Journal articles on the topic 'Glioma Cell Lines'

To see the other types of publications on this topic, follow the link: Glioma Cell Lines.

Create a spot-on reference in APA, MLA, Chicago, Harvard, and other styles

Select a source type:

Consult the top 50 journal articles for your research on the topic 'Glioma Cell Lines.'

Next to every source in the list of references, there is an 'Add to bibliography' button. Press on it, and we will generate automatically the bibliographic reference to the chosen work in the citation style you need: APA, MLA, Harvard, Chicago, Vancouver, etc.

You can also download the full text of the academic publication as pdf and read online its abstract whenever available in the metadata.

Browse journal articles on a wide variety of disciplines and organise your bibliography correctly.

1

Chen, Yi-Hsuan, Dueng-Yuan Hueng, and Wen-Chiuan Tsai. "Proteolipid Protein 2 Overexpression Indicates Aggressive Tumor Behavior and Adverse Prognosis in Human Gliomas." International Journal of Molecular Sciences 19, no. 11 (October 26, 2018): 3353. http://dx.doi.org/10.3390/ijms19113353.

Full text
Abstract:
Proteolipid protein 2 (PLP2), a membrane protein of the endoplasmic reticulum, is related to tumor proliferation and metastasis in some human cancers, but not in gliomas. First, we performed western-blot analysis, real-time quantitative PCR and immunohistochemical stains to detect PLP2 expression in 4 glioma cell lines and human glioma tissues. In addition, we used small interfering RNA (SiPLP2) and short hairpin RNA (shPLP2) to knockdown PLP2 expression in GBM8401 and LN229 glioma cell lines. After then, the alteration of PLP2 suppressed glioma cells behavior were examined by cell proliferation, wound healing, cell invasion, and colonies formation assays. Finally, the possible mechanism of PLP2 was analyzed by detecting the expression of the proteins related to cell-cycle checkpoints, cell-proliferative signaling factors, and cell-matrix interaction. Compared with normal brain cell lysates and mRNA, all glioma cell lines displayed PLP2 protein and mRNA overexpression. Besides, higher PLP2 IHC staining significantly correlated with more advanced tumor grades and poorer prognosis in human gliomas. Both siPLP2 transfected gliomas showed a clear inhibition of glioma cell proliferation, migration, and invasion as well as down-regulating p-p38, p-ERK, MMP-2, and MMP-9 expression. In conclusion, we successfully demonstrated that PLP2 overexpression played an oncogenic role in glioma development and aggressive tumor behavior.
APA, Harvard, Vancouver, ISO, and other styles
2

Tuma, Rabiya. "Repopulating glioma cell lines." Journal of Cell Biology 164, no. 3 (January 26, 2004): 334–35. http://dx.doi.org/10.1083/jcb1643rr2.

Full text
APA, Harvard, Vancouver, ISO, and other styles
3

Shi, Fei, Jie Hu, Ping Zheng, Yisong Lv, Hongyu Liu, Guiyun Zhang, and Hongyu Jiang. "LncRNA PANTR1 is Associated with Poor Prognostic and Suppresses Apoptosis in Glioma." Journal of Oncology 2023 (February 20, 2023): 1–14. http://dx.doi.org/10.1155/2023/8537036.

Full text
Abstract:
Glioma is the most common tumor in the central nervous system. High-grade gliomas confer a poor prognosis, being a serious health and economic burden. Current literature suggests the important role of long noncoding RNA (lncRNA) in mammals, especially in tumorigenesis of various tumors. The functions of lncRNA POU3F3 adjacent noncoding transcript 1 (PANTR1) have been investigated in hepatocellular carcinoma but remain yet unclear in gliomas. We evaluated the role of PANTR1 in glioma cells using published data from The Cancer Genome Atlas (TCGA), then validated it by ex vivo experiments. To investigate the potential cellular mechanism of different levels of PANTR1 expression in glioma cells, we used siRNA-mediated knockdown in low-grade (grade II) cell lines and GBM (grade IV) cell lines (SW1088 and SHG44, respectively). On the molecular level, low expression of PANTR1 caused significantly reduced glioma cell viability and enhanced cell death. Moreover, we identified the importance of PANTR1 expression for cell migration in both cell lines, a critical foundation for invasiveness in recurrent gliomas. In conclusion, this study provides the first evidence that PANTR1 has a relevant role in human glioma by influencing cell viability and cell death.
APA, Harvard, Vancouver, ISO, and other styles
4

Bota, Daniela A., Daniela Alexandru, Stephen T. Keir, Darell Bigner, James Vredenburgh, and Henry S. Friedman. "Proteasome inhibition with bortezomib induces cell death in GBM stem-like cells and temozolomide-resistant glioma cell lines, but stimulates GBM stem-like cells' VEGF production and angiogenesis." Journal of Neurosurgery 119, no. 6 (December 2013): 1415–23. http://dx.doi.org/10.3171/2013.7.jns1323.

Full text
Abstract:
Object Recurrent malignant gliomas have inherent resistance to traditional chemotherapy. Novel therapies target specific molecular mechanisms involved in abnormal signaling and resistance to apoptosis. The proteasome is a key regulator of multiple cellular functions, and its inhibition in malignant astrocytic lines causes cell growth arrest and apoptotic cell death. The proteasome inhibitor bortezomib was reported to have very good in vitro activity against malignant glioma cell lines, with modest activity in animal models as well as in clinical trials as a single agent. In this paper, the authors describe the multiple effects of bortezomib in both in vitro and in vivo glioma models and offer a novel explanation for its seeming lack of activity. Methods Glioma stem-like cells (GSCs) were obtained from resected glioblastomas (GBMs) at surgery and expanded in culture. Stable glioma cell lines (U21 and D54) as well as temozolomide (TMZ)-resistant glioma cells derived from U251 and D54-MG were also cultured. GSCs from 2 different tumors, as well as D54 and U251 cells, were treated with bortezomib, and the effect of the drug was measured using an XTT cell viability assay. The activity of bortezomib was then determined in D54-MG and/or U251 cells using apoptosis analysis as well as caspase-3 activity and proteasome activity measurements. Human glioma xenograft models were created in nude mice by subcutaneous injection. Bevacizumab was administered via intraperitoneal injection at a dose of 5 mg/kg daily. Bortezomib was administered by intraperitoneal injection 1 hour after bevacizumab administration in doses of at a dose of 0.35 mg/kg on days 1, 4, 8, and 11 every 21 days. Tumors were measured twice weekly. Results Bortezomib induced caspase-3 activation and apoptotic cell death in stable glioma cell lines and in glioma stem-like cells (GSCs) derived from malignant tumor specimens Furthermore, TMZ-resistant glioma cell lines retained susceptibility to the proteasome inhibition. The bortezomib activity was directly proportional with the cells' baseline proteasome activity. The proteasome inhibition stimulated both hypoxia-inducible factor (HIF)–1α and vascular endothelial growth factor (VEGF) production in malignant GSCs. As such, the VEGF produced by GSCs stimulated endothelial cell growth, an effect that could be prevented by the addition of bevacizumab (VEGF antibody) to the media. Similarly, administration of bortezomib and bevacizumab to athymic mice carrying subcutaneous malignant glioma xenografts resulted in greater tumor inhibition and greater improvement in survival than administration of either drug alone. These data indicate that simultaneous proteasome inhibition and VEGF blockade offer increased benefit as a strategy for malignant glioma therapy. Conclusions The results of this study indicate that combination therapies based on bortezomib and bevacizumab might offer an increased benefit when the two agents are used in combination. These drugs have a complementary mechanism of action and therefore can be used together to treat TMZ-resistant malignant gliomas.
APA, Harvard, Vancouver, ISO, and other styles
5

Han, Jizhong, Yu Xiong, Huajiang Deng, Jie Zhou, Lilei Peng, Wei Xiang, Yang Ming, and Ligang Chen. "MiR-455-3p regulates glioma cell proliferation by targeting PAX6." Tropical Journal of Pharmaceutical Research 18, no. 4 (May 17, 2021): 689–95. http://dx.doi.org/10.4314/tjpr.v18i4.2.

Full text
Abstract:
Purpose: To investigate the role of miR-455-3p in gliomas. Method: Quantitative real-time polymerase chain reaction was used to measure miR-455-3p and paired box 6 (PAX6) levels in glioma cell lines. Western blot analysis was used to determine the expression of cell cycle regulators. In addition to over-expression, silencing of miR-455-3p or PAX6 was performed to study the functions of miR-455-3p in gliomas. Results: The levels of miR-455-3p were significantly up-regulated in glioma cell lines (p < 0.05), while miR-455-3p over-expression increased glioma cell proliferation and interfered with the progress of the cell cycle (p < 0.01). Furthermore, endogenous miR-455-3p silencing prevented glioma cell proliferation by regulating cell cycle progression (p < 0.05).The results also showed that PAX6 controlled the cell cycle while PAX6 silencing selectively regulated p21 expression (p < 0.01). Furthermore, miR-455-3p and PAX6 influenced p53 expression. Re-introduction of PAX6 expressing vector into glioma cells rescued the pro-tumoral effect of miR-455-3p overexpression. Conclusion: These findings demonstrate the role of miR-455-3p as a tumour oncogene in gliomas via regulation of the cell cycle, indicating that miR-455-3p might act as a new treatment strategy for glioma cell tumours and a predictor of survival in glioma patients.
APA, Harvard, Vancouver, ISO, and other styles
6

Matsumoto, Tsuyoshi, Eiichi Tani, Keizo Kaba, Hideki Shindo, and Katsuya Miyaji. "Expression of P-glycoprotein in human glioma cell lines and surgical glioma specimens." Journal of Neurosurgery 74, no. 3 (March 1991): 460–66. http://dx.doi.org/10.3171/jns.1991.74.3.0460.

Full text
Abstract:
✓ The expression of P-glycoprotein, a product of multidrug resistance gene 1, was studied by Western blotting and immunohistochemistry in five human glioma cell lines. One glioma cell line was resistant to vincristine, Adriamycin (doxorubicin), and etoposide, and the other four glioma cell lines were sensitive to each drug. The multidrug-resistant cell line showed a high expression of P-glycoprotein in Western blot analysis and a positive immunostaining for P-glycoprotein mainly along the cell membrane, whereas all multidrug-sensitive glioma cell lines demonstrated no expression of P-glycoprotein in Western blotting and no immunostaining for P-glycoprotein, thus showing a good correlation between the expression level of P-glycoprotein and the extent of multidrug resistance. In 18 human surgical glioma specimens, there was no evidence of complete absence of immunostaining for P-glycoprotein. With a definition of more than 20% of P-glycoprotein-positive cells as positive, from 10% to 20% as intermediate, and less than 10% as negative, immunostaining for P-glycoprotein was positive in one specimen and intermediate in six of 15 specimens taken from virgin gliomas, and positive in two specimens and intermediate in one of three recurrent gliomas treated previously with irradiation, ACNU (1-(4-amino-2-methyl-pyrimidine-5-yl)-methyl-3-(2-chloroethyl)-3-nitrosourea hydrochloride), cisplatin, vincristine, and/or procarbazine.
APA, Harvard, Vancouver, ISO, and other styles
7

Nakano, Atsuhisa, Eiichi Tani, Kaoru Miyazaki, Yoshihiro Yamamoto, and Jun-ichi Furuyama. "Matrix metalloproteinases and tissue inhibitors of metalloproteinases in human gliomas." Journal of Neurosurgery 83, no. 2 (August 1995): 298–307. http://dx.doi.org/10.3171/jns.1995.83.2.0298.

Full text
Abstract:
✓ The gene expression of five matrix metalloproteinases (MMPs) and two tissue inhibitors of metalloproteinases (TIMPs) was studied in human gliomas in vivo and in vitro to evaluate their roles in glioma invasion. Simultaneous expression of one to four MMP genes and two TIMP genes was found in 17 surgical glioma specimens, and one MMP (gelatinase A) gene and two TIMP genes were simultaneously expressed in tissue of three brains. The concomitant overexpression of gelatinase A, gelatinase B, and occasional matrilysin genes was associated with the malignancy of gliomas and accompanied by overexpression of the TIMP-1 gene. In five human glioma cell lines, gelatinase A, TIMP-1, and TIMP-2 genes were constitutively expressed in all cell lines: the matrilysin gene in three cell lines; the stromelysin gene in two cell lines; and the interstitial collagenase gene in one cell line. There was a clear difference in the expression of gelatinase B and stromelysin genes between surgical glioma specimens and glioma cell lines: the gelatinase B gene was not expressed constitutively in vitro but was overexpressed in vivo, whereas the stromelysin gene was not expressed in vivo but was expressed in some cell lines. To find the cause of that difference in vivo and in vitro, the transcriptional regulations of MMP and TIMP genes by tumor promoter, growth factors, or cytokines were studied in vitro. Interstitial collagenase, gelatinase B, stromelysin, and TIMP-1 genes were upregulated in many cell lines by phorbol-12-myristate-13-acetate (PMA) and in some cell lines by epidermal growth factor, tumor necrosis factor-α, or interleukin-1β. Transforming growth factor-β1 (TGFβ1) upregulated gelatinase A and matrilysin genes in some cell lines, and there were no clear responses from any MMP and TIMP genes to interleukin-6. Thus, the transcriptional modulation of MMP genes by these growth factors and cytokines seemed insufficient to explain the difference in gelatinase B and stromelysin gene expressionsin vivo and in vitro and was suggestive of the genetic alteration of glioma cells in vitro, the heterogeneous cell population in glioma tissues, or both. Furthermore, the in vitro invasion of glioma cells through Matrigel in response to PMA, TGFβ1, or TIMP-1 was assessed by chemoinvasion assay. In most cell lines, invasion was significantly stimulated by PMA or TGFβ1 but suppressed by TIMP-1. These in vivo and in vitro studies are strongly suggestive of the important roles of some MMPs, especially gelatinase A, gelatinase B, and matrilysin, in the glioma invasion.
APA, Harvard, Vancouver, ISO, and other styles
8

Zhou, You-xin, San-song Chen, Ting-feng Wu, Da-dong Ding, Xiong-hui Chen, Jin-ming Chen, Zuo-peng Su, et al. "A Novel Gene RNF138 Expressed in Human Gliomas and Its Function in the Glioma Cell Line U251." Analytical Cellular Pathology 35, no. 3 (2012): 167–78. http://dx.doi.org/10.1155/2012/519037.

Full text
Abstract:
Background: The gliomas represent the most common primary malignant brain tumors; however, little is known about the molecular pathogenesis of these tumors. Recent research reveals that the oncogenesis and development of gliomas have a close relation to the overexpression of several oncogenes and the inactivation of tumor suppressor genes. Whether the RING finger protein, RNF138, a newly discovered protein, plays a role in glioma oncogenesis is unknown. The present study investigates the expression levels of RNF138 mRNA in glioma samples and noncancerous brain samples and its function in the human glioma cell line U251.Methods: RT-PCR was used to ascertain the expression of RNF138 mRNA in the glioma cell lines U251, SHG44, U87, A172, and U373. The RNF138 mRNA expression levels of 35 pathological confirmed glioma samples (Grade I – 4 cases, Grade II – 13 cases, Grade III – 11 cases, and Grade IV – 7 cases) and five noncancerous brain tissue samples were analyzed by real-time quantitative PCR. By RNA interference (RNAi) with the lentivirus vector system, the expression of RNF138 was inhibited in the human astrocytomas-glioblastoma multiforme cell line U251. The effects of RNF138-knockdown on cell proliferation were assessed by Cellomics, and cell cycle and cell apoptosis were assessed by FACS.Results: The RNF138 mRNA is expressed in the five glioma cell lines, and its expression level is significantly higher in glioma tissue than in noncancerous brain tissue. By down-regulation of RNF138 expression, U251 cell proliferation was inhibited and cell apoptosis increased. At the same time, S stage cells lessened and G2 stage cells increased.Conclusion: The RNF138 gene is highly expressed in glioma tissue and glioma cell lines. It plays an important role in glioma cell proliferation, apoptosis, and cell cycle.
APA, Harvard, Vancouver, ISO, and other styles
9

Alexiou, George A., Xanthi Xourgia, Evrysthenis Vartholomatos, Spyridon Tsiouris, John A. Kalef-Ezra, Andreas D. Fotopoulos, and Athanasios P. Kyritsis. "Comparison of 99mTc-Tetrofosmin and 99mTc-Sestamibi Uptake in Glioma Cell Lines: The Role of P-Glycoprotein Expression." International Journal of Molecular Imaging 2014 (November 10, 2014): 1–5. http://dx.doi.org/10.1155/2014/471032.

Full text
Abstract:
Tc-Tetrofosmin (Tc-TF) and Tc-Sestamibi (Tc-MIBI) are SPECT tracers that have been used for brain tumor imaging. Tumor’s multidrug resistance phenotype, namely, P-glycoprotein (p-gp), and the multidrug resistance related proteins (MRPs) expression have been suggested to influence both tracers’ uptake. In the present study we set out to compare Tc-MIBI uptake in high-grade glioma cell lines and to investigate the influence of gliomas p-gp expression on both tracers’ uptake. We used four glioma cell lines (U251MG, A172, U87MG, and T98G). The expression of p-gp protein was evaluated by flow cytometry. Twenty μCi (7.4·105 Bq) of Tc-TF and Tc-MIBI were used. The radioactivity in the cellular lysate was measured with a dose calibrator. P-gp was significantly expressed only in the U251MG cell line (). In all gliomas cell lines (U251MG, U87MG, A172, and T98G) the Tc-TF uptake was significantly higher than Tc-sestamibi. The U251MG cell line, in which significant p-gp expression was documented, exhibited the strongest uptake difference. Tc-TF uptake was higher than Tc-MIBI in all studied high-grade glioma cell lines. Thus, Tc-TF may be superior to Tc-MIBI for glioma imaging in vivo.
APA, Harvard, Vancouver, ISO, and other styles
10

Jung, Tae-Young, Shin Jung, Hyang-Hwa Ryu, Young-Il Jeong, Yong-Hao Jin, Shu-Guang Jin, In-Young Kim, Sam-Suk Kang, and Hyung-Seok Kim. "Role of galectin-1 in migration and invasion of human glioblastoma multiforme cell lines." Journal of Neurosurgery 109, no. 2 (August 2008): 273–84. http://dx.doi.org/10.3171/jns/2008/109/8/0273.

Full text
Abstract:
Object Galectin-1 is highly expressed in motile cell lines. The authors investigated whether galectin-1 actually modulates the migration and invasion of human glioblastoma multiforme (GBM) cell lines, and whether its expression with respect to invasion and prognosis is attributable to certain glioma subgroups. Methods In the human GBM cell lines U343MG-A, U87MG, and U87MG-10′, the RNA differential display was evaluated using Genefishing technology. The results were validated by reverse transcription polymerase chain reaction and Northern blot analysis to detect possible genetic changes as the determining factors for the motility of the malignant glioma. The migration and invasion abilities were investigated in human GBM cell lines and galectin-1 transfectant using an in vitro brain slice invasion model and a simple scratch technique. The morphological and cytoskeletal (such as the development of actin and vimentin) changes were examined under light and confocal microscopy. Galectin-1 expression was assessed on immunohistochemical tests and Western blot analysis. Results Endogenous galectin-1 expression in the human GBM cell lines was statistically correlated with migratory abilities and invasiveness. The U87-G-AS cells became more round than the U87MG cells and lacked lamellipodia. On immunohistochemical staining, galectin-1 expression was increased in higher-grade glioma subgroups (p = 0.027). Conclusions Diffuse gliomas demonstrated higher expression levels than pilocytic astrocytoma in the Western blot. Galectin-1 appears to modulate migration and invasion in human glioma cell lines and may play a role in tumor progression and invasiveness in human gliomas.
APA, Harvard, Vancouver, ISO, and other styles
11

Miyaji, Katsuya, Eiichi Tani, Hideki Shindo, Atsuhisa Nakano, and Takashi Tokunaga. "Effect of tyrphostin on cell growth and tyrosine kinase activity of epidermal growth factor receptor in human gliomas." Journal of Neurosurgery 81, no. 3 (September 1994): 411–19. http://dx.doi.org/10.3171/jns.1994.81.3.0411.

Full text
Abstract:
✓ The effects of tyrphostin, a selective protein tyrosine kinase inhibitor, on epidermal growth factor (EGF)-stimulated cell growth and EGF-receptor tyrosine kinase activity were studied in four human glioma cell lines. Stimulation by EGF induced variable enhancements of cell growth as well as tyrosine phosphorylation of EGF receptor and intracellular target proteins in all glioma cell lines. The level of immunoreactive EGF receptor detected with antibodies against extra- and intracellular domains was moderate in all four glioma cell lines, but markedly decreased with the latter antibody in two glioma cell lines. This variation was associated with considerable reduction of the EGF-stimulated tyrosine autophosphorylation level. Tyrphostin inhibited dose-dependently the EGF-stimulated cell growth and tyrosine autophosphorylation in all glioma cell lines, and the optimum time for the maximum inhibitory effect on tyrosine autophosphorylation was 12 to 18 hours after treatment with tyrphostin. The antiproliferative activity of tyrphostin nearly correlated quantitatively with its potency as an inhibitor of the EGF-stimulated EGF receptor tyrosine kinase activity. Tyrphostin had no significant effect on the immunoreactive EGF receptor levels, on the affinity constants and numbers of EGF receptor, or on the down-regulation and specific internalization of EGF receptor in any glioma cell line, suggesting that the effects of tyrphostin are not likely to be the results of reduction in EGF receptor and EGF binding capacity. In addition, the serum-stimulated cell growth was also inhibited dose-dependently by higher concentrations of tyrphostin in all glioma cell lines. It might be suggested, therefore, that tyrphostin inhibits EGF-stimulated cell growth by a specific suppression of EGF receptor tyrosine kinase activity, and at higher concentrations there appears to be some degree of either nonspecific inhibition or inhibition of serum-stimulated protein tyrosine kinase activity to induce the cell growth inhibition of gliomas.
APA, Harvard, Vancouver, ISO, and other styles
12

Muldoon, Daniel, Guisheng Zhao, Carly Batt, Mallika Singh, and Theodore Nicolaides. "MODL-17. SHP2 INHIBITORS SHOW ACTIVITY AGAINST NF1-DEFICIENT GLIOMAS AND ENHANCE MAPK PATHWAY INHIBITION IN BRAF-V600E MUTANT GLIOMAS." Neuro-Oncology 22, Supplement_3 (December 1, 2020): iii414. http://dx.doi.org/10.1093/neuonc/noaa222.591.

Full text
Abstract:
Abstract INTRODUCTION Activation of the RAS-MAPK signaling cascade is common in pediatric gliomas. Based on the role of SHP2 in RAS pathway signaling, we hypothesized that NF1-deficient pediatric glioma models would respond to SHP2 inhibitor monotherapy whereas BRAF-V600E gliomas would not. However, we postulated that the latter would exhibit increased sensitivity to a BRAF inhibitor (BRAFi) in combination with SHP2i. Here we demonstrate that the SHP2 inhibitors SHP099 and RMC-4550 (SHP2i) show significant single-agent activity in vitro against NF1-deficient glioma cells and that the combination of RMC-4550 with BRAFi shows increased activity in BRAF-V600E glioma cells relative to the single-agents. METHODS Using a panel of NF1 mutant/deficient and BRAF-V600E mutant glioma cell lines we examined effects on cell viability and protein expression levels of total and phosphorylated MEK, ERK, and AKT. RESULTS LN229 and U87 NF1-deficient glioma cells are sensitive to SHP2i alone but not A375 cells (melanoma, BRAF-V600E). Additionally, we show that in multiple BRAF-V600E glioma cell lines BRAFi sensitivity increases when combined with a SHP2i. Immunoblots show decreased expression of pERK and pMEK in LN229 cells following SHP2i exposure, while A375 cells maintain MAPK pathway signaling. A sustained decrease in the expression of pERK after 24 hours was observed in BRAF-V600E glioma cells with BRAFi in combination with SHP2i, consistent with relief of feedback inhibition. In vivo studies using orthotopic xenograft models are underway. CONCLUSION SHP2i shows preclinical activity in vitro against NF1-deficient pediatric glioma cell lines as a single-agent and against BRAF-V600E gliomas in combination with BRAFi.
APA, Harvard, Vancouver, ISO, and other styles
13

Bowie, Michelle, Seethalakshmi Hariharan, Janell Hostettler, Kristen Roso, Yiping He, Christopher Pirozzi, Martin Roskoski, et al. "IMMU-34. ATRX MUTATIONS PREDICT RESPONSE TO INNATE BASED THERAPY IN GLIOMA." Neuro-Oncology 21, Supplement_6 (November 2019): vi126. http://dx.doi.org/10.1093/neuonc/noz175.526.

Full text
Abstract:
Abstract BACKGROUND Innate based immunotherapies are becoming increasingly important for treating brain tumor patients. Gliomas carry recurrent mutations in regulatory genes that control innate immune signaling responses. About 71% of adult WHO grade II and III gliomas and 57% of secondary glioblastomas also carry a loss-of-function mutation in the ATRX gene. ATRX is a SWI-SNF chromatin remodeling protein that has major roles in processes such as cell cycle regulation and maintenance of genomic stability. Recent studies have implicated ATRX in dysfunctional innate immune signaling in cancer cells. However, the role of ATRX in mediating innate immune responses has not been investigated in gliomas. METHODS AND RESULTS Human and mouse glioma cell lines from a variety of genetic contexts have been examined including models which carry IDH/ATRX mutations, IDH 1p-/19q- and ATRX -/- status. Additionally, using Crispr-Cas9 technology and cloning cell lines with ATRX deletions, we have derived a series of immune competent and nude mice models. Treating these cell lines with double-stranded RNA based innate stimuli led to an enhanced early induction in phospho-interferon regulatory factor 3 (IRF3) and late induction in phospho-STAT1 in the ATRX knockout (KO) cell lines. A differential increase in interferon-stimulated gene 15 (ISG15) release was also noted in the ATRX KO cell lines, further suggesting that ATRX deletion may enable a potent activation of type I interferon production. A combination of patient-derived glioma cell lines in xenograft models and syngeneic murine glioma models derived from ATRX KO cell lines and controls confirm a survival advantage in both immuno-competent mice and xenografts. Our models are under evaluation with PVSRIPO and other innate based RNA therapies. CONCLUSION Our data suggests that ATRX mutations may confer sensitivity to RNA-based innate immune signaling agonists in gliomas. This potential vulnerability can be targeted in future therapies.
APA, Harvard, Vancouver, ISO, and other styles
14

Wang, Yue, Bingbing Wu, Shengrong Long, QiangLiu, and Guangyu Li. "WNK3 promotes the invasiveness of glioma cell lines under hypoxia by inducing the epithelial-to-mesenchymal transition." Translational Neuroscience 12, no. 1 (January 1, 2021): 320–29. http://dx.doi.org/10.1515/tnsci-2020-0180.

Full text
Abstract:
Abstract Background The primary features of malignant glioma include high rates of mortality and recurrence, uncontrollable invasiveness, strong angiogenesis, and widespread hypoxia. The hypoxic microenvironment is an important factor affecting the malignant progression of glioma. However, the molecular mechanisms underlying glioma adaption in hypoxic microenvironments are poorly understood. Objective The work presented in this paper focuses on the role of WNK3 gene in glioma invasion under hypoxic conditions. Furthermore, we aim to explore its role in epithelial-to-mesenchymal transition (EMT). Methods ShRNA targeting WNK3 transfection was used to knockdown the WNK3 expression in U87 cells. We used western blot analysis to detect the relative expression of proteins in U87 cells. The effect of WNK3 on cell migration was explored using a transwell assay in the U87 cell line. We also evaluated WNK3 expression levels in glioma samples by immunohistochemistry analysis. Results WNK3 expression was significantly higher in high-grade (III and IV) gliomas than in low-grade (I and II) gliomas. WNK3 expression was up-regulated in U87 cells when cultured in a hypoxic environment in addition; WNK3 knockdown inhibited the invasion of U87 glioma cells by regulating the EMT, especially under hypoxic conditions. Conclusion These findings suggested that WNK3 plays an important role in the hypoxic microenvironment of glioma and might also be a candidate for therapeutic application in the treatment of glioma.
APA, Harvard, Vancouver, ISO, and other styles
15

Li, Ruiting, Yinghui Li, Xin Hu, Haiwei Lian, Lei Wang, and Hui Fu. "Transcription factor 3 controls cell proliferation and migration in glioblastoma multiforme cell lines." Biochemistry and Cell Biology 94, no. 3 (June 2016): 247–55. http://dx.doi.org/10.1139/bcb-2015-0162.

Full text
Abstract:
Transcription factor 3 (TCF3) is a member of the T-cell factor/lymphoid enhancer factor (TCF/LEF) transcription factor family. Recent studies have demonstrated its potential carcinogenic properties. Here we show that TCF3 was upregulated in glioma tissues compared with normal brain tissues. This upregulation of the TCF3 gene probably has functional significance in brain-tumor progression. Our studies on glioblastoma multiforme (GBM) cell lines show that knock-down of TCF3 induced apoptosis and inhibited cell migration. Further analysis revealed that down-regulation of TCF3 gene expression inhibits Akt and Erk1/2 activation, suggesting that the carcinogenic properties of TCF3 in GBM are partially mediated by the phosphatidylinositol 3-kinase–Akt and MAPK–Erk signaling pathways. Considered together, the results of this study demonstrate that high levels of TCF3 in gliomas potentially promote glioma development through the Akt and Erk pathways.
APA, Harvard, Vancouver, ISO, and other styles
16

Baltuch, Gordon H., Nora P. Dooley, William T. Couldwell, and Voon Wee Yong. "Staurosporine differentially inhibits glioma versus non-glioma cell lines." Journal of Neuro-Oncology 16, no. 2 (June 1993): 141–47. http://dx.doi.org/10.1007/bf01324701.

Full text
APA, Harvard, Vancouver, ISO, and other styles
17

Rakotomalala, Andria, Quentin Bailleul, Clara Savary, Mélanie Arcicasa, Maud Hamadou, Paul Huchedé, Audrey Hochart, et al. "H3.3K27M Mutation Controls Cell Growth and Resistance to Therapies in Pediatric Glioma Cell Lines." Cancers 13, no. 21 (November 5, 2021): 5551. http://dx.doi.org/10.3390/cancers13215551.

Full text
Abstract:
High-grade gliomas represent the most lethal class of pediatric tumors, and their resistance to both radio- and chemotherapy is associated with a poor prognosis. Recurrent mutations affecting histone genes drive the tumorigenesis of some pediatric high-grade gliomas, and H3K27M mutations are notably characteristic of a subtype of gliomas called DMG (Diffuse Midline Gliomas). This dominant negative mutation impairs H3K27 trimethylation, leading to profound epigenetic modifications of genes expression. Even though this mutation was described as a driver event in tumorigenesis, its role in tumor cell resistance to treatments has not been deciphered so far. To tackle this issue, we expressed the H3.3K27M mutated histone in three initially H3K27-unmutated pediatric glioma cell lines, Res259, SF188, and KNS42. First, we validated these new H3.3K27M-expressing models at the molecular level and showed that K27M expression is associated with pleiotropic effects on the transcriptomic signature, largely dependent on cell context. We observed that the mutation triggered an increase in cell growth in Res259 and SF188 cells, associated with higher clonogenic capacities. Interestingly, we evidenced that the mutation confers an increased resistance to ionizing radiations in Res259 and KNS42 cells. Moreover, we showed that H3.3K27M mutation impacts the sensitivity of Res259 cells to specific drugs among a library of 80 anticancerous compounds. Altogether, these data highlight that, beyond its tumorigenic role, H3.3K27M mutation is strongly involved in pediatric glioma cells’ resistance to therapies, likely through transcriptomic reprogramming.
APA, Harvard, Vancouver, ISO, and other styles
18

Wilson, Diana E., Anthony DiGianfilippo, Frank G. Ondrey, Kenning M. Anderson, and Jules E. Harris. "Effect of nordihydroguaiaretic acid on cultured rat and human glioma cell proliferation." Journal of Neurosurgery 71, no. 4 (October 1989): 551–57. http://dx.doi.org/10.3171/jns.1989.71.4.0551.

Full text
Abstract:
✓ When cultured malignant cells derived from rat gliomas (C6 and 9L) and human gliomas (A-172 and T98G) were treated for 4 hours with 1 to 80 µm nordihydroguaiaretic acid (NDGA) or 5,8,11,14-eicosatetraynoic acid (ETYA), a dose-dependent inhibition of deoxyribonucleic acid (DNA) synthesis occurred. In a series of three experiments for each cell line, 40 µM NDGA suppressed 3H-thymidine incorporation in the rat and human glioma lines to an average of less than 3.1% and 5.6% of control uptake (counts per minute), respectively. Incubation with a higher concentration of ETYA (80 µM) resulted in inhibition of rat and human DNA synthesis to less than 53% and 62% of control levels, respectively. This inhibition was not associated with any loss of cell viability, as judged by trypan blue exclusion studies. Prolonged incubation (for 72 hours) of the rat and human glioma cells with NDGA markedly decreased cell proliferation with no loss of cell viability. The inhibition of human glioma cell division by NDGA was rapidly reversible after incubation for 24 hours and at least partially reversible after incubation for 96 hours. It is concluded that the inhibitors of eicosanoid biosynthesis, NDGA and (to a lesser extent) ETYA, reduce in vitro cell proliferation in two glioma lines from both the rat and human. Since neither indomethacin nor acetylsalicylic acid altered DNA synthesis in these cell lines, this implicates the lipoxygenase products of arachidonic acid metabolism as important positive modulators in glioma cell division. These findings warrant further study in an in vivo system.
APA, Harvard, Vancouver, ISO, and other styles
19

Irvin, David, Hannah Roberts, Vladislav Sharin, Ahsan Farooqi, Sharvari Dharmaiah, Christian Alvarez, and Jason Huse. "CSIG-09. ATRX DEFICIENCY IN GLIOMA IMPACTS TRANSCRIPTIONAL PROFILES AND THE IMMUNE MICROENVIRONMENT IN VIVO." Neuro-Oncology 22, Supplement_2 (November 2020): ii29. http://dx.doi.org/10.1093/neuonc/noaa215.121.

Full text
Abstract:
Abstract Current treatment for diffuse astrocytoma fails to address its underlying molecular mechanisms leading to inevitable disease progression and eventual patient death. Genomic studies have implicated ATRX alterations as critical to low grade glioma biology. Our lab has previously shown in vitro that ATRX influences glioma motility, cellular differentiation state, and epigenetic programming, however, the influence of ATRX alterations in vivo remains unclear. Here, we leveraged an RCAS/tva mouse tumor model to probe the role of ATRX deficiency in glioma. Atrx deficient murine tumors exhibited lower histopathological grade and were associated with longer survival than Atrx-intact counterparts, and syngeneic allografts of cell lines derived from primary tumors mirrored the differential degrees of aggressiveness seen in primary tumors. Tumor-derived Atrx-deficient cell lines showed increased susceptibility to G-quadruplex stabilizing compounds, pointing to increased replication stress and recapitulating a key phenotype of ATRX-mutant gliomas in humans. Transcriptional profiling revealed enrichments in MYC target genes, E2F targets as well as G2/M checkpoint pathways in Atrx-intact tumors and cells, and enrichment in RAS signaling in Atrx-deficient tumors and cells. Finally, Atrx deficient murine gliomas displayed increased levels of NK cells, a phenotype recapitulated in ATRX-mutant human gliomas, and primary Atrx-deficient glioma lines exhibited increased levels of NK cell-attracting cytokines. These latter findings suggest that ATRX deficiency could influence interactions between glioma cells and their immune microenvironment by way of phenotypically relevant molecular mechanisms.
APA, Harvard, Vancouver, ISO, and other styles
20

Timerman, Dmitriy, and Caleb M. Yeung. "Identity confusion of glioma cell lines." Gene 536, no. 1 (February 2014): 221–22. http://dx.doi.org/10.1016/j.gene.2013.11.096.

Full text
APA, Harvard, Vancouver, ISO, and other styles
21

Huang, Suning, Gang Chen, Yiwu Dang, and Long-Hua Chen. "Overexpression of DcR3 and Its Significance on Tumor Cell Differentiation and Proliferation in Glioma." Scientific World Journal 2014 (2014): 1–7. http://dx.doi.org/10.1155/2014/605236.

Full text
Abstract:
Background.Overexpression of decoy receptor 3 (DcR3) have been reported in various classes of malignancies. However, its expression and clinicopathological contribution in gliomas has not been fully elucidated.Objective.To explore the expression and clinical significance of DcR3 protein in relation to tumor cell differentiation and proliferation in glioma cell lines and tissues.Methods.One hundred and twenty-five samples of glioma patients and 18 cases of normal brain tissues were recruited. The expression of DcR3 protein was detected using immunohistochemistry. Tumor differentiation was assessed by histologic characters and the status of glial fibrillary acidic protein (GFAP). Tumor cell labeling indexes (LIs) of Ki-67 and PCNA were also obtained. The relationship between the DcR3 level and clinicopathological features was investigated, including tumor differentiation, LIs, and survival. Meanwhile, the expression of DcR3 protein was also measured in the supernatants of 8 glioma cell lines and glioma cells freshly prepared from 8 human glioblastoma specimens by using western blot.Results.The level of DcR3 protein in gliomas was significantly higher than that in normal brain tissues (P<0.01). DcR3 expression showed positive correlations with tumor pathological grade(r=0.621,P<0.01)and negative with GFAP expression (r=-0.489,P<0.01). Furthermore, there were positive correlations between DcR3 expression and Ki-67, PCNA LIs (r=0.529,P<0.01;r=0.556,P<0.01). The survival in the DcR3 negative group was 50 ± 1.79 months, longer than that of the DcR3 positive group (48.36 ± 2.90), however, without significance(P=0.149). Different levels of DcR3 could also be detected in the culturing supernatants of all the 8 glioma cell lines and glioma cells freshly obtained from 8 human glioblastoma specimens.Conclusions.The overexpression of DcR3 might play a crucial role in the tumorigenesis, differentiation, and proliferation of glioma.
APA, Harvard, Vancouver, ISO, and other styles
22

Wild-Bode, Christine, Michael Weller, and Wolfgang Wick. "Molecular determinants of glioma cell migration and invasion." Journal of Neurosurgery 94, no. 6 (June 2001): 978–84. http://dx.doi.org/10.3171/jns.2001.94.6.0978.

Full text
Abstract:
Object. Migration and invasion are important prerequisites for the infiltrative and destructive growth patterns of malignant gliomas. Infiltrative growth prevents complete tumor resection and causes significant neurological morbidity and mortality. Methods. The authors assessed the expression of matrix metalloproteinases (MMPs) at messenger RNA and protein levels, MMP-2 and MMP-9 activities, and expression levels of a panel of anti- and proapoptotic proteins of the BCL-2 family. They then correlated their findings with αVβ3 integrin expression and the migratory and invasive potentials in 12 human malignant glioma cell lines. Multiple MMPs were expressed by most cell lines. The levels of MMP-2 and MMP-3 and the activities of MMP-2 and MMP-9 correlated with tumor cell invasion. Migration and invasion were also correlated. Although the expression levels of αVβ3 integrin did not predict migration or invasion, a neutralizing αVβ3 integrin antibody inhibited migration and invasion selectively in cell lines that contained a high level of αVβ3 integrin expression, thus indicating the important role of αVβ3 integrin for migration and invasion in this subset of cell lines. An expression pattern of BCL-2 family proteins that favor resistance to apoptosis was associated with enhanced migration, invasion, and MMP activity. Wild-type p53 cell lines migrated farther than mutant p53 cell lines. Conclusions. Activities of MMP-2 and MMP-9 are the best predictors of glioma cell invasion. The αVβ3 integrin mediates migration and invasion in a subset of glioma cell lines, but these processes do not depend on αVβ3 integrin expression. Antiapoptotic BCL-2 family protein expression is a predictor of efficient migration and invasion.
APA, Harvard, Vancouver, ISO, and other styles
23

Clark, Aaron J., Wagner G. Dos Santos, Jessica Mccready, Mike Y. Chen, Timothy E. Van Meter, Joy L. Ware, Sharon B. Wolber, Helen Fillmore, and William C. Broaddus. "Wilms tumor 1 expression in malignant gliomas and correlation of +KTS isoforms with p53 status." Journal of Neurosurgery 107, no. 3 (September 2007): 586–92. http://dx.doi.org/10.3171/jns-07/09/0586.

Full text
Abstract:
Object The WT1 gene is overexpressed in many types of human cancer. It has been demonstrated that Wilms tumor 1 (WT1) promotes tumor cell proliferation and survival in some cell lines by inhibiting p53-mediated apoptosis; however, this relationship has not been investigated in gliomas. The goal in this study was to characterize the expression pattern of WT1 in human gliomas and to determine if a correlation exists between WT1 expression and p53 status. Methods The authors screened nine malignant glioma cell lines, 50 glioblastoma multiforme (GBM) samples, and 16 lower-grade glial tumors for WT1 expression. Results Five of nine cell lines, 44 of 50 GBM samples, and 13 of 16 lower-grade gliomas expressed WT1 mRNA on reverse transcriptase polymerase chain reaction (PCR) analysis. Expression of WT1 was not detected in normal astrocytes. Two WT1 isoforms, +/+ and −/+, were expressed in the majority of these samples. Real-time PCR analysis of the GBM cell lines revealed that the level of WT1 mRNA ranged from 6.33 to 214.70 ng per ng 18S ribosomal RNA. The authors screened the GBM samples for p53 mutation by using PCR and single-stranded conformational polymorphism analysis, and they demonstrated an association between WT1 expression and p53 status. Tumors that contained wild-type p53 were significantly more likely to express WT1 than tumors that contained mutant p53. Conclusions The presence of WT1 in glioma cell lines and the majority of primary tumor samples and its absence in normal astrocytes support the suggestion that WT1 expression is important in glioma biology.
APA, Harvard, Vancouver, ISO, and other styles
24

Vogelbaum, Michael A., Jianxin X. Tong, Rajashri Perugu, David H. Gutmann, and Keith M. Rich. "Overexpression of bax in human glioma cell lines." Journal of Neurosurgery 91, no. 3 (September 1999): 483–89. http://dx.doi.org/10.3171/jns.1999.91.3.0483.

Full text
Abstract:
Object. Cells that lose their ability to undergo apoptosis may promote the development of neoplasms and result in resistance to clinical treatment with DNA-damaging modalities such as radio- and chemotherapy. Four established human glioma cell lines that are resistant to apoptosis were transfected with the proapoptotic gene bax and assessed for their sensitivity to a proapoptotic stimulus.Methods. Two cell lines had a wild-type p53 genotype (U87 and D247MG) and two had mutant p53 genotypes (U138 and U373). Constitutive overexpression of murine bax was achieved in U138 and U373 only, which resulted in an increased sensitivity of these lines to the apoptosis-inducing effect of cytosine arabinoside (ara-C). Multiple attempts to produce constitutive overexpression of bax in U87 and D247MG cells resulted in spontaneous, near-complete cell loss. Vector-only control transfections were successful in all four cell lines. Inducible overexpression of bax was achieved in the U87 cells and elevated levels of BAX were observed as early as 6 hours after gene induction. This overexpression of BAX resulted in the spontaneous induction of apoptosis in these cells.Conclusions. Overexpression of BAX in four human glioma cell lines resulted in increased sensitivity to apoptosis. In the two lines that had a wild-type p53 genotype, overexpression of BAX produced spontaneous apoptosis. In contrast, the lines that had mutant, nonfunctional P53 did not undergo spontaneous apoptosis, but they were rendered more sensitive to the apoptosis-inducing effect of ara-C. Modulation of BAX expression may be a useful therapeutic modality for gliomas, regardless of p53 genotype.
APA, Harvard, Vancouver, ISO, and other styles
25

Kouchi, Masaaki, Yuki Shibayama, Daisuke Ogawa, Keisuke Miyake, Akira Nishiyama, and Takashi Tamiya. "(Pro)renin receptor is crucial for glioma development via the Wnt/β-catenin signaling pathway." Journal of Neurosurgery 127, no. 4 (October 2017): 819–28. http://dx.doi.org/10.3171/2016.9.jns16431.

Full text
Abstract:
OBJECTIVEThe (pro)renin receptor (PRR) plays an essential role in the early development of the central nervous system by activating the Wnt/β-catenin signaling pathway. The authors investigated the potential role of the PRR in the pathogenesis of glioma.METHODSThe authors performed immunohistochemical analysis to detect both the PRR and isocitrate dehydrogenase 1 with mutations involving arginine 132 (IDH1R132H) in paraffin sections of 31 gliomas. Expression of the PRR and Wnt pathway components in cultured human glioma cell lines (U251MG, U87MG, and T98G) was measured using Western blotting. The effects of PRR short interfering RNA (siRNA) on glioma cell proliferation (WST-1 assay and direct cell counting) and apoptosis (flow cytometry and the caspase-3 assay) were also examined.RESULTSPRR expression was significantly higher in glioblastoma than in normal tissue or in lower grade glioma, regardless of IDH1R132H mutation. PRR expression was also higher in human glioblastoma cell lines than in human astrocytes. PRR expression showed a significant positive correlation with the Ki-67 labeling index, while it had a significant negative correlation with the survival time of glioma patients. Treatment with PRR siRNA significantly reduced expression of Wnt2, activated β-catenin, and cyclin D1 by human glioblastoma cell lines, and it reduced the proliferative capacity of these cell lines and induced apoptosis.CONCLUSIONSThis is the first evidence that the PRR has an important role in development of glioma by aberrant activation of the Wnt/β-catenin signaling pathway. This receptor may be both a prognostic marker and a therapeutic target for glioma.
APA, Harvard, Vancouver, ISO, and other styles
26

Zhang, Danfeng, Dawei Dai, Mengxia Zhou, Zhenxing Li, Chunhui Wang, Yicheng Lu, Yiming Li, and Junyu Wang. "Inhibition of Cyclin D1 Expression in Human Glioblastoma Cells is Associated with Increased Temozolomide Chemosensitivity." Cellular Physiology and Biochemistry 51, no. 6 (2018): 2496–508. http://dx.doi.org/10.1159/000495920.

Full text
Abstract:
Background/Aims: Cyclin D1 (CCND1) is frequently overexpressed in malignant gliomas. We have previously shown ectopic overexpression of CCND1 in human malignant gliomas cell lines. Methods: Quantitative reverse transcriptase PCR (qRT-PCR) and Western Blot (WB) was performed to investigate the expression of CCND1 in glioma tissues and cell lines. The biological function of CCND1 was also investigated through knockdown and overexpression of BCYRN1 in vitro. Results: Here we reported that CCND1 expression was positively associated with the pathological grade and proliferative activity of astrocytomas, as the lowest expression was found in normal brain tissue (N = 3) whereas the highest expression was in high-grade glioma tissue (N = 25). Additionally, we found that the expression level of CCND1 was associated with IC50 values in malignant glioma cell lines. Forced inhibition of CCND1 increased temozolomide efficacy in U251 and SHG-44 cells. After CCND1 overexpression, the temozolomide efficacy decreased in U251 and SHG-44 cells. Colony survival assay and apoptosis analysis confirmed that CCND1 inhibition renders cells more sensitive to temozolomide treatment and temozolomide-induced apoptosis in U251 and SHG-44 cells. Inhibition of P-gp (MDR1) by Tariquidar overcomes the effects of CCND1 overexpression on inhibiting temozolomide-induced apoptosis. Inhibition of CCND1 inhibited cell growth in vitro and in vivo significantly more effectively after temozolomide treatments than single temozolomide treatments. Finally, inhibition of CCND1 in glioma cells reduced tumor volume in a murine model. Conclusion: Taken together, these data indicate that CCND1 overexpression upregulate P-gp and induces chemoresistance in human malignant gliomas cells and that inhibition of CCND1 may be an effective means of overcoming CCND1 associated chemoresistance in human malignant glioma cells.
APA, Harvard, Vancouver, ISO, and other styles
27

Xi, Yan-Guo, Deng-Peng Ren, Jing-Yun Jin, Lei Zhu, Tai-Long Yi, Xue-Fei Shao, Sheng-Kai Sun, Wen-Bin Zhang, and Shi-Xiang Cheng. "Casein Kinase 2 Interacting Protein-1 Suppresses Glioma Cell Proliferation via Regulating the AKT/GSK3β/β-Catenin Pathway." BioMed Research International 2019 (July 2, 2019): 1–11. http://dx.doi.org/10.1155/2019/5653212.

Full text
Abstract:
Objective. Casein kinase 2 interacting protein-1 (CKIP-1) has exhibited multiple functions in regulating cell proliferation, apoptosis, differentiation, and cytoskeleton. CKIP-1 also plays an important role as a critical regulator in tumorigenesis. The aim of this study is to further examine the function of CKIP-1 in glioma cells. Methods. The expression level of CKIP-1 protein was determined in gliomas tissues and cell lines by immunohistochemistry stain and western blotting while the association of CKIP-1 expression with prognosis was analyzed by Kaplan-Meier method and compared by log-rank test. CKIP-1 was overexpressed or silenced in gliomas cell lines. CCK-8, colony formation assay, and BrdU incorporation assay were used to determine cell proliferation and DNA synthesis. Cell cycle and apoptosis rate were determined with fluorescence-activated cell sorting (FACS) method. Then, expression of key members in AKT/GSK3β/β-catenin pathway was detected by western blot analysis. Results. In the present study, we reported new evidence that CKIP-1 was reversely associated with the proliferation of glioma cells and survival in glioma patients. Additionally, the overexpressed CKIP-1 significantly inhibited glioma cell proliferation. Further experiments revealed that CKIP-1 functioned through its antiproliferative and proapoptotic activity in glioma cells. Importantly, mechanistic investigations suggested that CKIP-1 sharply suppressed the activity of AKT by inhibiting the phosphorylation, markedly downregulated the phosphorylated GSK3β at Ser9, and promoted β-catenin degradation. Conclusions. Overall, our results provided new insights into the clinical significance and molecular mechanism of CKIP-1 in glioma, which indicated CKIP1 might function as a therapeutic target for clinical treatment of glioma.
APA, Harvard, Vancouver, ISO, and other styles
28

Zhou, Yan, Peter H. Larsen, Chunhai Hao, and V. Wee Yong. "CXCR4 Is a Major Chemokine Receptor on Glioma Cells and Mediates Their Survival." Journal of Biological Chemistry 277, no. 51 (October 17, 2002): 49481–87. http://dx.doi.org/10.1074/jbc.m206222200.

Full text
Abstract:
Chemokines were described originally in the context of providing migrational cues for leukocytes. They are now known to have broader activities, including those that favor tumor growth. We addressed whether and which chemokines may be important promoters of the growth of the incurable brain neoplasm, malignant gliomas. Analyses of 16 human glioma lines for the expression of chemokine receptors belonging to the CXCR and CCR series revealed low to negligible levels of all receptors, with the exception of CXCR4 that was expressed by 13 of 16 lines. All six resected human glioma specimens showed similarly high CXCR4 expression. The CXCR4 on glioma lines is a signaling receptor in that its agonist, stromal cell-derived factor-1 (SDF-1; CXCL12), produced rapid phosphorylation of mitogen-activated protein kinases. Furthermore, SDF-1 induced the phosphorylation of Akt (protein kinase B), a kinase associated with survival, and prevented the apoptosis of glioma cells when serum was withdrawn from the culture medium. SDF-1 also mediated glioma chemotaxis, in accordance with this better known role of chemokines. We conclude that glioma cells express a predominant chemokine receptor, CXCR4, and that this functions to regulate survival in part through activating pathways such as Akt.
APA, Harvard, Vancouver, ISO, and other styles
29

Rao, Aparna, Xiaoran Zhang, Christopher Deibert, Paola Sette, Paola Grandi, and Nduka Amankulor. "IDH Mutant Gliomas Escape Natural Killer Cell Immune Surveillance by Downregulation of NKG2D Ligand Expression." Journal of Immunology 196, no. 1_Supplement (May 1, 2016): 142.11. http://dx.doi.org/10.4049/jimmunol.196.supp.142.11.

Full text
Abstract:
Abstract Background Diffuse gliomas are fatal primary brain tumors that are poorly immunogenic. The basis for insufficient anti-tumor immunity in diffuse gliomas is not understood. Mutations in isocitrate dehydrogenases (IDH1 and IDH2) promote diffuse glioma formation through epigenetic reprogramming of a number of genes, including immune-related genes. Here, we identify epigenetic dysregulation of natural killer (NK) cell ligand genes as significant contributors to immune escape in glioma. Methods We analyzed the TCGA database for immune gene expression patterns in IDH mutant or wild-type gliomas and identified differentially expressed immune genes. NKG2D ligands expression levels and NK cell-mediated lysis were measured in patient-derived glioma stem cells and in genetically engineered astrocytes. Finally, we assessed the impact of hypomethylating agent Decitabine as a potential NK cell sensitizing agent in IDH mutant cells. Results All IDH mutant glioma stem-like cell lines exhibited significantly lower expression of NKG2D ligands compared with IDH wild-type cells. Consistent with these findings, IDH mutant glioma cells and astrocytes were resistant to NK cell-mediated lysis. The hypomethylating agent Decitabine increased NKG2D ligand expression, and restored NK- mediated lysis of IDH mutant cells in an NKG2D-dependent manner. Conclusions IDH mutant glioma cells acquire resistance to NK cells through epigenetic silencing of NKG2D ligands ULBP1 and ULPB3. Decitabine-mediated hypomethylation restores ULBP1 and ULBP3 expression in IDH mutant glioma cells and may provide a clinically useful method to sensitize IDH mutant gliomas to NK cell-mediated immune surveillance in patients with IDH mutated diffuse gliomas.
APA, Harvard, Vancouver, ISO, and other styles
30

Jin, Linchun, Haitao Ge, Changlin Yang, Yu Long, Yifan(Emily) Chang, Luyan Mu, Elias Sayour, et al. "CD70 as a novel target of CAR-T-cell therapy for gliomas." Journal of Clinical Oncology 35, no. 7_suppl (March 1, 2017): 148. http://dx.doi.org/10.1200/jco.2017.35.7_suppl.148.

Full text
Abstract:
148 Background: Gliomas are the most common primary malignant brain tumor and are uniformly lethal. Cancer immunotherapy has the potential to target gliomas; however, its antitumor effects are restricted by limitations in clinically useful tumor specific targets. Chimeric antigen receptor modified T-cell (CAR-T) therapy is a highly promising option for cancer treatment, due to its combination of precision antibody recognition and T-cell tumor-specific killing. CD70 is an antigen expressed by limited subsets of normal lymphocytes and dendritic cells but is aberrantly overexpressed by glioma cells, which makes it an outstanding glioma-antigen target. Methods: The gene and protein expression of CD70 were evaluated to identify its potential as a glioma target. Human and mouse versions of CD70-specific CAR-T cells were generated, and human primary GBM lines as well as murine lines (GL-261, KR-158B) were used as human and mouse tumor targets, respectively. The antitumor effect of the human and mouse CD70-sepecific CARs were tested in vitro and in orthotopic xenograft and syngeneic murine models. Results: CD70 is only overexpressed by tumor cells in a subset of low-grade gliomas and GBM. The elevated gene and protein expression are associated with increased tumor grade and poor patient survival. Co-culturing CD70-specific CAR-T cells with CD70-positive glioma cells resulted in potent secretion of IFN-gamma and tumor-specific killing in a CD70-dependent manner. Irradiation enhances CD70 expression on glioma cells and thus increases CAR T-cell recognition. Adoptive transfer of the human and mouse CD70 CAR-T cells resulted in tumor regression of immunocompetent and immunodeficient mice, respectively. Conclusions: CD70 can be an excellent tumor target for gliomas, and CD70-specific CAR-T cells have potent antitumor activity against CD70-positive gliomas both in vitro and in vivo. Our study provides crucial preclinical evidence to support the future clinical application of CD70 CAR-T cells to treat gliomas.
APA, Harvard, Vancouver, ISO, and other styles
31

Tabatabai, Ghazaleh, Shanmugarajan Krishnan, Ana-Maria Florea, Karl Frei, Kathy Hasenbach, Guido Reifenberger, Niklaus Krayenbuehl, Michael Weller, and Hans-Georg Wirsching. "Effect of silencing thymosin beta 4 gene expression on stemness and invasiveness in glioblastoma." Journal of Clinical Oncology 31, no. 15_suppl (May 20, 2013): 2081. http://dx.doi.org/10.1200/jco.2013.31.15_suppl.2081.

Full text
Abstract:
2081 Background: Thymosin β4 (TB4) is a pleiotropic actin-sequestering polypeptide that is involved in wound healing and developmental processes. TB4 gene silencing promotes differentiation of neural progenitor cells whereas TB4 overexpression initiates cortical folding of developing brain hemispheres. However, a role of TB4 in malignant gliomas has not yet been investigated. Methods: We first analyzed TB4 expression on tissue microarrays and performed REMBRANDT and TCGA database interrogations. We analyzed TB4 expression in a panel of 8 long-term glioma cell lines and 7 glioma-initiating cell lines. Using lentiviral transduction, we modulated TB4 expression in LNT-229, U87MG and the glioma-initiating cell line GS-2. We studied clonogenic survival, migration, invasion, self-renewal, differentiation capacity of TB4-depleted or TB4-overexpressing glioma cells in vitro and tumorigenicity upon orthotopic implantation in vivo. Finally, we performed an Affymetrix gene chip analysis to unravel the molecular network of TB4 signaling effects. Results: TB4 expression increased with the grade of malignancy in gliomas and correlated with patient survival. In vitro, TB4 gene silencing by lentiviral transduction decreased migration, invasion, growth and self-renewal, and promoted differentiation and the susceptibility to undergo apoptotic cell death upon nutrient depletion in LNT-229, U87MG and the glioma stem-cell line GS2, respectively. In vivo, survival of nude mice bearing tumors derived from TB4-depleted glioma cells was improved and the tumorigenicity of the GS2 glioma stem-cell line was decreased. The gene expression pattern was shifted from the mesenchymal towards the pro-neural gene signature upon TB4 gene silencing. The clustering of differentially regulated genes involved TGF-β and p53 signaling networks. Conclusions: TB4 may be a key regulator of malignancy in glioblastoma and therefore a novel candidate molecular target for anti-glioma therapies.
APA, Harvard, Vancouver, ISO, and other styles
32

Qing, Mingjie, Jiahao Zhou, Weijian Chen, and Lijuan Cheng. "Highly Expressed CYBRD1 Associated with Glioma Recurrence Regulates the Immune Response of Glioma Cells to Interferon." Evidence-Based Complementary and Alternative Medicine 2021 (July 16, 2021): 1–12. http://dx.doi.org/10.1155/2021/2793222.

Full text
Abstract:
Invasiveness, resistance to treatment, and recurrence of gliomas are significant hurdles to successful treatment regimens. Data sets from Gene Expression Omnibus (GEO), CGGA-RNAseq, and The Cancer Genome Atlas Glioblastoma Multiforme (TCGA-GBM) were analyzed, and an increased expression of Cytochrome B Reductase 1 (CYBRD1) was identified and could be associated with aggravated clinical outcomes. Gene ontology (GO) enrichment analysis indicated that CYBRD1 co-expressed genes are enriched during an immune response. CYBRD1 overexpression in glioma cell lines is enhanced, whereas CYBRD1 silencing attenuated the aggressiveness of glioma cells. In IFN-α-treated glioma cells, IFN-α suppressed the viability and migratory ability and invasive ability of glioma cells, whereas CYBRD1 overexpression attenuated the antitumor effects of IFN-α. CYBRD1 could potentially serve as a biomarker for glioma recurrence. CYBRD1 overexpression enhances glioma cell aggressiveness and attenuates glioma cell response to IFN-α.
APA, Harvard, Vancouver, ISO, and other styles
33

Wakabayashi, Toshihiko, Jun Yoshida, Hisao Seo, Kyoto Kazo, Yoshiharu Murata, Nobuo Matsui, and Naoki Kageyama. "Characterization of neuroectodermal antigen by a monoclonal antibody and its application in CSF diagnosis of human glioma." Journal of Neurosurgery 68, no. 3 (March 1988): 449–55. http://dx.doi.org/10.3171/jns.1988.68.3.0449.

Full text
Abstract:
✓ Monoclonal antibodies were produced by immunization of the human glioma cell line SK-MG-4. One of the antibodies, designated G-22, reacted with 18 of 20 glioma cell lines, two melanoma cell lines, and three lung cancer cell lines, but not with 39 cell lines derived from sarcoma, carcinoma, or hematopoietic tumors. The antigen was expressed in the brain of human fetuses in early gestation (9 weeks) but not in late gestation (8 months) or in normal adult brain, suggesting that the antibody recognizes neural differentiation antigens expressed by neuroectodermal origin. A high incidence of positive antigens has been observed in gliomas but not in the other neural tumors, such as ependymomas, meningiomas, and neuroblastomas. Thus, the antigen defined by the G-22 monoclonal antibody could be defined as glioma-associated antigen. Pulse-labeling with tritiated leucine and subsequent immunoprecipitation of the solubilized cell membrane revealed that the antigen recognized by this antibody had a molecular weight of 67 kD on sodium dodecyl sulfate-poly-acrylamide gel electrophoresis (SDS-PAGE). It was shown by dot-blot enzyme-linked immunospecific assay (ELISA) that the antigen could be detected in the cerebrospinal fluid (CSF) from patients with gliomas. From analysis of affinity chromatography and SDS-PAGE, the antigen present in the CSF had a molecular weight similar to that of a 1% Nonidet P-40 (NP-40) extract from a glioma cell line. When the antigen in CSF was quantitatively assayed by ELISA, the mean antigen level (expressed as optical density at 450 nm) in the CSF of seven patients was 0.8 ± 0.28 (mean ± standard deviation), which was significantly higher than the 0.38 ± 0.14 level observed in the CSF of 15 patients with nonglioma brain tumors and the 0.23 ± 0.09 level in the CSF of four patients without brain tumors. These results indicate that the monoclonal antibody G-22 is useful for the diagnosis of glioma.
APA, Harvard, Vancouver, ISO, and other styles
34

Wang, Zengliang, Yirizhati Aili, Yongxin Wang, Nuersimanguli Maimaitiming, Hu Qin, Wenyu Ji, Guofeng Fan, and Bo Li. "The RPL4P4 Pseudogene Is a Prognostic Biomarker and Is Associated with Immune Infiltration in Glioma." Oxidative Medicine and Cellular Longevity 2022 (August 9, 2022): 1–28. http://dx.doi.org/10.1155/2022/7967722.

Full text
Abstract:
Objective. Research over the past decade has suggested important roles for pseudogenes in gliomas. Our previous study found that the RPL4P4 pseudogene is highly expressed in gliomas. However, its biological function in gliomas remains unclear. Methods. In this study, we analyzed clinical data on patients with glioma obtained from The Cancer Genome Atlas (TCGA), the Chinese Glioma Genome Atlas (CGGA), the Genotype-Tissue Expression (GTEx), and the GEPIA2 databases. We used the R language for the main analysis. Correlations among RPL4P4 expression, pathological characteristics, clinical outcome, and biological function were evaluated. In addition, the correlations of RPL4P4 expression with immune cell infiltration and glioma progression were analyzed. Finally, wound healing, Transwell, and CCK-8 assays were performed to analyze the function of RPL4P4 in glioma cells. Result. We found that RPL4P4 is highly expressed in glioma tissues and is associated with poor prognosis, IDH1 wild type, codeletion of 1p19q, and age. Multivariate analysis and the nomogram model showed that high RPL4P4 expression was an independent risk factor for glioma prognosis and had better prognostic prediction power. Moreover, high RPL4P4 expression correlated with immune cell infiltration, which showed a significant positive association with M2-type macrophages. Finally, RPL4P4 knockdown in glioma cell lines caused decreased glioma cell proliferation, invasion, and migration capacity. Conclusion. Our data suggest that RPL4P4 can function as an independent prognostic predictor of glioma. It also shows that RPL4P4 expression correlates with immune cell infiltration and that targeting RPL4P4 may be a new strategy for the treatment of glioma patients.
APA, Harvard, Vancouver, ISO, and other styles
35

Elahi, Lubayna, Matthew Garrett, Lea Guo, Michael Condro, Riki Kawaguchi, Shwetal Mehta, Albert Lai, Whitney Pope, and Harley Kornblum. "CBMT-31. HDAC INHIBITION DIMINISHES THE GROWTH OF ENDOGENOUS IDH MUTANT GLIOMAS." Neuro-Oncology 21, Supplement_6 (November 2019): vi39—vi40. http://dx.doi.org/10.1093/neuonc/noz175.153.

Full text
Abstract:
Abstract Histone deacetylase inhibitors (HDACi’s) have emerged as a promising class of drugs for treatment of malignancies such as glioblastoma (GBM). Several studies have demonstrated the anti-tumor property of HDACi’s against GBM in both in vitro and in vivo experiments. Nonetheless, in clinical trials, HDACi only marginally increased overall survival of patients with GBM. The mixed results of trials with HDACi’s in glioma have prompted us to hypothesize that improved selection of patients by tumor characteristics could enhance the efficacy of therapy. We specifically tested the effects of valproic acid (VPA), a HDACi and an antiepileptic drug against IDH mutant gliomas. We have previously demonstrated that our IDH mutant glioma cell lines have gene expression and methylation patterns highly similar to IDH mutant tumors in situ. Mutant IDH1 alters the epigenetic landscape of gliomas leading to the hypermethylation phenotype and transcriptional repression of genes. This repression of genes may contribute to tumorigenesis and progression of IDH mutant gliomas. We found that VPA inhibits the growth of patient-derived IDH1 mutant glioma lines. In addition, RNA sequencing analysis of vehicle and VPA-treated IDH1 mutant glioma cells showed de-repression of several genes previously shown to be downregulated in IDH1 mutant glioma cell lines. We also treated cells with another HDACi LBH589 and found that both VPA and LBH589 upregulates similar gene sets suggesting that HDAC inhibition promotes de-repression of previously repressed genes. Ongoing studies are aimed at determining the molecular mechanism by which VPA regulates the growth of IDH1 mutant tumors.
APA, Harvard, Vancouver, ISO, and other styles
36

Pollack, Ian F., Margaret S. Randall, Matthew P. Kristofik, Robert H. Kelly, Robert G. Selker, and Frank T. Vertosick. "Response of malignant glioma cell lines to activation and inhibition of protein kinase C-mediated pathways." Journal of Neurosurgery 73, no. 1 (July 1990): 98–105. http://dx.doi.org/10.3171/jns.1990.73.1.0098.

Full text
Abstract:
✓ To evaluate the role of protein kinase C-mediated pathways in the proliferation of malignant gliomas, this study examined the effect of a protein kinase C (PKC)-activating phorbol ester (12-O-tetradecanoyl-13-phorbol acetate or TPA) and a protein kinase C inhibitor (polymyxin B) on deoxyribonucleic acid (DNA) synthesis of malignant glioma cells in vitro. A serum-free chemically defined medium, MCDB 105, was employed for all studies. Two established human malignant glioma cell lines (T98G and U138), two rat glioma lines (9L and C6), and two low-passage human glioma lines (obtained from surgical specimens) were studied. With the exception of the C6 line, all tumors responded in a dose-dependent fashion to nanomolar concentrations of TPA with a median effective dose that varied from 0.5 ng/ml for the U138 glioma to 1 ng/ml for the T98G glioma. At optimal concentrations (5 to 10 ng/ml), TPA produced a two- to five-fold increase in the rate of DNA synthesis (p < 0.05) as assessed by incorporation of 3H-thymidine. However, TPA had no additive effect on the mitogenic response produced by epidermal growth factor (EGF) or platelet-derived growth factor (PDGF). Inhibition of PKC using the antibiotic polymyxin B (20 µg/ml) abolished the TPA-induced mitogenic response in the five responsive lines tested. In two tumors (U138 and 9L), polymyxin B also eliminated EGF-, PDGF-, and serum-induced DNA synthesis as well as abolishing baseline DNA synthesis. These cells remained viable, however, as assessed by trypan blue exclusion; after removal of polymyxin B from the medium, they were able to resume DNA synthesis in response to TPA and serum. In the three other tumors (T98G and the two low-passage human glioma lines), growth factor-induced and serum-induced DNA synthesis were inhibited by approximately 25% to 85%. It is concluded that PKC-mediated pathways affect DNA synthesis in the human malignant glial tumors studied. The response of the glioma cells to TPA is similar to the responses seen in fetal astrocytes, but differs significantly from those reported for normal adult glial cultures. Because the response of the 9L glioma to TPA is similar to the responses seen in the human tumors, the 9L rat glioma model may prove useful for examining the role of PKC-mediated pathways in controlling glioma growth in vivo.
APA, Harvard, Vancouver, ISO, and other styles
37

Verheul, C., T. V. Kers, A. Van Der Ploeg, M. Van Der Kaaij, M. Aghababazadeh, M. De Wit, Y. Hoogstrate, et al. "P11.47 Generation, characterisation and drug screening of patient-derivedIDH1-mutated glioma cell lines." Neuro-Oncology 21, Supplement_3 (August 2019): iii54. http://dx.doi.org/10.1093/neuonc/noz126.193.

Full text
Abstract:
Abstract BACKGROUND Despite considerable scientific efforts, endogenous in vitro isocitrate dehydrogenase (IDH)-mutated glioma models remain scarce. Availability of these models is key to understanding underlying molecular mechanisms and vital for the development of new therapeutic interventions. We established and characterized a set of seven cell lines derived from IDH1-mutated gliomas and utilized them to investigate IDH-mutant glioma drug-response in vitro. MATERIAL AND METHODS Fresh tumor material was collected directly from the operating room, mechanically and enzymatically dissociated and cultured under serum-free culture conditions. IDH mutation status was verified at several passages with Sanger sequencing, as well as with whole exome sequencing. D-2-hydroxyglutarate (D2-HG) levels were determined with mass-spectrometry. Genome-wide methylation profiling was performed using the Infinium MethylationEpic BeadChip array. Cell viability was measured using the ATP-based CellTiter-Glo assay. Cell proliferation was determined through cell counting. Drug screens were performed with an FDA-approved anti-cancer drug set from the NIH as well as two IDH-mutant specific inhibitors. RESULTS Over the last decade our lab processed over 800 glioma samples from all WHO grades and IDH mutational status. Optimisation of culture conditions led to the establishment of seven IDH1-mutant glioma cell lines. The IDH1-mutation was present during all tested passages in these cell lines. Whole exome sequencing showed a high concordance between tumor and cell lines with regard to driver gene mutations. Similarly, copy-number changes based on genome-wide methylation data also show a close resemblance between the parental tumor and resulting cell lines. The Heidelberg methylation profiler classified each cell line and its parental tumor as IDH mutant glioma. When exposed to an IDH1-mutant specific inhibitor, all cell lines showed a concentration-dependent decrease of D2-HG levels. The inhibitors showed little to no effect on viability up to 10uM during 8 days, however, long term treatment up to 4 weeks revealed a decrease in cell doubling time in 2 of 6 cultures. Drug screen results either alone or in combination with an IDH1-mutant specific inhibitor identified several interesting candidates that are currently followed up in additional experiments. CONCLUSION We established a unique set of patient-derived IDH1-mutant glioma cell lines that closely resemble their respective tumors and can be used to identify new therapeutic strategies.
APA, Harvard, Vancouver, ISO, and other styles
38

Zhang, Lei, Eiji Sato, Kenichi Amagasaki, Atsuhito Nakao, and Hirofumi Naganuma. "Participation of an abnormality in the transforming growth factor–β signaling pathway in resistance of malignant glioma cells to growth inhibition induced by that factor." Journal of Neurosurgery 105, no. 1 (July 2006): 119–28. http://dx.doi.org/10.3171/jns.2006.105.1.119.

Full text
Abstract:
Object Malignant glioma cells secrete and activate transforming growth factor–β (TGFβ) and are resistant to growth inhibition by that factor. Nevertheless, the mechanism underlying this effect remains poorly understood. In this study, the mechanism of the resistance to growth inhibition induced by TGFβ was investigated. Methods The authors examined the expression of downstream components of the TGFβ receptor, including Smad2, Smad3, Smad4, and Smad7, and the effect of TGFβ1 treatment on the phosphorylation of Smad2 and the nuclear translocation of Smad2 and Smad3 by using 10 glioma cell lines and the A549 cell line, which is sensitive to TGFβ-mediated growth inhibition. The expression of two transcriptional corepressor proteins, SnoN and Ski, and the effect of TGFβ1 treatment on the expression of the SnoN protein and the cell cycle regulators p21, p15, cyclin-dependent kinase–4 (CDK4), and cyclin D1 were also examined. Expression of the Smad2 and Smad3 proteins was lower in the glioma cell lines than in the A549 cell line and in normal astrocytes. In particular, Smad3 expression was low or very low in nine of the 10 malignant glioma cell lines. Expression of Smad4 was low in four glioma cell lines, and expression of the Smad7 protein was similar when compared with protein expression in the A549 cell line and in normal astrocytes. The levels of Smad2 phosphorylation after TGFβ1 treatment were lower in glioma cell lines than in the A549 cell line, except for one glioma cell line. Seven of the 10 glioma cell lines exhibited lower levels of nuclear translocation of Smad2 and Smad3, and two cell lines that expressed very low levels of Smad3 protein showed no nuclear translocation. All glioma cell lines expressed the SnoN protein and its expression was unaltered by treatment with TGFβ1. Three glioma cell lines expressed high levels of the Ski protein. The expression of the p21cip1, p15INK4B, CDK4, and cyclin D1 proteins was not altered by TGFβ1 treatment, except in one cell line that displayed a slight increase in p21 protein. Overall, the expression of the Smad2 and Smad3 proteins was low in the glioma cell lines, the phosphorylation and nuclear translocation of Smad2 and Smad3 were impaired, and the TGFβ receptor signal did not affect the expression of the SnoN, p21, p15, cyclin D1, and CDK4 proteins. Conclusions These results suggest that the ability to resist TGFβ-mediated growth inhibition in malignant glioma cells is due to abnormalities in the TGFβ signaling pathway.
APA, Harvard, Vancouver, ISO, and other styles
39

Cerchia, Laura, Carla Lucia Esposito, Andreas H. Jacobs, Bertrand Tavitian, and Vittorio de Franciscis. "Differential SELEX in Human Glioma Cell Lines." PLoS ONE 4, no. 11 (November 24, 2009): e7971. http://dx.doi.org/10.1371/journal.pone.0007971.

Full text
APA, Harvard, Vancouver, ISO, and other styles
40

MARK, JOACHIM, BENGT WESTERMARK, JAN PONTÉN, and RUNE HUGOSSON. "Banding patterns in human glioma cell lines." Hereditas 87, no. 2 (February 12, 2009): 243–60. http://dx.doi.org/10.1111/j.1601-5223.1978.tb01267.x.

Full text
APA, Harvard, Vancouver, ISO, and other styles
41

de Menezes, Weder Pereira, Viviane Aline Oliveira Silva, Izabela Natália Faria Gomes, Marcela Nunes Rosa, Maria Luisa Corcoll Spina, Adriana Cruvinel Carloni, Ana Laura Vieira Alves, et al. "Loss of 5′-Methylthioadenosine Phosphorylase (MTAP) is Frequent in High-Grade Gliomas; Nevertheless, it is Not Associated with Higher Tumor Aggressiveness." Cells 9, no. 2 (February 20, 2020): 492. http://dx.doi.org/10.3390/cells9020492.

Full text
Abstract:
The 5’-methylthioadenosine phosphorylase (MTAP) gene is located in the chromosomal region 9p21. MTAP deletion is a frequent event in a wide variety of human cancers; however, its biological role in tumorigenesis remains unclear. The purpose of this study was to characterize the MTAP expression profile in a series of gliomas and to associate it with patients’ clinicopathological features. Moreover, we sought to evaluate, through glioma gene-edited cell lines, the biological impact of MTAP in gliomas. MTAP expression was evaluated in 507 glioma patients by immunohistochemistry (IHC), and the expression levels were associated with patients’ clinicopathological features. Furthermore, an in silico study was undertaken using genomic databases totalizing 350 samples. In glioma cell lines, MTAP was edited, and following MTAP overexpression and knockout (KO), a transcriptome analysis was performed by NanoString Pan-Cancer Pathways panel. Moreover, MTAP’s role in glioma cell proliferation, migration, and invasion was evaluated. Homozygous deletion of 9p21 locus was associated with a reduction of MTAP mRNA expression in the TCGA (The Cancer Genome Atlas) - glioblastoma dataset (p < 0.01). In addition, the loss of MTAP expression was markedly high in high-grade gliomas (46.6% of cases) determined by IHC and Western blotting (40% of evaluated cell lines). Reduced MTAP expression was associated with a better prognostic in the adult glioblastoma dataset (p < 0.001). Nine genes associated with five pathways were differentially expressed in MTAP-knockout (KO) cells, with six upregulated and three downregulated in MTAP. Analysis of cell proliferation, migration, and invasion did not show any significant differences between MTAP gene-edited and control cells. Our results integrating data from patients as well as in silico and in vitro models provide evidence towards the lack of strong biological importance of MTAP in gliomas. Despite the frequent loss of MTAP, it seems not to have a clinical impact in survival and does not act as a canonic tumor suppressor gene in gliomas.
APA, Harvard, Vancouver, ISO, and other styles
42

Smith, Louise, Sajib Chakraborty, Anbarasu Lourdusamy, Alan McIntyre, and Stuart Smith. "The Role of miRNAs in Gliomas in Response to Hypoxia." Neuro-Oncology 24, Supplement_4 (October 1, 2022): iv9. http://dx.doi.org/10.1093/neuonc/noac200.041.

Full text
Abstract:
Abstract AIMS Identifying significant miRNAs that are either under or overexpressed in hypoxia compared to normoxia in glioma cells. Further exploring the effect and role of hypoxia on miR-92a-3p and miR-149-5p in gliomas in apoptotic and cellular senescence pathways. METHOD A range of glioma cells were used for screening, including primary cell lines: GIN28 and GIN31; low-grade cell lines: LGG19 and LGG24; a paediatric cell line: SF188 and a commercially available glioblastoma cell line U87. These cells were cultured at both 1% (hypoxia) and 20% (normoxia) oxygen levels. Screening of miRNAs was achieved by quantitative polymerase chain reaction (qPCR) using miRNA specific primers. Knockdowns/in were achieved by transfecting with miR-92a-3p and miR-149-5p mimics and inhibitors. Caspase-glo assay was used to assess the effect of hypoxia on apoptosis. RESULTS A range of miRNAs were differentially expressed in hypoxia in a range of glioma cell lines. miRNA 92a-3p and miR-149-5p were found to be downregulated and upregulated respectively in primary gliomablastoma cells in hypoxia compared to normoxia. These particular miRNAs are also found to have multiple targets in apoptosis and cellular senescence. CONCLUSION The screening of 90 miRNAs among the different categories of gliomas highlighted multiple miRNAs that were significant in hypoxia compared to normoxia. Using primary cell lines have identified hypoxia-affected miRNAs in glioblastomas. This discovery will increase our knowledge and understanding of the hypoxia effect on miRNAs which may aid and direct targeted therapy to conquer hypoxia in glioblastomas.
APA, Harvard, Vancouver, ISO, and other styles
43

Keough, M., K. Shamardani, and M. Monje. "P.103 Imaging neuron-glioma cell interactions in freely behaving animals with a novel implantable mini-microscope." Canadian Journal of Neurological Sciences / Journal Canadien des Sciences Neurologiques 50, s2 (June 2023): S85. http://dx.doi.org/10.1017/cjn.2023.196.

Full text
Abstract:
Background: High grade gliomas (HGG) are diffusely infiltrative brain tumours with dismal prognosis. Recent studies from our lab have demonstrated that glioma cells form synapses with surrounding neurons, and proliferate in response to neuronal input. How these neuron-glioma networks develop, and are influenced by experience, is currently unknown. We aimed to develop a novel imaging tool to study neuron-glioma cell interactions in freely behaving animals. Methods: Several patient-derived HGG cell lines were transfected to express the green calcium indicator GCaMP6s. These cells were xenografted into the premotor cortex of mice, along with a virus expressing the red calcium indicator jRGECO1a under a neuron-specific synapsin promotor to allow dual-color imaging of neurons and glioma cells. The Inscopix mini-microscope system was implanted into the cortex to allow real-time live calcium imaging in freely behaving animals. Results: Several HGG cell lines effectively expressed the GCaMP6s calcium indicator. In vivo, we were successfully able to image both neurons and glioma cells simultaneously in freely behaving mice in real time. Conclusions: The Inscopix system has been modified for studying cancer cells for the first time. This technology will be used to study how pharmacological agents and neuronal experience shape neuron-glioma circuit dynamics, to develop new therapeutic strategies for HGG.
APA, Harvard, Vancouver, ISO, and other styles
44

Pal, Sangita, Adam Boynton, Ryan Johnston, Naomi Currimjee, Kenin Qian, Mehdi Touat, Charlotte Bellamy, et al. "EXTH-95. UNCOVERING THERAPEUTIC VULNERABILITIES IN MISMATCH REPAIR-DEFICIENT GLIOMAS." Neuro-Oncology 24, Supplement_7 (November 1, 2022): vii231—vii232. http://dx.doi.org/10.1093/neuonc/noac209.893.

Full text
Abstract:
Abstract Approximately 25% of recurrent gliomas exhibit hypermutation, which is most often acquired in chemotherapy sensitive gliomas post-treatment with standard of care alkylating agent temozolomide, Intriguingly, nearly all of these recurrent hypermutant tumors harbor deficiencies in mismatch repair (MMR) genes. Unlike other MMR-deficient cancers, hypermutant gliomas do not exhibit detectable microsatellite instability (MSI) at the population level, and do not share similar dependencies. Thus, strategies to therapeutically target MMR-deficient and hypermutant gliomas are urgently needed, and likely to impact many patients. To evaluate vulnerabilities associated with MMR-deficient gliomas, we have generated isogenic MMR-deficient models by systematic ablation of core MMR genes MSH2, MSH6, MLH1, and PMS2, in multiple patient-derived glioma cell lines using a robust all-in-one CRISPR-Cas9 system. We characterized these isogenic MMR-deficient glioma lines in comparison to respective MMR-proficient control line by performing differential gene expression analysis and subsequent gene set enrichment analyses. Our analyses reveal in an unbiased fashion an enrichment of several hallmark gene sets including cell cycle control and DNA repair in the MMR-deficient cell lines. We performed a CRISPR-Cas9 knockout screen in these MMR deficient and hypermutant models and identified candidate genes involved in DNA repair, cell cycle, RNA metabolism and other pathways as preferential dependencies in the MMR-deficient glioma cells. We also screened multiple MMR-deficient isogenic models against the high-throughput drug repurposing (REPO) chemical library and our preliminary results reveal a number of compounds from a variety of drug classes that selectively kill the MMR-deficient glioma cells, indicating that the loss of MMR confers differential dependencies to these small molecule inhibitors. The candidate drugs and genes identified from these high-throughput assays are currently being subjected to further validation, and they possibly will help us identify novel therapeutic strategies or improve existing therapeutic strategies to target recurrent MMR-deficient gliomas.
APA, Harvard, Vancouver, ISO, and other styles
45

Kashiwagi, Hideki, Yoshihide Hattori, Shinji Kawabata, Ryo Kayama, Kohei Yoshimura, Yusuke Fukuo, Takuya Kanemitsu, et al. "Multi-Targeted Neutron Capture Therapy Combined with an 18 kDa Translocator Protein-Targeted Boron Compound Is an Effective Strategy in a Rat Brain Tumor Model." Cancers 15, no. 4 (February 6, 2023): 1034. http://dx.doi.org/10.3390/cancers15041034.

Full text
Abstract:
Background: Boron neutron capture therapy (BNCT) has been adapted to high-grade gliomas (HG); however, some gliomas are refractory to BNCT using boronophenylalanine (BPA). In this study, the feasibility of BNCT targeting the 18 kDa translocator protein (TSPO) expressed in glioblastoma and surrounding environmental cells was investigated. Methods: Three rat glioma cell lines, an F98 rat glioma bearing brain tumor model, DPA-BSTPG which is a boron-10 compound targeting TSPO, BPA, and sodium borocaptate (BSH) were used. TSPO expression was evaluated in the F98 rat glioma model. Boron uptake was assessed in three rat glioma cell lines and in the F98 rat glioma model. In vitro and in vivo neutron irradiation experiments were performed. Results: DPA-BSTPG was efficiently taken up in vitro. The brain tumor has 16-fold higher TSPO expressions than its brain tissue. The compound biological effectiveness value of DPA-BSTPG was 8.43 to F98 rat glioma cells. The boron concentration in the tumor using DPA-BSTPG convection-enhanced delivery (CED) administration was approximately twice as high as using BPA intravenous administration. BNCT using DPA-BSTPG has significant efficacy over the untreated group. BNCT using a combination of BPA and DPA-BSTPG gained significantly longer survival times than using BPA alone. Conclusion: DPA-BSTPG in combination with BPA may provide the multi-targeted neutron capture therapy against HG.
APA, Harvard, Vancouver, ISO, and other styles
46

Chen, Zhong-ping, Qing-wu Wu, and Jing Wang. "Antitumor effect of levetiracetam combined with temozolomide in glioma cell lines." Journal of Clinical Oncology 35, no. 15_suppl (May 20, 2017): e13530-e13530. http://dx.doi.org/10.1200/jco.2017.35.15_suppl.e13530.

Full text
Abstract:
e13530 Background: The antitumor effect oftemozolomide (TMZ) is still remitted for portion of glioma patients, and if anti-epilepcy drug levetiracetam (LEV) may help TMZ for anti-glioma is not clear. Methods: The cell proliferation with Cell Counting Kit-8 (CCK-8) assay was performed in two O6-methylguanine DNA methyltransferase (MGMT) positive cell lines (T98G and U138) and two other MGMT negative cell lines (SKMG-4 and U87). The cells were treated with LEV and TMZ alone or in combination. Results: LEV inhibited the proliferation of glioma cell lines but not reach therapeutic effect, especially in U87cell line. The 50% inhibitory concentration (IC50) of TMZ was significantly decreased in cell lines (T98G, U138 and SKMG-4, P< 0.05) which were treated with TMZ and LEV, but no significantly changed in U87 (P> 0.05). Conclusions: LEV may inhibit the proliferation of glioma cell lines (both MGMT- positive and MGMT-negative) and also sensitizes cell lines (T98G, U138 and SKMG-4, but not U87) to TMZ.
APA, Harvard, Vancouver, ISO, and other styles
47

Wang, Weijun, Nian-Ling Zhu, Jason Chua, Steve Swenson, Fritz K. Costa, Stephanie Schmitmeier, Barbara A. Sosnowski, Toshiaki Shichinohe, Noriyuki Kasahara, and Thomas C. Chen. "Retargeting of adenoviral vector using basic fibroblast growth factor ligand for malignant glioma gene therapy." Journal of Neurosurgery 103, no. 6 (December 2005): 1058–66. http://dx.doi.org/10.3171/jns.2005.103.6.1058.

Full text
Abstract:
Object. Adenovirus vector (AdV)—mediated gene delivery has been recently demonstrated in clinical trials as a novel potential treatment for malignant gliomas. Combined coxsackievirus B and adenovirus receptor (CAR) has been shown to function as an attachment receptor for multiple adenovirus serotypes, whereas the vitronectin integrins (αvβ3 and αvβ5) are involved in AdV internalization. In resected glioma specimens, the authors demonstrated that malignant gliomas have varying levels of CAR, αvβ3, and αvβ5 expression. Methods. A correlation between CAR expression and the transduction efficiency of AdV carrying the green fluorescent protein in various human glioblastoma multiforme (GBM) cell lines and GBM primary cell lines was observed. To increase transgene activity in in vitro glioma cells with low or deficient levels of CAR, the authors used basic fibroblast growth factor (FGF2) as a targeting ligand to redirect adenoviral infection through its cognate receptor, FGF receptor 1 (FGFR1), which was expressed at high levels by all glioma cells. These findings were confirmed by in vivo study data demonstrating enhanced transduction efficiency of FGF2-retargeted AdV in CAR-negative intracranial gliomas compared with AdV alone, without evidence of increased angiogenesis. Conclusions. Altogether, the results demonstrated that AdV-mediated gene transfer using the FGF2/FGFR system is effective in gliomas with low or deficient levels of CAR and suggested that FGF2-retargeting of AdV may be a promising approach in glioma gene therapy.
APA, Harvard, Vancouver, ISO, and other styles
48

Wang, Tuo, Yan Zhang, Bo Cui, Maode Wang, Ya Li, and Ke Gao. "miR-4530 inhibits the malignant biological behaviors of human glioma cells by directly targeting RTEL1." Acta Biochimica et Biophysica Sinica 52, no. 12 (November 17, 2020): 1394–403. http://dx.doi.org/10.1093/abbs/gmaa126.

Full text
Abstract:
Abstract Human glioma is the most common primary brain tumor and is associated with high morbidity and mortality. Aberrant expressions of microRNAs (miRNAs) are involved in glioma progression. In the present study, we aimed to elucidate the roles of miR-4530 in the pathogenesis of gliomas. miR-4530 expression was examined in human glioma clinical tissues and cell lines including U251 and T98G. The target gene of miR-4530, RTEL1, was predicted with online tools and validated by luciferase reporter assay. Lentivirus infection, transfection of plasmids, and miRNA mimics were used to manipulate gene expression. Cell proliferation was determined using the CCK-8 method, and migration and invasion assays were determined with transwell experiments. Colony formation was measured by crystal violet staining, while apoptosis was determined by Annexin V/PI staining. The anti-tumor effects of miR-4530 were evaluated in nude mice xenografted using U251 cells. Our results showed that miR-4530 was significantly down-regulated in human glioma tissues and cell lines. miR-4530 over-expression inhibited the malignant behaviors of U251 and T98G cells, including reduced proliferation, diminished colony formation, migration and invasion, and increased apoptosis. Further mechanistic investigations revealed that RTEL1 is a direct functional target of miR-4530 in gliomas, and its over-expression remarkably reverses the effects of miR-4530 mimics on inhibiting these malignant behaviors. In addition, miR-4530 over-expression inhibited the growth of xenografted U251 glioma in nude mice. Therefore, miR-4530 acts as a tumor-suppressor gene and inhibits the malignant biological behaviors of human glioma cells, which is associated with directly targeting RTEL1. The miR-4530/RTEL1 axis is a potential therapeutic target for gliomas.
APA, Harvard, Vancouver, ISO, and other styles
49

Park, Jong-Whi, Felix Sahm, Bianca Steffl, Isabel Arrillaga-Romany, Daniel Cahill, Michelle Monje, Christel Herold-Mende, Wolfgang Wick, and Sevin Turcan. "CBIO-20. HIGH LEVELS OF TERT CONFER SENSITIVITY TO THE DNA HYPOMETHYLATING AGENT DECITABINE (DAC) IN GLIOMAS." Neuro-Oncology 23, Supplement_6 (November 2, 2021): vi31. http://dx.doi.org/10.1093/neuonc/noab196.120.

Full text
Abstract:
Abstract BACKGROUND Decitabine (DAC)-incorporated DNA binds DNMT1 enzyme and subsequently triggers DNMT1 degradation. Previously, we showed that DAC can mediate the anti-tumor effect in a preclinical model of IDH-mutant gliomas. Here, we further investigate molecular determinants of response to DAC in gliomas. METHODS DAC response was assessed by soft agar anchorage independent growth assays and cell proliferation measurements. Patient-derived IDH-mutant chromosome 1p/19q codeleted (codel) and non-codel glioma lines upon vehicle and DAC treatment were used for RNA sequencing and Gene Set Enrichment Analysis (GSEA). RESULTS We found that DAC treatment is effective in high TERT-expressing gliomas including IDH-mutant and IDH-wildtype glioma lines. In contrast, pharmacological inhibition of TERT reduces DAC response in glioma lines. Interestingly, transcriptomic profiling showed that DAC reduces the expression of TERT, along with increased CDKN1A/p21 expression. We experimentally validated that TERT expression depends on CDKN1A/p21. Furthermore, p53 is required for DAC-mediated CDKN1A/p21 induction. Importantly, DAC-mediated proliferation defects in TERT-proficient glioma cells are abolished by DNMT1 knockdown, indicative of an expected DAC mechanism. CONCLUSIONS DAC could elicit the pronounced anti-tumor response in IDH-mutant codel oligodendroglioma and IDH-wildtype glioblastoma with TERT activating mutations.
APA, Harvard, Vancouver, ISO, and other styles
50

Tamai, Sho, Sabit Hemragul, Jiapaer Shabierjiang, Guangtao Zhang, Jiakang Zhang, Yi Wang, Shingo Tanaka, Masashi Kinoshita, Atsushi Hirao, and Mitsutoshi Nakada. "CBMS-12 PENTAMIDINE; TRANSLATIONAL RESEARCH FOR A NEW CHEMOTHERAPY TARGETING ON GLIOMA CELLS AND GLIOMA STEM CELLS USING DRUG REPOSITIONING." Neuro-Oncology Advances 1, Supplement_2 (December 2019): ii6—ii7. http://dx.doi.org/10.1093/noajnl/vdz039.029.

Full text
Abstract:
Abstract INTRODUCTION Glioblastoma (GBM) is primary malignant brain tumor with poor prognosis. Despite aggressive chemoradiotherapies, GBM has resistance and finally relapses. Recently, it is revealed that glioma stem cells (GSCs) are forming tumors and induce the recurrence. However, there is no effective therapy for GSCs. Herein, we newly identified pentamidine, an antiprotozoal drug, is effective for not only glioma cells but also GSCs by using drug repositioning approach. METHOD We used two glioma cell lines, A172 and T98, and patient-derived glioma stem cell lines KGS01, KGS07 which were established at Kanazawa University. We investigated proliferation ability, stemness and intracellular signal change by proliferation assay, sphere forming assay and western blotting, respectively. RESULT Proliferation ability was prohibited by pentamidine in both glioma cell lines and GSC lines. The half maximal inhibitory concentrations were 5–10 μM in glioma cell lines and 1–5 μM in GSC lines. Sphere forming assay revealed that size and number of spheres were reduced in both GSC lines, depending on concentration of pentamidine. In all cell lines, phosphorylation of extracellular signal-related kinase (ERK) and signal transducer and activator of transcription 3 (STAT3) were suppressed by pentamidine. DISCUSSION Pentamidine is known as the therapeutic drug for pneumocystis jirovecii. In this study, pentamidine suppressed proliferation activity in all cell lines, and stemness in both GSCs. Previous papers revealed pentamidine had anti-tumor effects for some types of tumor cell lines, however, therapeutic effect for tumor stem cells have never been mentioned. CONCLUSION These results suggest that pentamidine would be therapeutic drug for not only glioma cells but also GSCs by suppressing phosphorylation of ERK and STAT3.
APA, Harvard, Vancouver, ISO, and other styles
We offer discounts on all premium plans for authors whose works are included in thematic literature selections. Contact us to get a unique promo code!

To the bibliography