scholarly journals LB916 Effects of the platelet-activating factor-receptor and microRNA-149 in lung cancer growth and therapy effectiveness

2020 ◽  
Vol 140 (7) ◽  
pp. B3
Author(s):  
S.J. Chauhan ◽  
A. Thyagarajan ◽  
R.P. Sahu
2020 ◽  
Vol 21 (22) ◽  
pp. 8517
Author(s):  
Shreepa J. Chauhan ◽  
Anita Thyagarajan ◽  
Yanfang Chen ◽  
Jeffrey B. Travers ◽  
Ravi P. Sahu

Microvesicle particles (MVP) secreted by a variety of cell types in response to reactive oxygen species (ROS)-generating pro-oxidative stressors have been implicated in modifying the cellular responses including the sensitivity to therapeutic agents. Our previous studies have shown that expression of a G-protein coupled, platelet-activating factor-receptor (PAFR) pathway plays critical roles in pro-oxidative stressors-mediated cancer growth and MVP release. As most therapeutic agents act as pro-oxidative stressors, the current studies were designed to determine the role of the PAFR signaling in targeted therapies (i.e., gefitinib and erlotinib)-mediated MVP release and underlying mechanisms using PAFR-expressing human A549 and H1299 non-small cell lung cancer (NSCLC) cell lines. Our studies demonstrate that both gefitinib and erlotinib generate ROS in a dose-dependent manner in a process blocked by antioxidant and PAFR antagonist, verifying their pro-oxidative stressor’s ability, and the role of the PAFR in this effect. We observed that these targeted therapies induce MVP release in a dose- and time-dependent manner, similar to a PAFR-agonist, carbamoyl-PAF (CPAF), and PAFR-independent agonist, phorbol myristate acetate (PMA), used as positive controls. To confirm the PAFR dependency, we demonstrate that siRNA-mediated PAFR knockdown or PAFR antagonist significantly blocked only targeted therapies- and CPAF-mediated but not PMA-induced MVP release. The use of pharmacologic inhibitor strategy suggested the involvement of the lipid ceramide-generating enzyme, acid sphingomyelinase (aSMase) in MVP biogenesis, and observed that regardless of the stimuli used, aSMase inhibition significantly blocked MVP release. As mitogen-activated protein kinase (MAPK; ERK1/2 and p38) pathways crosstalk with PAFR, their inhibition also significantly attenuated targeted therapies-mediated MVP release. These findings indicate that PAFR signaling could be targeted to modify cellular responses of targeted therapies in lung cancer cells.


2021 ◽  
Author(s):  
Daniela Di Paolo ◽  
Francesca Pontis ◽  
Massimo Moro ◽  
Giovanni Centonze ◽  
Giulia Bertolini ◽  
...  

2021 ◽  
Author(s):  
Jie Zhang ◽  
Yuanyuan Zeng ◽  
Yueping Xing ◽  
Xiangrong Li ◽  
Lingqin Zhou ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document