In contrast to numerous previous reports that focused on the roles of IDH1 mutations in tumorigenesis (13,18C21), the present study was, to the best of our knowledge, the first study on the function of wild-type IDH1 in primary GBM cell migration

In contrast to numerous previous reports that focused on the roles of IDH1 mutations in tumorigenesis (13,18C21), the present study was, to the best of our knowledge, the first study on the function of wild-type IDH1 in primary GBM cell migration. of phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) pathway-regulated genes, including Myc, Bisoprolol Snail family transcriptional repressor 2 and Twist-related protein 1, which are primarily cell migration regulatory factors. Western blotting revealed that the overexpression or knockdown of IDH1 promoted or inhibited the PI3K/AKT/mTOR pathway, respectively. -KG treatment of primary GBM cells also promoted the PI3K/AKT/mTOR pathway. Furthermore, IDH1-overexpressing and -KG-treated U87 cells were incubated with rapamycin, an mTOR-specific inhibitor, and the results revealed that rapamycin treatment reversed the increased cell migration caused by IDH1 overexpression and -KG treatment. The results indicated that IDH1 regulated the migration of primary GBM cells by altering -KG levels and that the Bisoprolol function of the IDH1/-KG axis may rely on PI3K/AKT/mTOR pathway regulation. (15) reported that IDH1 suppression via shRNA or specific inhibitors inhibited primary GBM growth and facilitated cellular differentiation. However, the role of IDH1 in primary GBM cell migration remains elusive. Considering that primary GBM is a type of cancer that exhibits relatively high migratory abilities (27,28), excess IDH1 was hypothesized to contribute to primary GBM migration. The current Bisoprolol study discovered that IDH1 knockdown or overexpression led to repressed or improved cell migration, respectively. -KG is primarily produced by IDH1 via oxidative decarboxylation (38,39). Therefore, whether -KG mediated the effect of wild-type IDH1 on primary GBM cell migration was investigated. Cellular -KG levels were positively associated with changes in IDH1 levels. By treating U87 cells with different concentrations of -KG, dose-dependent increases in the migration rates of primary GBM cells were observed. However, to the best of our knowledge, there is no applicable method to directly reduce -KG levels in live cells, which impeded the current study to examine the effect of decreased -KG on cell migration. The most common way to reduce -KG levels in live cells is to repress enzymes that catalyze the production of -KG, such as IDH1. Therefore, the current study investigated the effect of IDH1 knockdown and the results matched expectations. Thus, the changes in the migration of primary GBM cells mediated by changes in IDH1 levels may occur by altering -KG levels. The PI3K/AKT/mTOR pathway regulates multiple cellular events, including growth, proliferation, motility and survival, which are often dysregulated in cancer (40). Although numerous previous studies (41C43) have reported the effect of the IDH1 R132H mutation on the PI3K/AKT/mTOR pathway, the correlation remains unclear. While IDH1 R132H and D-2HG were discovered to inhibit the PI3K/AKT/mTOR pathway in human glioma samples (41), another previous study reported that IDH1 R132H and 2-HG promoted the PI3K/AKT/mTOR pathway, resulting in upregulated glioma migration (44,45). Despite previous reports on mutated IDH1, the function of wild-type IDH1 and -KG levels on the PI3K/AKT/mTOR pathway remain unclear. The current study revealed that PI3K/AKT/mTOR pathway activity was enhanced by IDH1 overexpression and -KG treatment, and repressed by IDH1 knockdown. To further investigate whether the IDH1/-KG axis regulated primary GBM cell migration by modulating the PI3K/AKT/mTOR pathway, rapamycin treatment combined with IDH1 overexpression or -KG supplementation was employed. The results demonstrated that the increased cell migration of primary GBM cells was reversed, indicating that the IDH1/-KG axis regulated cell migration in primary GBM cells via the PI3K/AKT/mTOR pathway. Despite the mechanism revealed in the present study, there are areas of research that require further study. Firstly, as the results of the current study demonstrated that IDH1 may be a potential therapeutic target or diagnostic marker in primary GBM, further investigations are required prior to clinical application. Secondly, although -KG levels were reported to Rabbit Polyclonal to PGD regulate the PI3K/AKT/mTOR pathway in primary GBM cells, the detailed mechanism of.