Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Int J Mol Sci ; 24(8)2023 Apr 16.
Article in English | MEDLINE | ID: mdl-37108511

ABSTRACT

Multiple metabolic pathways are utilized to maintain cellular homeostasis. Given the evidence that altered cell metabolism significantly contributes to glioma biology, the current research efforts aim to improve our understanding of metabolic rewiring between glioma's complex genotype and tissue context. In addition, extensive molecular profiling has revealed activated oncogenes and inactivated tumor suppressors that directly or indirectly impact the cellular metabolism that is associated with the pathogenesis of gliomas. The mutation status of isocitrate dehydrogenases (IDHs) is one of the most important prognostic factors in adult-type diffuse gliomas. This review presents an overview of the metabolic alterations in IDH-mutant gliomas and IDH-wildtype glioblastoma (GBM). A particular focus is placed on targeting metabolic vulnerabilities to identify new therapeutic strategies for glioma.


Subject(s)
Brain Neoplasms , Glioblastoma , Glioma , Adult , Humans , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Glioma/genetics , Glioma/pathology , Mutation , Genotype , Isocitrate Dehydrogenase/genetics
2.
Int J Mol Sci ; 24(19)2023 Sep 30.
Article in English | MEDLINE | ID: mdl-37834227

ABSTRACT

Glioblastoma (GBM) is the most lethal brain cancer, causing inevitable deaths of patients owing to frequent relapses of cancer stem cells (CSCs). The significance of the NOTCH signaling pathway in CSCs has been well recognized; however, there is no NOTCH-selective treatment applicable to patients with GBM. We recently reported that Jagged1 (JAG1), a NOTCH ligand, drives a NOTCH receptor-independent signaling pathway via JAG1 intracellular domain (JICD1) as a crucial signal that renders CSC properties. Therefore, mechanisms regulating the JICD1 signaling pathway should be elucidated to further develop a selective therapeutic regimen. Here, we identified annexin A2 (ANXA2) as an essential modulator to stabilize intrinsically disordered JICD1. The binding of ANXA2 to JICD1 prevents the proteasomal degradation of JICD1 by heat shock protein-70/90 and carboxy-terminus of Hsc70 interacting protein E3 ligase. Furthermore, JICD1-driven propagation and tumor aggressiveness were inhibited by ANXA2 knockdown. Taken together, our findings show that ANXA2 maintains the function of the NOTCH receptor-independent JICD1 signaling pathway by stabilizing JICD1, and the targeted suppression of JICD1-driven CSC properties can be achieved by blocking its interaction with ANXA2.


Subject(s)
Annexin A2 , Glioblastoma , Humans , Annexin A2/genetics , Annexin A2/metabolism , Cell Line, Tumor , Glioblastoma/metabolism , Jagged-1 Protein/genetics , Jagged-1 Protein/metabolism , Neoplasm Recurrence, Local , Receptors, Notch/metabolism
3.
Sci Rep ; 14(1): 1566, 2024 Jan 18.
Article in English | MEDLINE | ID: mdl-38238447

ABSTRACT

In this study, an intense pulsed light (IPL) annealing process for a printed multi-layered indium-gallium-zinc-oxide (IGZO) and silver (Ag) electrode structure was developed for a high performance all-printed inorganic thin film transistor (TFT). Through a solution process using IGZO precursor and Ag ink, the bottom gate structure TFT was fabricated. The spin coating method was used to form the IGZO semiconductor layer on a heavily-doped silicon wafer covered with thermally grown silicon dioxide. The annealing process of the IGZO layer utilized an optimized IPL irradiation process. The Ag inks were printed on the IGZO layer by screen printing to form the source and drain (S/D) pattern. This S/D pattern was dried by near infrared radiation (NIR) and the dried S/D pattern was sintered with intense pulsed light by varying the irradiation energy. The performances of the all-printed TFT such as the field effect mobility and on-off ratio electrical transfer properties were measured by a parameter analyzer. The interfacial analysis including the contact resistance and cross-sectional microstructure analysis is essential because diffusion phenomenon can occur during the annealing and sintering process. Consequently, this TFT device showed noteworthy performance (field effect mobility: 7.96 cm2/V s, on/off ratio: 107). This is similar performance compared to a conventional TFT, which is expected to open a new path in the printed metal oxide-based TFT field.

4.
Acta Neuropathol Commun ; 11(1): 13, 2023 01 16.
Article in English | MEDLINE | ID: mdl-36647117

ABSTRACT

Capicua (CIC) is an important downstream molecule of RTK/RAS/MAPK pathway. The regulatory mechanism of CIC underlying tumorigenesis in oligodendroglioma, where CIC is frequently mutated, has yet to be fully elucidated. Using patient-derived glioma lines, RNA-sequencing and bioinformatic analysis of publicly available databases, we investigated how CIC loss- or gain-of-function regulates its downstream targets, cell proliferation and glutamate release. Our results indicate an increased frequency of CIC truncating mutations in oligodendroglioma during progression. In vitro, CIC modulation had a modest effect on cell proliferation in glioma lines, and no significant changes in the expression of ETV1, ETV4 and ETV5. Transcriptional repression of known CIC targets was observed in gliomas expressing non-phosphorylatable CIC variant on Ser173 which was unable to interact with 14-3-3. These data outline a mechanism by which the repressor function of CIC is inhibited by 14-3-3 in gliomas. Using transcriptional profiling, we found that genes related to glutamate release were upregulated because of CIC depletion. In addition, loss of CIC leads to increased extracellular glutamate. Consistent with this, CIC restoration in an oligodendroglioma line reduced the levels of extracellular glutamate, neuronal toxicity and xCT/SLC7A11 expression. Our findings may provide a molecular basis for the prevention of glioma-associated seizures.


Subject(s)
Amino Acid Transport System y+ , Glioma , Oligodendroglioma , Repressor Proteins , Humans , Amino Acid Transport System y+/metabolism , Glioma/genetics , Glutamic Acid , Neurons/metabolism , Oligodendroglioma/genetics , Repressor Proteins/genetics
5.
Cell Death Dis ; 14(12): 822, 2023 12 13.
Article in English | MEDLINE | ID: mdl-38092725

ABSTRACT

Jagged1 (JAG1) is a Notch ligand that correlates with tumor progression. Not limited to its function as a ligand, JAG1 can be cleaved, and its intracellular domain translocates to the nucleus, where it functions as a transcriptional cofactor. Previously, we showed that JAG1 intracellular domain (JICD1) forms a protein complex with DDX17/SMAD3/TGIF2. However, the molecular mechanisms underlying JICD1-mediated tumor aggressiveness remains unclear. Here, we demonstrate that JICD1 enhances the invasive phenotypes of glioblastoma cells by transcriptionally activating epithelial-to-mesenchymal transition (EMT)-related genes, especially TWIST1. The inhibition of TWIST1 reduced JICD1-driven tumor aggressiveness. Although SMAD3 is an important component of transforming growth factor (TGF)-ß signaling, the JICD1/SMAD3 transcriptional complex was shown to govern brain tumor invasion independent of TGF-ß signaling. Moreover, JICD1-TWIST1-MMP2 and MMP9 axes were significantly correlated with clinical outcome of glioblastoma patients. Collectively, we identified the JICD1/SMAD3-TWIST1 axis as a novel inducer of invasive phenotypes in cancer cells.


Subject(s)
Glioblastoma , Humans , Cell Line, Tumor , Epithelial-Mesenchymal Transition/genetics , Glioblastoma/genetics , Homeodomain Proteins/metabolism , Ligands , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , Signal Transduction , Smad3 Protein/genetics , Smad3 Protein/metabolism , Transforming Growth Factor beta/metabolism , Twist-Related Protein 1/genetics , Twist-Related Protein 1/metabolism
6.
Neuro Oncol ; 24(11): 1911-1924, 2022 11 02.
Article in English | MEDLINE | ID: mdl-35468210

ABSTRACT

BACKGROUND: Glioblastoma (GBM) is an aggressive tumor that frequently exhibits gain of chromosome 7, loss of chromosome 10, and aberrantly activated receptor tyrosine kinase signaling pathways. Previously, we identified Mesenchyme Homeobox 2 (MEOX2), a gene located on chromosome 7, as an upregulated transcription factor in GBM. Overexpressed transcription factors can be involved in driving GBM. Here, we aimed to address the role of MEOX2 in GBM. METHODS: Patient-derived GBM tumorspheres were used to constitutively knockdown or overexpress MEOX2 and subjected to in vitro assays including western blot to assess ERK phosphorylation. Cerebral organoid models were used to investigate the role of MEOX2 in growth initiation. Intracranial mouse implantation models were used to assess the tumorigenic potential of MEOX2. RNA-sequencing, ACT-seq, and CUT&Tag were used to identify MEOX2 target genes. RESULTS: MEOX2 enhanced ERK signaling through a feed-forward mechanism. We identified Ser155 as a putative ERK-dependent phosphorylation site upstream of the homeobox-domain of MEOX2. S155A substitution had a major effect on MEOX2 protein levels and altered its subnuclear localization. MEOX2 overexpression cooperated with p53 and PTEN loss in cerebral organoid models of human malignant gliomas to induce cell proliferation. Using high-throughput genomics, we identified putative transcriptional target genes of MEOX2 in patient-derived GBM tumorsphere models and a fresh frozen GBM tumor. CONCLUSIONS: We identified MEOX2 as an oncogenic transcription regulator in GBM. MEOX2 increases proliferation in cerebral organoid models of GBM and feeds into ERK signaling that represents a core signaling pathway in GBM.


Subject(s)
Glioblastoma , Glioma , Mice , Animals , Humans , Genes, Homeobox , Homeodomain Proteins/genetics , Glioma/genetics , Glioblastoma/pathology , Cell Proliferation , Transcription Factors/genetics , Carcinogenesis/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic
7.
Neuro Oncol ; 23(1): 76-87, 2021 01 30.
Article in English | MEDLINE | ID: mdl-32882013

ABSTRACT

BACKGROUND: Decitabine (DAC) is an FDA-approved DNA methyltransferase (DNMT) inhibitor that is used in the treatment of patients with myelodysplastic syndromes. Previously, we showed that DAC marks antitumor activity against gliomas with isocitrate dehydrogenase 1 (IDH1) mutations. Based on promising preclinical results, a clinical trial has been launched to determine the effect of DAC in IDH-mutant gliomas. The next step is to comprehensively assess the efficacy and potential determinants of response to DAC in malignant gliomas. METHODS: The expression and activity of telomerase reverse transcriptase (TERT) and DNMT1 were manipulated in patient-derived IDH1-mutant and -wildtype glioma lines, followed by assessment of cell proliferation with DAC treatment alone or in combination with telomerase inhibitors. RNA sequencing, Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment, and correlation analysis were performed. RESULTS: IDH1-mutant glioma tumorspheres with hemizygous codeletion of chromosome arms 1p/19q were particularly sensitive to DAC and showed significant inhibition of DNA replication genes. Our transcriptome analysis revealed that DAC induced expression of cyclin-dependent kinase inhibitor 1A/p21 (CDKN1A), along with downregulation of TERT. These molecular changes were also observed following doxorubicin treatment, supporting the importance of DAC-induced DNA damage in contributing to this effect. We demonstrated that knockdown of p21 led to TERT upregulation. Strikingly, TERT overexpression increased DNMT1 levels and DAC sensitivity via a telomerase-independent mechanism. Furthermore, RNA inhibition (RNAi) targeting of DNMT1 abrogated DAC response in TERT-proficient glioma cells. CONCLUSIONS: DAC downregulates TERT through p21 induction. Our data point to TERT and DNMT1 levels as potential determinants of response to DAC treatment.


Subject(s)
Brain Neoplasms , Glioma , Telomerase , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Decitabine/pharmacology , Glioma/drug therapy , Glioma/genetics , Humans , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , Mutation , Promoter Regions, Genetic , Telomerase/genetics
8.
Cancers (Basel) ; 11(10)2019 Oct 22.
Article in English | MEDLINE | ID: mdl-31652645

ABSTRACT

Targeting the epigenome has been considered a compelling treatment modality for several cancers, including gliomas. Nearly 80% of the lower-grade gliomas and secondary glioblastomas harbor recurrent mutations in isocitrate dehydrogenase (IDH). Mutant IDH generates high levels of 2-hydroxyglutarate (2-HG) that inhibit various components of the epigenetic machinery, including histone and DNA demethylases. The encouraging results from current epigenetic therapies in hematological malignancies have reinvigorated the interest in solid tumors and gliomas, both preclinically and clinically. Here, we summarize the recent advancements in epigenetic therapy for lower-grade gliomas and discuss the challenges associated with current treatment options. A particular focus is placed on therapeutic mechanisms underlying favorable outcome with epigenetic-based drugs in basic and translational research of gliomas. This review also highlights emerging bridges to combination treatment with respect to epigenetic drugs. Given that epigenetic therapies, particularly DNA methylation inhibitors, increase tumor immunogenicity and antitumor immune responses, appropriate drug combinations with immune checkpoint inhibitors may lead to improvement of treatment effectiveness of immunotherapy, ultimately leading to tumor cell eradication.

9.
Front Mol Neurosci ; 12: 73, 2019.
Article in English | MEDLINE | ID: mdl-30983969

ABSTRACT

Sprouty proteins act ubiquitously as signaling integrators and inhibitors of receptor tyrosine kinase (RTK) activated pathways. Among the four Sprouty isoforms, Sprouty2 is a key regulator of growth factor signaling in several neurological disorders. High protein levels correlate with reduced survival of glioma patients. We recently demonstrated that abrogating its function inhibits tumor growth by overstimulation of ERK and induction of DNA replication stress. The important role of Sprouty2 in the proliferation of malignant glioma cells prompted us to investigate its subcellular localization applying super-resolution fluorescence and immunoelectron microscopy. We found that cytoplasmic Sprouty2 is not homogenously distributed but localized to small spots (<100 nm) partly attached to vimentin filaments and co-localized with activated ERK. The protein is associated with early, late and recycling endosomes in response to but also independently of growth factor stimulation. The subcellular localization of Sprouty2 in all areas exhibiting strong RTK activities may reflect a protective response of glioma cells to limit excessive ERK activation and to prevent cellular senescence and apoptosis.

10.
Acta Neuropathol Commun ; 7(1): 201, 2019 12 05.
Article in English | MEDLINE | ID: mdl-31806013

ABSTRACT

The presence of genome-wide DNA hypermethylation is a hallmark of lower grade gliomas (LGG) with isocitrate dehydrogenase (IDH) mutations. Further molecular classification of IDH mutant gliomas is defined by the presence (IDHmut-codel) or absence (IDHmut-noncodel) of hemizygous codeletion of chromosome arms 1p and 19q. Despite the DNA hypermethylation seen in bulk tumors, intra-tumoral heterogeneity at the epigenetic level has not been thoroughly analyzed. To address this question, we performed the first epigenetic profiling of single cells in a cohort of 5 gliomas with IDH1 mutation using single nucleus Assay for Transposase-Accessible Chromatin with high-throughput sequencing (snATAC-seq). Using the Fluidigm HT IFC microfluidics platform, we generated chromatin accessibility maps from 336 individual nuclei, and identified variable promoter accessibility of non-coding RNAs in LGGs. Interestingly, local chromatin structures of several non-coding RNAs are significant factors that contribute to heterogeneity, and show increased promoter accessibility in IDHmut-noncodel samples. As an example for clinical significance of this result, we identify CYTOR as a poor prognosis factor in gliomas with IDH mutation. Open chromatin assay points to differential accessibility of non-coding RNAs as an important source of epigenetic heterogeneity within individual tumors and between molecular subgroups. Rare populations of nuclei that resemble either IDH mutant molecular group co-exist within IDHmut-noncodel and IDHmut-codel groups, and along with non-coding RNAs may be an important issue to consider for future studies, as they may help guide predict treatment response and relapse.A web-based explorer for the data is available at shiny.turcanlab.org.


Subject(s)
Brain Neoplasms/genetics , Cell Nucleus/genetics , Chromatin/genetics , Epigenesis, Genetic/genetics , Glioma/genetics , Isocitrate Dehydrogenase/genetics , Brain Neoplasms/pathology , Cell Nucleus/pathology , Chromatin/pathology , Cohort Studies , Glioma/pathology , Humans , Mutation/genetics , Sequence Analysis, RNA/methods
11.
Biochem Biophys Res Commun ; 365(3): 496-502, 2008 Jan 18.
Article in English | MEDLINE | ID: mdl-18021740

ABSTRACT

Here, we show that H-ras(V12) causes the p53-knockout mouse astrocytes (p53-/- astrocytes) to be transformed into brain cancer stem-like cells. H-ras(V12) triggers the p53-/- astrocytes to express a Nestin and a Cd133, which are expressed in normal and cancer neural stem cells. H-ras(V12) also induces the formation of a single cell-derived neurosphere under neural stem cell culture conditions. Furthermore, H-ras(V12)-overexpressing p53-/- astrocytes (p53-/-ast-H-ras(V12)) possess an in vitro self-renewal capacity, and are aberrantly differentiated into Tuj1-positve neurons both in vitro and in vivo. Amongst a variety of Ras-mediated canonical signaling pathways, we demonstrated that the MEK/ERK signaling pathway is responsible for neurosphere formation in p53-deficient astrocytes, whereas the PI3K/AKT signaling pathway is involved in oncogenic transformation in these cells. These findings suggest that the activation of Ras signaling pathways promotes the generation of brain cancer stem-like cells from p53-deficient mouse astrocytes by changing cell fate and transforming cell properties.


Subject(s)
Astrocytes/pathology , Brain Neoplasms/etiology , Brain Neoplasms/pathology , Cell Transformation, Neoplastic/pathology , Neoplastic Stem Cells/pathology , Oncogene Protein p21(ras)/metabolism , Animals , Astrocytes/metabolism , Biomarkers/analysis , Brain Neoplasms/metabolism , Cell Transformation, Neoplastic/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Intermediate Filament Proteins/analysis , Intermediate Filament Proteins/metabolism , MAP Kinase Kinase Kinases/metabolism , Mice , Mice, Knockout , Neoplastic Stem Cells/chemistry , Neoplastic Stem Cells/metabolism , Nerve Tissue Proteins/analysis , Nerve Tissue Proteins/metabolism , Nestin , Neurons/chemistry , Neurons/pathology , Oncogene Protein p21(ras)/analysis , Oncogene Protein p21(ras)/genetics , Phosphatidylinositol 3-Kinases/metabolism , Tubulin/analysis , Tumor Suppressor Protein p53/genetics
12.
Mol Cancer Ther ; 6(8): 2178-87, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17699715

ABSTRACT

We have established several glioma-relevant oncogene-engineered cancer cells to reevaluate the oncogene-selective cytotoxicity of previously well-characterized anticancer drugs, such as etoposide, doxorubicin, staurosporine, and carmustine. Among several glioma-relevant oncogenes (activated epidermal growth factor receptor, Ras, and Akt, as well as Bcl-2 and p53DD used in the present study), the activated epidermal growth factor receptor, Ras, and Akt exerted oncogenic transformation of Ink4a/Arf(-/-) murine astrocyte cells. We identified that etoposide, a topoisomerase II inhibitor, caused selective killing of myristylated Akt (Akt-myr)-transduced Ink4a/Arf(-/-) astrocytes and U87MG cells in a dose- and time-dependent manner. Etoposide-selective cytotoxicity in the Akt-myr-transduced cells was shown to be caused by nonapoptotic cell death and occurred in a p53-independent manner. Etoposide caused severe reactive oxygen species (ROS) accumulation preferentially in the Akt-myr-transduced cells, and elevated ROS rendered these cells highly sensitive to cell death. The etoposide-selective cell death of Akt-myr-transduced cells was attenuated by pepstatin A, a lysosomal protease inhibitor. In the present study, we show that etoposide might possess a novel therapeutic activity for oncogenic Akt-transduced cancer cells to kill preferentially through ROS-mediated damage.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/pathology , Cell Transformation, Neoplastic/drug effects , Etoposide/pharmacology , Oncogene Protein v-akt/metabolism , Reactive Oxygen Species/metabolism , Transduction, Genetic , Animals , Astrocytes/drug effects , Cell Death/drug effects , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Dose-Response Relationship, Drug , Mice , Mice, Nude , Oncogene Protein v-akt/genetics , Pepstatins/pharmacology , Time Factors , Tumor Suppressor Protein p53/metabolism
13.
Neuro Oncol ; 20(8): 1044-1054, 2018 07 05.
Article in English | MEDLINE | ID: mdl-29635363

ABSTRACT

Background: Sprouty2 (SPRY2), a feedback regulator of receptor tyrosine kinase (RTK) signaling, has been shown to be associated with drug resistance and cell proliferation in glioblastoma (GBM), but the underlying mechanisms are still poorly defined. Methods: SPRY2 expression and survival patterns of patients with gliomas were analyzed using publicly available databases. Effects of RNA interference targeting SPRY2 on cellular proliferation in established GBM or patient-derived GBM stemlike cells were examined. Loss- or gain-of-function of SPRY2 to regulate the tumorigenic capacity was assessed in both intracranial and subcutaneous xenografts. Results: SPRY2 was found to be upregulated in GBM, which correlated with reduced survival in GBM patients. SPRY2 knockdown significantly impaired proliferation of GBM cells but not of normal astrocytes. Silencing of SPRY2 increased epidermal growth factor-induced extracellular signal-regulated kinase (ERK) and Akt activation causing premature onset of DNA replication, increased DNA damage, and impaired proliferation, suggesting that SPRY2 suppresses DNA replication stress. Abrogating SPRY2 function strongly inhibited intracranial tumor growth and led to significantly prolonged survival of U87 xenograft-bearing mice. In contrast, SPRY2 overexpression promoted tumor propagation of low-tumorigenic U251 cells. Conclusions: The present study highlights an antitumoral effect of SPRY2 inhibition that is based on excessive activation of ERK signaling and DNA damage response, resulting in reduced cell proliferation and increased cytotoxicity, proposing SPRY2 as a promising pharmacological target in GBM patients.


Subject(s)
Brain Neoplasms/pathology , Carcinogenesis/pathology , Gene Expression Regulation, Neoplastic , Glioblastoma/pathology , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Animals , Apoptosis , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Carcinogenesis/genetics , Carcinogenesis/metabolism , Cell Proliferation , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Glioblastoma/genetics , Glioblastoma/metabolism , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mice , Prognosis , RNA, Small Interfering/genetics , Signal Transduction , Survival Rate , Tumor Cells, Cultured , Up-Regulation , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL