@article{PeixotoJanakiRamanSchlickeretal.2021, author = {Peixoto, Joana and Janaki-Raman, Sudha and Schlicker, Lisa and Schmitz, Werner and Walz, Susanne and Winkelkotte, Alina M. and Herold-Mende, Christel and Soares, Paula and Schulze, Almut and Lima, Jorge}, title = {Integrated metabolomics and transcriptomics analysis of monolayer and neurospheres from established glioblastoma cell lines}, series = {Cancers}, volume = {13}, journal = {Cancers}, number = {6}, issn = {2072-6694}, doi = {10.3390/cancers13061327}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-234110}, year = {2021}, abstract = {Altered metabolic processes contribute to carcinogenesis by modulating proliferation, survival and differentiation. Tumours are composed of different cell populations, with cancer stem-like cells being one of the most prominent examples. This specific pool of cells is thought to be responsible for cancer growth and recurrence and plays a particularly relevant role in glioblastoma (GBM), the most lethal form of primary brain tumours. Here, we have analysed the transcriptome and metabolome of an established GBM cell line (U87) and a patient-derived GBM stem-like cell line (NCH644) exposed to neurosphere or monolayer culture conditions. By integrating transcriptome and metabolome data, we identified key metabolic pathways and gene signatures that are associated with stem-like and differentiated states in GBM cells, and demonstrated that neurospheres and monolayer cells differ substantially in their metabolism and gene regulation. Furthermore, arginine biosynthesis was identified as the most significantly regulated pathway in neurospheres, although individual nodes of this pathway were distinctly regulated in the two cellular systems. Neurosphere conditions, as opposed to monolayer conditions, cause a transcriptomic and metabolic rewiring that may be crucial for the regulation of stem-like features, where arginine biosynthesis may be a key metabolic pathway. Additionally, TCGA data from GBM patients showed significant regulation of specific components of the arginine biosynthesis pathway, providing further evidence for the importance of this metabolic pathway in GBM.}, language = {en} } @article{JungwirthYuMoustafaetal.2019, author = {Jungwirth, Gerhard and Yu, Tao and Moustafa, Mahmoud and Rapp, Carmen and Warta, Rolf and Jungk, Christine and Sahm, Felix and Dettling, Steffen and Zweckberger, Klaus and Lamszus, Katrin and Senft, Christian and Loehr, Mario and Keßler, Almuth F. and Ketter, Ralf and Westphal, Manfred and Debus, Juergen and von Deimling, Andreas and Simon, Matthias and Unterberg, Andreas and Abdollahi, Amir and Herold-Mende, Christel}, title = {Identification of KIF11 as a Novel Target in Meningioma}, series = {Cancers}, volume = {11}, journal = {Cancers}, number = {4}, issn = {2072-6694}, doi = {10.3390/cancers11040545}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-197402}, year = {2019}, abstract = {Kinesins play an important role in many physiological functions including intracellular vesicle transport and mitosis. The emerging role of kinesins in different cancers led us to investigate the expression and functional role of kinesins in meningioma. Therefore, we re-analyzed our previous microarray dataset of benign, atypical, and anaplastic meningiomas (n = 62) and got evidence for differential expression of five kinesins (KIFC1, KIF4A, KIF11, KIF14 and KIF20A). Further validation in an extended study sample (n = 208) revealed a significant upregulation of these genes in WHO°I to °III meningiomas (WHO°I n = 61, WHO°II n = 88, and WHO°III n = 59), which was most pronounced in clinically more aggressive tumors of the same WHO grade. Immunohistochemical staining confirmed a WHO grade-associated upregulated protein expression in meningioma tissues. Furthermore, high mRNA expression levels of KIFC1, KIF11, KIF14 and KIF20A were associated with shorter progression-free survival. On a functional level, knockdown of kinesins in Ben-Men-1 cells and in the newly established anaplastic meningioma cell line NCH93 resulted in a significantly inhibited tumor cell proliferation upon siRNA-mediated downregulation of KIF11 in both cell lines by up to 95\% and 71\%, respectively. Taken together, in this study we were able to identify the prognostic and functional role of several kinesin family members of which KIF11 exhibits the most promising properties as a novel prognostic marker and therapeutic target, which may offer new treatment options for aggressive meningiomas.}, language = {en} } @article{SchulzeHuttererSaboetal.2018, author = {Schulze, Markus and Hutterer, Maria and Sabo, Anja and Hoja, Sabine and Lorenz, Julia and Rothhammer-Hampl, Tanja and Herold-Mende, Christel and Floßbach, Lucia and Monoranu, Camelia and Riemenschneider, Markus J.}, title = {Chronophin regulates active vitamin B6 levels and transcriptomic features of glioblastoma cell lines cultured under non-adherent, serum-free conditions}, series = {BMC Cancer}, volume = {18}, journal = {BMC Cancer}, doi = {10.1186/s12885-018-4440-4}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-234645}, year = {2018}, abstract = {Background The phosphatase chronophin (CIN/PDXP) has been shown to be an important regulator of glioma cell migration and invasion. It has two known substrates: p-Ser3-cofilin, the phosphorylated form of the actin binding protein cofilin, and pyridoxal 5′-phosphate, the active form of vitamin B6. Phosphoregulation of cofilin, among other functions, plays an important role in cell migration, whereas active vitamin B6 is a cofactor for more than one hundred enzymatic reactions. The role of CIN has yet only been examined in glioblastoma cell line models derived under serum culture conditions. Results We found that CIN is highly expressed in cells cultured under non-adherent, serum-free conditions that are thought to better mimic the in vivo situation. Furthermore, the substrates of CIN, p-Ser3-cofilin and active vitamin B6, were significantly reduced as compared to cell lines cultured in serum-containing medium. To further examine its molecular role we stably knocked down the CIN protein with two different shRNA hairpins in the glioblastoma cell lines NCH421k and NCH644. Both cell lines did not show any significant alterations in proliferation but expression of differentiation markers (such as GFAP or TUBB3) was increased in the knockdown cell lines. In addition, colony formation was significantly impaired in NCH644. Of note, in both cell lines CIN knockdown increased active vitamin B6 levels with vitamin B6 being known to be important for S-adenosylmethionine biosynthesis. Nevertheless, global histone and DNA methylation remained unaltered as was chemoresistance towards temozolomide. To further elucidate the role of phosphocofilin in glioblastoma cells we applied inhibitors for ROCK1/2 and LIMK1/2 to our model. LIMK- and ROCK-inhibitor treatment alone was not toxic for glioblastoma cells. However, it had profound, but antagonistic effects in NCH421k and NCH644 under chemotherapy. Conclusion In non-adherent glioblastoma cell lines cultured in serum-free medium, chronophin knockdown induces phenotypic changes, e.g. in colony formation and transcription, but these are highly dependent on the cellular background. The same is true for phenotypes observed after treatment with inhibitors for kinases regulating cofilin phosphorylation (ROCKs and LIMKs). Targeting the cofilin phosphorylation pathway might therefore not be a straightforward therapeutic option in glioblastoma.}, language = {en} }