Share this post on:

To their volume at randomization. Each group contained six mice. *p = 0.0009. (C) U251 cells were injected s.c. in a mouse flank model. When tumors reached ,500 mm3 in size, mice were exposed to either PBS alone or EGF-SubA (125 mg/kg). Mice were then sacrificed 24 h after treatment and stated tissue 1326631 was dissected, flash frozen, and tissue lysates were generated to evaluate for GRP78 cleavage by immunoblot. doi:10.1371/journal.pone.0052265.gStatisticsThe statistical analysis was done for the described treatment conditions using a Student’s t test. A probability level of a P value of ,0.05 was considered significant.Results and DiscussionPrevious investigations have demonstrated aberrant expression of GRP78 in malignant glioma at the transcriptional level [10,11], although its expression at the protein level has yet to be comprehensively quantified. We therefore performed immunohistochemical staining on a glioma tissue microarray (TMA). The TMA consisted of a total of 56 glioma specimens, 11 samples were Grade II and 45 samples were Grade III/IV; individual histologies are provided in Fig. S1. The expression level of GRP78 ranged from no expression (0) to diffuse expression (3+), with representative images provided in Fig. 1A. Although various levels of focal GRP78 expression was present in the different grades of glioma, diffuse expression (3+) was only present in Grade III and IV tumors (n = 21/45), further P7C3 custom synthesis supporting the biologic relevance the UPR plays in malignant glioma and its potential to serve as a molecular target. We have previously demonstrated selective cleavage of GRP78 in EGFR-positive prostate and breast cancer cells exposed to EGF-SubA; thereby confirming the receptor-binding activity of the EGF moiety and the proteolytic activity of the SubA moiety [16]. We now extend these studies to explore the potential of both the fusion protein EGF-SubA and the SubA toxin alone to cleave GRP78 in glioblastoma models. As demonstrated in Fig. 2A, EGFSubA demonstrated potent proteolytic activity, cleaving GRP78 atconcentrations ranging from 0.5 to 2.5 pM in established glioblastoma cell lines (U251 and T98G) and the glioblastoma neural stem (GNS) cell line G179. These concentrations were over 20 fold lower when compared to the SubA toxin alone, which required approximately 50 pM to induce GRP78 cleavage, confirming increased 58-49-1 chemical information potency of the fusion protein EGF-SubA. Time course studies demonstrated maximal cleavage of GRP78 within 16 h of EGF-SubA exposure (Fig. 2B). Conversely, cleavage of GRP78 in normal human astrocytes (NHA) required significantly higher concentrations of EGF-SubA when compared to the glioblastoma cell lines, supporting the tumor specificity of this approach. Interestingly, the glioblastoma cell line U87 required considerably higher concentrations of EGF-SubA and SubA toxin to induce GRP78 cleavage. As an initial investigation, based on the mechanism of action of EGF-SubA, we performed western blot analysis to determine if the relative expression of EGFR or GRP78 could contribute to the observed differential response of EGF-SubA. As demonstrated in Fig. S2, expression levels of these proteins did not appear to be significantly different between the cancer cell lines tested. As we reported earlier, EGF-SubA induced 12926553 toxicity is EGFR-dependent, but does not directly correspond to the EGFR expression level, reflecting a more complex cell-specific process of EGFR-mediated internalization and trafficking, as well as the m.To their volume at randomization. Each group contained six mice. *p = 0.0009. (C) U251 cells were injected s.c. in a mouse flank model. When tumors reached ,500 mm3 in size, mice were exposed to either PBS alone or EGF-SubA (125 mg/kg). Mice were then sacrificed 24 h after treatment and stated tissue 1326631 was dissected, flash frozen, and tissue lysates were generated to evaluate for GRP78 cleavage by immunoblot. doi:10.1371/journal.pone.0052265.gStatisticsThe statistical analysis was done for the described treatment conditions using a Student’s t test. A probability level of a P value of ,0.05 was considered significant.Results and DiscussionPrevious investigations have demonstrated aberrant expression of GRP78 in malignant glioma at the transcriptional level [10,11], although its expression at the protein level has yet to be comprehensively quantified. We therefore performed immunohistochemical staining on a glioma tissue microarray (TMA). The TMA consisted of a total of 56 glioma specimens, 11 samples were Grade II and 45 samples were Grade III/IV; individual histologies are provided in Fig. S1. The expression level of GRP78 ranged from no expression (0) to diffuse expression (3+), with representative images provided in Fig. 1A. Although various levels of focal GRP78 expression was present in the different grades of glioma, diffuse expression (3+) was only present in Grade III and IV tumors (n = 21/45), further supporting the biologic relevance the UPR plays in malignant glioma and its potential to serve as a molecular target. We have previously demonstrated selective cleavage of GRP78 in EGFR-positive prostate and breast cancer cells exposed to EGF-SubA; thereby confirming the receptor-binding activity of the EGF moiety and the proteolytic activity of the SubA moiety [16]. We now extend these studies to explore the potential of both the fusion protein EGF-SubA and the SubA toxin alone to cleave GRP78 in glioblastoma models. As demonstrated in Fig. 2A, EGFSubA demonstrated potent proteolytic activity, cleaving GRP78 atconcentrations ranging from 0.5 to 2.5 pM in established glioblastoma cell lines (U251 and T98G) and the glioblastoma neural stem (GNS) cell line G179. These concentrations were over 20 fold lower when compared to the SubA toxin alone, which required approximately 50 pM to induce GRP78 cleavage, confirming increased potency of the fusion protein EGF-SubA. Time course studies demonstrated maximal cleavage of GRP78 within 16 h of EGF-SubA exposure (Fig. 2B). Conversely, cleavage of GRP78 in normal human astrocytes (NHA) required significantly higher concentrations of EGF-SubA when compared to the glioblastoma cell lines, supporting the tumor specificity of this approach. Interestingly, the glioblastoma cell line U87 required considerably higher concentrations of EGF-SubA and SubA toxin to induce GRP78 cleavage. As an initial investigation, based on the mechanism of action of EGF-SubA, we performed western blot analysis to determine if the relative expression of EGFR or GRP78 could contribute to the observed differential response of EGF-SubA. As demonstrated in Fig. S2, expression levels of these proteins did not appear to be significantly different between the cancer cell lines tested. As we reported earlier, EGF-SubA induced 12926553 toxicity is EGFR-dependent, but does not directly correspond to the EGFR expression level, reflecting a more complex cell-specific process of EGFR-mediated internalization and trafficking, as well as the m.

Share this post on: