3)

3). the timing of the measurement following radiation. In their study, the sphingolipid analysis was performed within hours following radiation; on the other hand, in our study, the analysis was performed once survived cells grew to confluence, a process that required approximately one month. Following radiation, most cells died within one week; 1% of cells survived ionizing radiation and grew to confluence after a month of tradition. The data suggest that only cells that express a high level of ASAH1 could survive radiation. Nr4a1 Since Mahdy performed sphingolipid analysis within hours following radiation, their results likely included cells that would not survive radiation long-term (those that contained lower levels of ASAH1 and higher levels of ceramides). With our study, the longer time interval selected out these cells (as they died within 1 week), where final analysis involved only cells that survived radiation. To confirm the upregulation of ASAH1 and Sph-1P like a mechanism Acetylcysteine of radioresistance, we performed Acetylcysteine western blotting and IHC on GBM cell lines and individual GBM cells. IHC staining of both U87 and U87-10gy cells with humanized anti-Sph-1P exposed increased levels of Sph-1P in irradiated U87-10gy (Fig. 3). Similar to the western blot data, ASAH1 IHC analysis of four different Acetylcysteine units of data from your same patient (pre- and post-radiation GBM Acetylcysteine specimens) confirmed the upregulation of ASAH1 in post-radiation samples, ranging from 1.5- to 60-fold higher in staining intensity as assessed by ImageJ (Fig. 4). This getting was further supported by data showing a significantly lower Allred median ASAH1 staining score for non-irradiated GBMs in comparison to radiated GBM samples (Fig. 8). Consistent with earlier data (13), we showed that irradiated GBMs also have a Acetylcysteine higher protein expression of CD133 than non-irradiated GBMs. Given the concomitant high expression level of ASAH1 in irradiated GBMs, this raises the possibility that CD133+ cells or CSCs are the cells that survive radiation and overexpress ASAH1, as shown in western blot and IHC studies (Fig. 4A). These results indicate that this U87-10gy cell collection is usually a potential, clinically-relevant model to study recurrent GBMs, especially in studies that target the sphingolipid metabolism pathway. ASAH1 was shown in this study to be secreted into the extracellular space (Figs. 4, ?,5,5, and ?and8),8), which is consistent with other reports that document secretion of Sph-1P into the extracellular space as well (21,26,28,29). Consequently, cancer cells with increased secretion of ASAH1 and Sph-1P produce a tumor microenvironment that favors cancer survival by virtue of the ASAH1 and Sph-1P known tumor-promoting functions (21,27,29C31). In support of this microenvironment theory, we exhibited that media from SJGBM2-10gy cells, which secreted a high amount of ASAH1, promoted 50% more cell growth than media from SJGBM2 cells that contained a lower amount of secreted ASAH1 (Fig. 5). In addition, staining of irradiated GBMs also exhibited significant ASAH1 staining in the extracellular space, suggesting that irradiated GBMs also secrete ASAH1 into the extracellular space (Figs. 4 and ?and8).8). The presence of tumor promoters ASAH1 and Sph-1P outside the intracellular space provides a unique opportunity to target these molecules with antibodies. Employing this strategy, we found that treatment of U87-10gy cells with anti-ASAH1 antibody reduced cell growth by 50% (Fig. 6). A similar 50% reduction in cell growth was observed in U87-10gy treated with the humanized anti-Sph-1P antibody (Fig. 6). This reduction in cell growth is likely attributed to the ability of antibodies to disrupt the functions that ASAH1 and Sph-1 have in the promotion of cell growth and survival (3,21,28,29,31C33). The benefit of an anti-ASAH1 antibody was.