. 5113-5124 ◽  
Author(s):  
Li-Li Liu ◽  
Shi-Xun Lu ◽  
Min Li ◽  
Lin-Zi Li ◽  
Jia Fu ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document