Your browser doesn't support javascript.
loading
Platelet-Activating Factor-Receptor Signaling Mediates Targeted Therapies-Induced Microvesicle Particles Release in Lung Cancer Cells.
Chauhan, Shreepa J; Thyagarajan, Anita; Chen, Yanfang; Travers, Jeffrey B; Sahu, Ravi P.
Afiliação
  • Chauhan SJ; Department of Pharmacology and Toxicology, Boonshoft School of Medicine Wright State University, Dayton, OH 45345, USA.
  • Thyagarajan A; Department of Pharmacology and Toxicology, Boonshoft School of Medicine Wright State University, Dayton, OH 45345, USA.
  • Chen Y; Department of Pharmacology and Toxicology, Boonshoft School of Medicine Wright State University, Dayton, OH 45345, USA.
  • Travers JB; Department of Pharmacology and Toxicology, Boonshoft School of Medicine Wright State University, Dayton, OH 45345, USA.
  • Sahu RP; Department of Dermatology, Wright State Physicians, Wright State University, Dayton, OH 45345, USA.
Int J Mol Sci ; 21(22)2020 Nov 12.
Article em En | MEDLINE | ID: mdl-33198218
ABSTRACT
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.
Assuntos
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Glicoproteínas da Membrana de Plaquetas / Transdução de Sinais / Receptores Acoplados a Proteínas G / Microvasos / Micropartículas Derivadas de Células / Neoplasias Pulmonares Idioma: En Ano de publicação: 2020 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Glicoproteínas da Membrana de Plaquetas / Transdução de Sinais / Receptores Acoplados a Proteínas G / Microvasos / Micropartículas Derivadas de Células / Neoplasias Pulmonares Idioma: En Ano de publicação: 2020 Tipo de documento: Article