Abstract 4145: Brain-derived endothelial cells stimulate migration of different human, mouse, and rat glioma cell lines in vivo and in vitro

Author(s):  
Viveka N. Yadav ◽  
Gregory J. Baker ◽  
Samanthi Narayanan ◽  
Maria G. Castro ◽  
Pedro R. Lowenstein
2019 ◽  
Vol 21 (Supplement_6) ◽  
pp. vi39-vi40
Author(s):  
Lubayna Elahi ◽  
Matthew Garrett ◽  
Lea Guo ◽  
Michael Condro ◽  
Riki Kawaguchi ◽  
...  

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.


1992 ◽  
Vol 28 (1) ◽  
pp. 47-56 ◽  
Author(s):  
Aziz Ghahary ◽  
R. Bhatnagar ◽  
Karen Price ◽  
Norine L. Forsyth ◽  
You Jun Shen ◽  
...  

1996 ◽  
Vol 84 (6) ◽  
pp. 1024-1032 ◽  
Author(s):  
Ian F. Pollack ◽  
Stephanie Kawecki ◽  
John S. Lazo

✓ Seven-hydroxystaurosporine (UCN-01) is a derivative of the nonselective protein kinase inhibitor staurosporine that exhibits significant selectivity for protein kinase C (PKC) in comparison to a variety of other intracellular kinases and appears to be well tolerated in vivo at concentrations sufficient to achieve effective inhibition of PKC. Because recent studies have indicated that the proliferation of malignant gliomas may result from activation of PKC-mediated pathways and, conversely, may be inhibited by blocking PKC, the authors examined the efficacy of this agent as an inhibitor of proliferation in three established and three low-passage malignant glioma cell lines in vitro. A striking inhibition of proliferation was produced by UCN-01 in each of the cell lines, with a median effective concentration of 20 to 100 nM, which correlated with the median in vitro PKC inhibitory concentration of 20 to 60 nM for this agent in the U-87 and SG-388 glioma cell lines. Inhibition-recovery studies of clonogenic activity indicated that UCN-01 had both cytostatic and cytotoxic effects on the treated cells. Proliferation resumed after short-term (6- and 24-hour) exposures to this agent; in contrast, with longer exposures, recovery of proliferative activity was severely compromised. In addition, UCN-01 enhanced the inhibition of glioma cell proliferation achieved with conventional chemotherapeutic agents, exhibiting synergistic effects with cisplatin and additive effects with 1,3-bis(2-chloroethyl)-1-nitrosourea. In vivo studies in which UCN-01 was administered by continuous intraperitoneal infusion in subcutaneous and intracranial intraparenchymal nude rat models demonstrated significant activity against U-87 glioma xenografts at dose levels that were well tolerated. It is concluded that UCN-01 is an effective agent for the inhibition of glioma proliferation in vitro and in vivo and has potential for clinical applicability in the treatment of human gliomas.


2019 ◽  
Vol 21 (Supplement_3) ◽  
pp. iii2-iii2
Author(s):  
T Weiss ◽  
H Meister ◽  
M Weller ◽  
C Sentman ◽  
P Roth

Abstract BACKGROUND Cancer immunotherapy with genetically engineered T cells that express a chimeric antigen receptor (CAR) has led to impressive responses in extracranial malignancies and is also explored against glioblastoma. However, CAR T cell strategies that are currently being explored against glioblastoma target single tumor antigens, which are non-homogeneously expressed and are prone to antigen escape. Furthermore, the immunosuppressive brain tumor microenvironment hampers anti-tumor efficacy. METHODS By immunohistochemistry and flow cytometry, we investigated the expression of CD155 and CD112, which are ligands to the activating immune cell receptor DNAX accessory molecule-1 (DNAM-1), in human and mouse glioma cell lines as well as in human glioblastoma samples. To understand their functional role, we generated CD155 or CD112 knock-out glioma cell lines using CRISPR/Cas9 and studied proliferation, sensitivity to irradiation or temozolomide as well as migration. To exploit the promiscuous binding features of DNAM-1, we generated different first or second-generation CAR T cells that use DNAM-1 as a tumor-binding domain. Subsequently, we investigated their anti-tumor activity in vitro in co-culture assays and in vivo in syngeneic orthotopic murine glioma models. RESULTS CD155 and CD112 are homogenously expressed in human and mouse glioma cell lines and human glioblastoma tissues. Knock-out of these ligands affected the migration of tumor cells, but did not affect proliferation or sensitivity to irradition or temozolomide. DNAM-1-based CAR T cells demonstrated high cytolytic activity and effector cytokine secretion in vitro. In vivo, DNAM-1 based CAR T cells reached to the tumor site in the brain upon intravenous administration, prolonged survival of orthotopic glioma-bearing mice and led to a durable anti-tumor response in a fraction of mice. The treatment was tolerated without toxicities. CONCLUSION We elucidated the tumor-intrinisic role of CD155 and CD112 and provide the first systematical preclincal assessment of DNAM-1 CAR T cells against glioma. These findings provide a rationale to test this immunotherapeutic strategy also in human glioma patients.


2021 ◽  
Author(s):  
Wenjin Qiu ◽  
Xiaomin Cai ◽  
Kaya Xu ◽  
Shibin Song ◽  
Zumu Xiao ◽  
...  

Abstract Background Regenerating liver phosphatase 1 (PRL1) is an established oncogene in various cancers, although its biological functions and the underlying mechanisms in glioblastoma multiforme (GBM) remain unclear. Methods PRL1 expression levels were analyzed in glioma tissues and cell lines. Multiple glioma cell lines were transfected with PRL1-overexpressing and shRNA constructs. In vitro proliferation, migration and invasion assays were conducted. Western blot and ubiquitylation assays were performed for molecular and mechanistic analyses. PRL1 expression levels were correlated with downstream ubiquitin pathway and clinical parameters using archival GBM samples. Results PRL1 was significantly upregulated in glioma tissues and cell lines, and positively correlated with the tumor grade. Ectopic expression of PRL1 in glioma cell lines significantly enhanced their tumorgenicity and invasion both in vitro and in vivo by promoting EMT. Conversely, knocking down PRL1 blocked EMT in the GBM cells, and inhibited their invasion, migration and tumorigenic growth. PRL1 also stabilized Snail2 through deubiquitination by activating USP36. Snail2 was identified as a crucial mediator of the oncogenic effects of PRL1 in GBM. Finally, PRL1 protein levels were positively correlated with that of Snail2 and predicted poor outcome of GBMs. Conclusions PRL1 promotes GBM progression by activating USP36-mediated Snail2 deubiquitination. This novel PRL1/USP36/Snail2 axis may be a promising therapeutic target for glioblastoma.


2020 ◽  
Vol 8 (1) ◽  
Author(s):  
Victor Ruiz-Rodado ◽  
Adrian Lita ◽  
Tyrone Dowdy ◽  
Orieta Celiku ◽  
Alejandra Cavazos Saldana ◽  
...  

Abstract Background Targeting glutamine metabolism in cancer has become an increasingly vibrant area of research. Mutant IDH1 (IDH1mut) gliomas are considered good candidates for targeting this pathway because of the contribution of glutamine to their newly acquired function: synthesis of 2-hydroxyglutarate (2HG). Methods We have employed a combination of 13C tracers including glutamine and glucose for investigating the metabolism of patient-derived IDH1mut glioma cell lines through NMR and LC/MS. Additionally, genetic loss-of-function (in vitro and in vivo) approaches were performed to unravel the adaptability of these cell lines to the inhibition of glutaminase activity. Results We report the adaptability of IDH1mut cells’ metabolism to the inhibition of glutamine/glutamate pathway. The glutaminase inhibitor CB839 generated a decrease in the production of the downstream metabolites of glutamate, including those involved in the TCA cycle and 2HG. However, this effect on metabolism was not extended to viability; rather, our patient-derived IDH1mut cell lines display a metabolic plasticity that allows them to overcome glutaminase inhibition. Conclusions Major metabolic adaptations involved pathways that can generate glutamate by using alternative substrates from glutamine, such as alanine or aspartate. Indeed, asparagine synthetase was upregulated both in vivo and in vitro revealing a new potential therapeutic target for a combinatory approach with CB839 against IDH1mut gliomas.


2011 ◽  
Vol 69 (3) ◽  
pp. 455-463 ◽  
Author(s):  
Linda B. C. Bralten ◽  
Nanne K. Kloosterhof ◽  
Rutger Balvers ◽  
Andrea Sacchetti ◽  
Lariesa Lapre ◽  
...  

2014 ◽  
Vol 121 (6) ◽  
pp. 1483-1491 ◽  
Author(s):  
Ho-Shin Gwak ◽  
Myung-Jin Park ◽  
In-Chul Park ◽  
Sang Hyeok Woo ◽  
Hyeon-Ok Jin ◽  
...  

Object Local invasiveness of malignant glioma is a major reason for the failure of current treatments including surgery and radiation therapy. Tetraarsenic oxide (As4O6 [TAO]) is a trivalent arsenic compound that has potential anticancer and antiangiogenic effects in selected cancer cell lines at a lower concentration than arsenic trioxide (As2O3 [ATO]), which has been more widely tested in vitro and in vivo. The authors tried to determine the cytotoxic concentration of TAO in malignant glioma cell lines and whether TAO would show anti-invasive effects under conditions independent of cell death or apoptosis. Methods The human phosphatase and tensin homolog (PTEN)-deficient malignant glioma cell lines U87MG, U251MG, and U373MG together with PTEN-functional LN428 were cultured with a range of micromolar concentrations of TAO. The invasiveness of the glioma cell lines was analyzed. The effect of TAO on matrix metalloproteinase (MMP) secretion and membrane type 1 (MT1)-MMP expression was measured using gelatin zymography and Western blot, respectively. Akt, or protein kinase B, activity, which is a downstream effector of PTEN, was assessed with a kinase assay using glycogen synthesis kinase-3β (GSK-3β) as a substrate and Western blotting of phosphorylated Akt. Results Tetraarsenic oxide inhibited 50% of glioma cell proliferation at 6.3–12.2 μM. Subsequent experiments were performed under the same TAO concentrations and exposure times, avoiding the direct tumoricidal effect of TAO, which was confirmed with apoptosis markers. An invasion assay revealed a dose-dependent decrease in invasiveness under the influence of TAO. Both the gelatinolytic activity of MMP-2 and MT1-MMP expression decreased in a dose-dependent manner in all cell lines, which was in accordance with the invasion assay results. The TAO decreased kinase activity of Akt on GSK-3β assay and inhibited Akt phosphorylation in a dose-dependent manner in all cell lines regardless of their PTEN status. Conclusions These results showed that TAO effectively inhibits proliferation of glioblastoma cell lines and also exerts an anti-invasive effect via decreased MMP-2 secretion, decreased MT1-MMP expression, and the inhibition of Akt phosphorylation under conditions devoid of cytotoxicity. Further investigations using an in vivo model are needed to evaluate the potential role of TAO as an anti-invasive agent.


2007 ◽  
Vol 85 (2) ◽  
pp. 123-132 ◽  
Author(s):  
Takashi Sasayama ◽  
Kazuhiro Tanaka ◽  
Katsu Mizukawa ◽  
Atsufumi Kawamura ◽  
Takeshi Kondoh ◽  
...  

2006 ◽  
Vol 14 (9) ◽  
pp. 614-622 ◽  
Author(s):  
Berta Sanchez De Juan ◽  
Hagen Von Briesen ◽  
Svetlana E. Gelperina ◽  
Jörg Kreuter

Sign in / Sign up

Export Citation Format

Share Document