Remarkably, our recent studies revealed that EPCR could function as a negative regulator of cancer progression in malignant pleural mesothelioma (MPM)21

Remarkably, our recent studies revealed that EPCR could function as a negative regulator of cancer progression in malignant pleural mesothelioma (MPM)21. founded MPM originating from MPM cells lacking EPCR reduced the progression of tumor growth. Ad.EPCR treatment elicited recruitment of macrophages and NK cells into the tumor microenvironment and increased IFN and TNF levels in the pleural space. Ad.EPCR treatment resulted in a marked increase in tumor cell apoptosis. In summary, our data display that EPCR manifestation in MPM cells promotes tumor cell apoptosis, and intrapleural EPCR gene therapy suppresses MPM progression. Endothelial cell protein C receptor (EPCR) was first recognized and isolated like a cellular receptor for protein C on endothelial cells1. EPCR takes on a crucial part in the protein C anticoagulant pathway by advertising protein C activation2. EPCR also serves as the cellular receptor for triggered protein C (APC) and helps APC-mediated vascular protecting signaling via activation of protease-activated receptors. (PARs)3,4. Although originally identified as an endothelial cell receptor, EPCR offers PK 44 phosphate since been recognized in a variety of cell types5, including hematopoietic, epithelial progenitor cells, and malignancy cells6,7,8,9. Recent studies discovered novel ligands for EPCR4, such as erythrocyte membrane protein 110, and a specific variant of the T-cell receptor11. These observations have opened unsuspected fresh tasks for EPCR beyond hemostasis4. EPCR-mediated cell signaling, in general, was shown to contribute to cell survival and anti-apoptotic pathways3,4,12. EPCR-APC-induced cell signaling was shown to inhibit apoptosis in endothelial cells, malignancy cells, and additional cell types13,14,15,16,17. The EPCR-APC axis advertised the survival of lung adenocarcinoma cells by avoiding their apoptosis18. EPCR expressing breast tumor stem cells were shown to have improved tumor cell-initiating activity compared to cells lacking EPCR19. EPCR overexpression in breast cancer cells improved the tumor growth potential at an initial stage20. Remarkably, our recent studies exposed that EPCR could function as a negative regulator of malignancy progression in malignant pleural mesothelioma (MPM)21. These studies showed that transduction of EPCR gene manifestation in aggressive REN MPM cells that communicate oncogenic tissue element (TF) but lack EPCR markedly attenuated the tumorigenicity of REN MPM cells21. Confirming the tumor suppressive effect of EPCR in MPM, the knock-down of EPCR in non-aggressive TF expressing MPM cells that constitutively communicate EPCR improved the tumorigenicity of the non-aggressive MPM cells21. This study also exposed that EPCR in MPM cells promotes tumor cell apoptosis and studies performed here display that EPCR manifestation, in itself, does not promote apoptosis in MPM cells. However, EPCR manifestation in MPM cells makes them highly susceptible to TNF?+?IFN-induced cell death. It is unlikely that EPCR-APC or EPCR-FVIIa-mediated cell signaling is responsible for advertising TNF?+?IFN-induced cell death of MPM cells since no APC or FVIIa was added in our experimental treatment. Furthermore, treatment of cells with EPCR obstructing antibody that prevents APC and FVIIa binding to EPCR did not block the EPCR-mediated apoptosis (data not demonstrated). Furthermore, all published literature using several other cell types showed that EPCR-APC-mediated cell signaling activates antiapoptotic and not proapoptotic pathways3,4,49. Consistent with this, we also found that addition of APC to MPM cells expressing EPCR reduced MPM cell apoptosis (data not demonstrated). The proapoptotic function of EPCR appears to be limited to MPM cells once we found no significant variations in apoptosis in MDA231 breast cancer cells lacking noticeable EPCR levels and MDA 231 cells transduced to overexpress EPCR (data PK 44 phosphate not demonstrated). Genome-wide manifestation profiling of mRNA in REN cells, REN cells transfected to express EPCR, MS-1, and M9K cells that constitutively communicate EPCR showed that EPCR manifestation alters the transcription profile in MPM cells. A most striking alteration is in the manifestation of malignancy/testis (CT) antigens (GAGEs, XAGE 2B, MAGE, and CT45A4). Manifestation of these genes was markedly reduced, 50 to 100-fold, in MPM cells expressing PK 44 phosphate EPCR (REN(+EPCR), MS-1, M9K) in comparison to REN MPM cells lacking EPCR. These data were confirmed in qRT-PCR (data not demonstrated). In normal health, CT antigen manifestation is definitely purely restricted to the testes, but they are aberrantly indicated in various cancers50, including mesothelioma51. Recent studies suggest that CT antigens contribute to the pathogenesis of malignancy by suppressing apoptosis and advertising cell survival52,53,54. GAGE was shown to render tumor cells resistant to apoptosis mediated by IFN-, Fas, taxol, and -irradiation55. Therefore, it is possible that EPCR-mediated down-regulation Bmp6 of GAGE and additional CT antigens in MPM cells makes EPCR expressing MPM cells highly PK 44 phosphate susceptible to TNF?+?IFN-induced apoptosis. A well-designed.

Scroll to top