Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Neurooncol Adv ; 5(1): vdad132, 2023.
Article in English | MEDLINE | ID: mdl-38130900

ABSTRACT

Background: Epidermal growth factor receptor (EGFR) amplification is found in nearly 40%-50% of glioblastoma cases. Several EGFR inhibitors have been tested in glioblastoma but have failed to demonstrate long-term therapeutic benefit, presumably because of acquired resistance. Targeting EGFR downstream signaling with mitogen-activated protein kinase kinase 1 and 2 (MEK1/2) inhibitors would be a more effective approach to glioblastoma treatment. We tested the therapeutic potential of MEK1/2 inhibitors in glioblastoma using 3D cultures of glioma stem-like cells (GSCs) and mouse models of glioblastoma. Methods: Several MEK inhibitors were screened in an unbiased high-throughput platform using GSCs. Cell death was evaluated using flow cytometry and Western blotting (WB) analysis. RNA-seq, real-time quantitative polymerase chain reaction, immunofluorescence, and WB analysis were used to identify and validate neuronal differentiation. Results: Unbiased screening of multiple MEK inhibitors in GSCs showed antiproliferative and apoptotic cell death in sensitive cell lines. An RNA-seq analysis of cells treated with trametinib, a potent MEK inhibitor, revealed upregulation of neurogenesis and neuronal differentiation genes, such as achaete-scute homolog 1 (ASCL1), delta-like 3 (DLL3), and neurogenic differentiation 4 (NeuroD4). We validated the neuronal differentiation phenotypes in vitro and in vivo using selected differentiation markers (ß-III-tubulin, ASCL1, DLL3, and NeuroD4). Oral treatment with trametinib in an orthotopic GSC xenograft model significantly improved animal survival, with 25%-30% of mice being long-term survivors. Conclusions: Our findings demonstrated that MEK1/2 inhibition promotes neuronal differentiation in glioblastoma, a potential additional mechanism of action of MEK1/2 inhibitors. Thus, MEK inhibitors could be efficacious in glioblastoma patients with activated EGFR/MAPK signaling.

2.
Cancers (Basel) ; 13(21)2021 Oct 21.
Article in English | MEDLINE | ID: mdl-34771447

ABSTRACT

Interferon (IFN) signaling contributes to stemness, cell proliferation, cell death, and cytokine signaling in cancer and immune cells; however, the role of IFN signaling in glioblastoma (GBM) and GBM stem-like cells (GSCs) is unclear. Here, we investigated the role of cancer-cell-intrinsic IFN signaling in tumorigenesis in GBM. We report here that GSCs and GBM tumors exhibited differential cell-intrinsic type I and type II IFN signaling, and high IFN/STAT1 signaling was associated with mesenchymal phenotype and poor survival outcomes. In addition, chronic inhibition of IFN/STAT1 signaling decreased cell proliferation and mesenchymal signatures in GSCs with intrinsically high IFN/STAT1 signaling. IFN-ß exposure induced apoptosis in GSCs with intrinsically high IFN/STAT1 signaling, and this effect was abolished by the pharmacological inhibitor ruxolitinib and STAT1 knockdown. We provide evidence for targeting IFN signaling in a specific sub-group of GBM patients. IFN-ß may be a promising candidate for adjuvant GBM therapy.

3.
Neurooncol Adv ; 3(1): vdab015, 2021.
Article in English | MEDLINE | ID: mdl-33738447

ABSTRACT

Glioblastoma (GBM), the most aggressive primary brain tumor, has a dismal prognosis. Despite our growing knowledge of genomic and epigenomic alterations in GBM, standard therapies and outcomes have not changed significantly in the past two decades. There is therefore an urgent unmet need to develop novel therapies for GBM. The inter- and intratumoral heterogeneity of GBM, inadequate drug concentrations in the tumor owing to the blood-brain barrier, redundant signaling pathways contributing to resistance to conventional therapies, and an immunosuppressive tumor microenvironment, have all hindered the development of novel therapies for GBM. Given the high frequency of DNA damage pathway alterations in GBM, researchers have focused their efforts on pharmacologically targeting key enzymes, including poly(ADP-ribose) polymerase (PARP), DNA-dependent protein kinase, ataxia telangiectasia-mutated, and ataxia telangiectasia and Rad3-related. The mainstays of GBM treatment, ionizing radiation and alkylating chemotherapy, generate DNA damage that is repaired through the upregulation and activation of DNA damage response (DDR) enzymes. Therefore, the use of PARP and other DDR inhibitors to render GBM cells more vulnerable to conventional treatments is an area of intense investigation. In this review, we highlight the growing body of data behind DDR inhibitors in GBM, with a focus on putative predictive biomarkers of response. We also discuss the challenges involved in the successful development of DDR inhibitors for GBM, including the intracranial location and predicted overlapping toxicities of DDR agents with current standards of care, and propose promising strategies to overcome these hurdles.

4.
Am J Cancer Res ; 10(11): 3765-3783, 2020.
Article in English | MEDLINE | ID: mdl-33294266

ABSTRACT

The Cdc2-like kinases (CLKs) regulate RNA splicing and have been shown to suppress cell growth. Knockdown of CLK2 was found to block glioma stem-like cell (GSC) growth in vivo through the AKT/FOXO3a/p27 pathway without activating mTOR and MAPK signaling, suggesting that these pathways mediate resistance to CLK2 inhibition. We identified CLK2 binding partners using immunoprecipitation assays and confirmed their interactions in vitro in GSCs. We then tested the cellular viability of several signaling inhibitors in parental and CLK2 knockdown GSCs. Our results demonstrate that CLK2 binds to 14-3-3τ isoform and prevents its ubiquitination in GSCs. Stable CLK2 knockdown increased PP2A activity and activated PI3K signaling. Treatment with a PI3K/mTOR inhibitor in CLK2 knockdown cells led to a modest reduction in cell viability compared to drug treatment alone at a lower dose. However, FGFR inhibitor in CLK2 knockdown cells led to a decrease in cell viability and increased apoptosis. Reduced expression of CLK2 in glioblastoma, in combination with FGFR inhibitors, led to synergistic apoptosis induction and cell cycle arrest compared to blockade or either kinase alone.

5.
Int J Mol Sci ; 21(6)2020 Mar 13.
Article in English | MEDLINE | ID: mdl-32182988

ABSTRACT

Recent efforts in brain tumor research have been directed towards the modulation of the immune system for therapeutic interventions. Several human cancers, including gliomas, are infiltrated with immune cell types-including neutrophils and myeloid-derived suppressor cells-that contribute to tumor progression, invasiveness, and treatment resistance. The role of tumor-associated neutrophils and myeloid-derived suppressor cells in cancer biology remains elusive, as these cells can exert a multitude of pro-tumor and antitumor effects. In this review, we provide the current understanding and novel insights on the role of neutrophils and myeloid-derived suppressor cells in glioma progression and treatment resistance, as well as the mechanisms of pleiotropic behaviors in these cells during disease progression, with an emphasis on possible strategies to reprogram these cells towards their antitumor actions.


Subject(s)
Brain Neoplasms/immunology , Drug Resistance, Neoplasm , Glioma/immunology , Myeloid-Derived Suppressor Cells/immunology , Neutrophils/immunology , Animals , Brain Neoplasms/drug therapy , Brain Neoplasms/radiotherapy , Glioma/drug therapy , Glioma/radiotherapy , Humans , Radiation Tolerance
6.
Nat Cancer ; 1: 423-436, 2020 04.
Article in English | MEDLINE | ID: mdl-33521652

ABSTRACT

The natural history of small cell lung cancer (SCLC) includes rapid evolution from chemosensitivity to chemoresistance, although mechanisms underlying this evolution remain obscure due to scarcity of post-relapse tissue samples. We generated circulating tumor cell (CTC)-derived xenografts (CDXs) from SCLC patients to study intratumoral heterogeneity (ITH) via single-cell RNAseq of chemo-sensitive and -resistant CDXs and patient CTCs. We found globally increased ITH including heterogeneous expression of therapeutic targets and potential resistance pathways, such as EMT, between cellular subpopulations following treatment-resistance. Similarly, serial profiling of patient CTCs directly from blood confirmed increased ITH post-relapse. These data suggest that treatment-resistance in SCLC is characterized by coexisting subpopulations of cells with heterogeneous gene expression leading to multiple, concurrent resistance mechanisms. These findings emphasize the need for clinical efforts to focus on rational combination therapies for treatment-naïve SCLC tumors to maximize initial responses and counteract the emergence of ITH and diverse resistance mechanisms.


Subject(s)
Lung Neoplasms , Neoplastic Cells, Circulating , Small Cell Lung Carcinoma , Humans , Lung Neoplasms/drug therapy , Neoplasm Recurrence, Local/drug therapy , Neoplastic Cells, Circulating/metabolism , Single-Cell Analysis , Small Cell Lung Carcinoma/drug therapy
7.
Neuro Oncol ; 22(2): 169-170, 2020 02 20.
Article in English | MEDLINE | ID: mdl-31863119
8.
Oncotarget ; 9(12): 10497-10509, 2018 Feb 13.
Article in English | MEDLINE | ID: mdl-29535822

ABSTRACT

BACKGROUND: Despite the availability of hundreds of cancer drugs, there is insufficient data on the efficacy of these drugs on the extremely heterogeneous tumor cell populations of glioblastoma (GBM). RESULTS: The PKIS of 357 compounds was initially evaluated in 15 different GSC lines which then led to a more focused screening of the 21 most highly active compounds in 11 unique GSC lines using HTS screening for cell viability. We further validated the HTS result with the second-generation PLK1 inhibitor volasertib as a single agent and in combination with ionizing radiation (IR). In vitro studies showed that volasertib inhibited cell viability, and high levels of the anti-apoptotic protein Bcl-xL expression were highly correlated with volasertib resistance. Volasertib sensitized GSCs to radiation therapy by enhancing G2/M arrest and by inducing apoptosis. Colony-formation assay demonstrated that volasertib plus IR synergistically inhibited colony formation. In intracranial xenograft mouse models, the combination of volasertib and radiation significantly inhibited GSC tumor growth and prolonged median survival compared with radiation treatment alone due to inhibition of cell proliferation, enhancement of DNA damage, and induction of apoptosis. CONCLUSIONS: Our results reinforce the potential therapeutic efficacy of volasertib in combination with radiation for the treatment of GBM. METHODS: We used high-throughput screening (HTS) to identify drugs, out of 357 compounds in the published Protein Kinase Inhibitor Set, with the greatest efficacy against a panel of glioma stem cells (GSCs), which are representative of the classic cancer genome atlas (TCGA) molecular subtypes.

9.
Oncotarget ; 8(57): 96970-96983, 2017 Nov 14.
Article in English | MEDLINE | ID: mdl-29228586

ABSTRACT

Intercellular cell adhesion molecule 1 (ICAM-1; also known as CD54) is overexpressed in bevacizumab-resistant glioblastoma. In the present study, we tested our hypothesis that highly expressed ICAM-1 mediates glioblastoma's resistance to antiangiogenic therapy. We validated ICAM-1 overexpression in tumors resistant to antiangiogenic therapy using real-time polymerase chain reaction, immunohistochemistry, and Western blotting. We also detected ICAM1 expression in most glioma stem cells (GSCs). We investigated the mechanism of ICAM-1 overexpression after bevacizumab treatment and found that ICAM-1 protein expression was markedly increased in a time-dependent manner in GSC11 and GSC17 cells under hypoxic conditions in vitro. We also found that hypoxia induced ICAM-1 overexpression through the up-regulation of phosphorylated signal transducer and activator of transcription (p-STAT3). Hypoxia-induced p-STAT3 increased the mRNA transcription of ICAM-1, which we could inhibit with the STAT3 inhibitor AZD1480. Next, we used GFP-tagged ICAM-1 shRNA lentivirus to knock down ICAM-1 in GSC11 and GSC17 glioma cell lines. Then, we injected shICAM-1 GSC11 and scramble glioma stem cells into the brains of nude mice. Mice bearing tumors from shICAM-1 GSC11 cells survived significantly longer than mice injected with control cells did. The tumor sizes was significantly decreased in mice bearing tumors from shICAM-1 cells than that in mice bearing tumors from GFP-tagged GSC11 control cells. Knocking down ICAM-1 suppressed tumor invasion in vitro and in vivo and inhibited macrophage infiltration to the tumor site in bevacizumab-treated mice. Our findings suggest that ICAM-1 is a potentially important mediator of tumor migration and invasion in bevacizumab-resistant glioblastoma. Targeting ICAM-1 may provide a new strategy for enhancing the efficacy of antiangiogenic therapy against glioblastoma and preventing the invasive phenotype of the disease.

10.
Cell Rep ; 21(8): 2183-2197, 2017 Nov 21.
Article in English | MEDLINE | ID: mdl-29166609

ABSTRACT

A mesenchymal transition occurs both during the natural evolution of glioblastoma (GBM) and in response to therapy. Here, we report that the adhesion G-protein-coupled receptor, GPR56/ADGRG1, inhibits GBM mesenchymal differentiation and radioresistance. GPR56 is enriched in proneural and classical GBMs and is lost during their transition toward a mesenchymal subtype. GPR56 loss of function promotes mesenchymal differentiation and radioresistance of glioma initiating cells both in vitro and in vivo. Accordingly, a low GPR56-associated signature is prognostic of a poor outcome in GBM patients even within non-G-CIMP GBMs. Mechanistically, we reveal GPR56 as an inhibitor of the nuclear factor kappa B (NF-κB) signaling pathway, thereby providing the rationale by which this receptor prevents mesenchymal differentiation and radioresistance. A pan-cancer analysis suggests that GPR56 might be an inhibitor of the mesenchymal transition across multiple tumor types beyond GBM.


Subject(s)
Brain Neoplasms/metabolism , Glioblastoma/metabolism , Neoplastic Stem Cells/metabolism , Receptors, G-Protein-Coupled/metabolism , Cell Differentiation/physiology , Cell Line, Tumor , Humans , NF-kappa B/metabolism , Signal Transduction/physiology
11.
Neuro Oncol ; 19(9): 1152-1153, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28821207
12.
J Bone Miner Metab ; 35(1): 21-30, 2017 Jan.
Article in English | MEDLINE | ID: mdl-26747612

ABSTRACT

New developments in stem cell biology offer alternatives for the reconstruction of critical-sized bone defects. One of these developments is the use of induced pluripotent stem (iPS) cells. These stem cells are similar to embryonic stem (ES) cells, but can be generated from adult somatic cells and therefore do not raise ethical concerns. Proper characterization of iPS-derived osteoblasts is important for future development of safe clinical applications of these cells. For this reason, we differentiated mouse ES and iPS cells toward osteoblasts using osteogenic medium and compared their functionality. Immunocytochemical analysis showed significant expression of bone markers (osteocalcin and collagen type I) in osteoblasts differentiated from ES and iPS cells on days 7 and 30. An in vitro mineralization assay confirmed the functionality of osteogenically differentiated ES and iPS cells. Gene expression arrays focusing on osteogenic differentiation were performed in order to compare the gene expression pattern in both differentiated and undifferentiated ES cells and iPS cells. We observed a significant upregulation of osteogenesis-related genes such as Runx2, osteopontin, collagen type I, Tnfsf11, Csf1, and alkaline phosphatase upon osteogenic differentiation of the ES and iPS cells. We further validated the expression of key osteogenic genes Runx2, osteopontin, osteocalcin, collagen type I, and osterix in both differentiated and undifferentiated ES and iPS cells by means of quantified real-time polymerase chain reaction. We conclude that ES and iPS cells are similar in their osteogenic differentiation capacities, as well as in their gene expression patterns.


Subject(s)
Cell Differentiation , Induced Pluripotent Stem Cells/metabolism , Mouse Embryonic Stem Cells/metabolism , Osteoblasts/metabolism , Osteogenesis , Animals , Antigens, Differentiation/biosynthesis , Cell Line , Immunohistochemistry , Induced Pluripotent Stem Cells/cytology , Mice , Mouse Embryonic Stem Cells/cytology , Osteoblasts/cytology , Up-Regulation
13.
Oncotarget ; 6(31): 31007-17, 2015 Oct 13.
Article in English | MEDLINE | ID: mdl-26307681

ABSTRACT

Differentiation has been proposed as a therapeutic strategy for glioblastoma (GBM) in part due to observations of stem-like cells in GBM that have been shown to undergo terminal differentiation in response to growth factor withdrawal and BMP activation. However, the effects of long term exposure to serum culture conditions on glioma sphere cultures/glioma stem-like cells (GSCs) have not been examined. Here we show that GSCs retained both neurosphere formation and tumor initiation abilities after short or long term serum exposure. Under these conditions, GSCs expressed both neural lineage and stem cell markers, highlighting the aberrant pseudo-differentiation state. GSCs maintained under adherent serum cultured conditions continued to proliferate and initiate tumor formation with efficiencies similar to GSCs maintained under proliferating (neurosphere) conditions. Proneural (PN) GSCs under serum exposure showed an induction of mesenchymal (MES) gene expression signatures. Our data indicate that exposure to serum containing media result in aberrant differentiation (e.g. toward MES lineage) and activation of alternative oncogenic pathways in GSCs.


Subject(s)
Brain Neoplasms/pathology , Cell Differentiation , Cell Transformation, Neoplastic/pathology , Glioma/pathology , Mesoderm/pathology , Neoplastic Stem Cells/pathology , Animals , Apoptosis , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Gene Expression Regulation, Neoplastic , Glioma/genetics , Glioma/metabolism , Humans , Mesoderm/metabolism , Mice , Mice, Nude , Neoplastic Stem Cells/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
14.
Mol Cell Neurosci ; 65: 21-30, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25662290

ABSTRACT

The present study describes for the first time the neural expression and distribution of UGS148, a protein encoded by the RIKEN cDNA63330403K07 gene that has been shown to be prominently and characteristically expressed in neural stem cells (NSCs). Based on its molecular structure, UGS148 is an intracellular protein expected to be involved in intracellular sorting, trafficking, exocytosis and membrane insertion of proteins. We demonstrate that UGS148 is highly expressed in embryonic NSCs as well as, albeit at low level, in the adult neurogenic niches, the subventricular zone and the hippocampal dentate gyrus. Interestingly, the highest expression level of UGS148 in the adult mouse brain was observed specifically in the neurogenic cells lining the third ventricle, the tanycytes. Our in vitro studies show the involvement of UGS148 in the regulation of the proliferation of NSCs.


Subject(s)
Dentate Gyrus/metabolism , Ependymoglial Cells/metabolism , Nerve Tissue Proteins/genetics , Neural Stem Cells/metabolism , Amino Acid Sequence , Animals , Cells, Cultured , Dentate Gyrus/cytology , Dentate Gyrus/growth & development , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Nerve Tissue Proteins/metabolism , Protein Transport
15.
Cancer Lett ; 359(1): 107-16, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25592037

ABSTRACT

Glioblastoma (GBM) is the most common brain tumor in adults and the mesenchymal GBM subtype was reported to be the most malignant, presenting severe hypoxia and necrosis. Here, we investigated the possible role of a hypoxic microenvironment for inducing a mesenchymal and invasive phenotype. The exposure of non-mesenchymal SNB75 and U87 cells to hypoxia induced a strong change in cell morphology that was accompanied by enhanced invasive capacity and the acquisition of mesenchymal marker expression. Further analyses showed the induction of HIF1α and HIF2α by hypoxia and exposure to digoxin, a cardiac glycoside known to inhibit HIF1/2 expression, was able to prevent hypoxia-induced mesenchymal transition. ShRNA-mediated knockdown of HIF1α, and not HIF2α, prevented this transition, as well as the knockdown of the EMT transcription factor ZEB1. We provide further evidence for a hypoxia-induced mesenchymal shift in GBM primary material by showing co-localization of GLUT1, ZEB1 and the mesenchymal marker YKL40 in hypoxic regions of the tumor. Collectively, our results identify a HIF1α-ZEB1 signaling axis that promotes hypoxia induced mesenchymal shift and invasion in GBM in a cell line dependent fashion.


Subject(s)
Brain Neoplasms/metabolism , Cell Movement , Epithelial-Mesenchymal Transition , Glioblastoma/metabolism , Homeodomain Proteins/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Transcription Factors/metabolism , Adipokines/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Hypoxia , Cell Line, Tumor , Cell Movement/drug effects , Cell Shape , Chitinase-3-Like Protein 1 , Digoxin/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Glioblastoma/genetics , Glioblastoma/pathology , Glucose Transporter Type 1/metabolism , Homeodomain Proteins/genetics , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Lectins/metabolism , Necrosis , Neoplasm Invasiveness , Phenotype , RNA Interference , Signal Transduction , Transcription Factors/genetics , Transfection , Zinc Finger E-box-Binding Homeobox 1
16.
PLoS One ; 9(12): e115687, 2014.
Article in English | MEDLINE | ID: mdl-25546404

ABSTRACT

Molecular signatures in Glioblastoma (GBM) have been described that correlate with clinical outcome and response to therapy. The Proneural (PN) and Mesenchymal (MES) signatures have been identified most consistently, but others including Classical (CLAS) have also been reported. The molecular signatures have been detected by array techniques at RNA and DNA level, but these methods are costly and cannot take into account individual contributions of different cells within a tumor. Therefore, the aim of this study was to investigate whether subclasses of newly diagnosed GBMs could be assessed and assigned by application of standard pathology laboratory procedures. 123 newly diagnosed GBMs were analyzed for the tumor cell expression of 23 pre-identified proteins and EGFR amplification, together allowing for the subclassification of 65% of the tumors. Immunohistochemistry (IHC)-based profiling was found to be analogous to transcription-based profiling using a 9-gene transcriptional signature for PN and MES subclasses. Based on these data a novel, minimal IHC-based scheme for subclass assignment for GBMs is proposed. Positive staining for IDH1R132H can be used for PN subclass assignment, high EGFR expression for the CLAS subtype and a combined high expression of PTEN, VIM and/or YKL40 for the MES subclass. The application of the proposed scheme was evaluated in an independent tumor set, which resulted in similar subclass assignment rates as those observed in the training set. The IHC-based subclassification scheme proposed in this study therefore could provide very useful in future studies for stratification of individual patient samples.


Subject(s)
Biomarkers, Tumor/metabolism , Gene Expression Profiling/methods , Glioblastoma/metabolism , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , ErbB Receptors/genetics , ErbB Receptors/metabolism , Female , Glioblastoma/classification , Glioblastoma/pathology , Humans , Immunohistochemistry/methods , Male , Middle Aged , Transcriptome
17.
Cancer Cell ; 24(3): 331-46, 2013 Sep 09.
Article in English | MEDLINE | ID: mdl-23993863

ABSTRACT

Despite extensive study, few therapeutic targets have been identified for glioblastoma (GBM). Here we show that patient-derived glioma sphere cultures (GSCs) that resemble either the proneural (PN) or mesenchymal (MES) transcriptomal subtypes differ significantly in their biological characteristics. Moreover, we found that a subset of the PN GSCs undergoes differentiation to a MES state in a TNF-α/NF-κB-dependent manner with an associated enrichment of CD44 subpopulations and radioresistant phenotypes. We present data to suggest that the tumor microenvironment cell types such as macrophages/microglia may play an integral role in this process. We further show that the MES signature, CD44 expression, and NF-κB activation correlate with poor radiation response and shorter survival in patients with GBM.


Subject(s)
Glioblastoma/genetics , Glioblastoma/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Radiation Tolerance/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/genetics , Cluster Analysis , DNA Methylation , Disease Models, Animal , Epigenesis, Genetic , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Glioblastoma/mortality , Humans , Hyaluronan Receptors/genetics , Hyaluronan Receptors/metabolism , Mice , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Oligodendrocyte Transcription Factor 2 , Prognosis , Signal Transduction , Transcriptome , Tumor Necrosis Factor-alpha/pharmacology , Xenograft Model Antitumor Assays
18.
Biochem Pharmacol ; 85(4): 478-85, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23159669

ABSTRACT

Transforming growth factor-ß (TGF-ß) is a cytokine with a key role in tissue homeostasis and cancer. TGF-ß elicits both tumor suppressive and tumor promoting functions during cancer progression, in a wide range of cancers. Here, we review the tumor promoting function of TGF-ß and its possible promise as a therapeutic target in high grade gliomas, including glioblastoma multiforme (GBM), a disease with very poor prognosis. TGF-ß signaling is highly active in high grade gliomas and elevated TGF-ß activity has been associated with poor clinical outcome in this deadly disease. Common features of GBMs include fast cell proliferation, invasion into normal brain parenchyma, hypoxia, high angiogenic - and immunosuppressive activity, characteristics that all have been linked to activation of the TGF-ß pathway. TGF-ß signaling has also been connected with the cancer stem cell (CSC) phenotype in GBM. CSCs represent a subset of GBM cells thought to be responsible for tumor initiation, progression and relapse of disease. Following the description of these different properties of TGF-ß signaling and the underlying mechanisms identified thus far, the promise of TGF-ß targeted therapy in malignant gliomas is discussed. Several drugs targeting TGF-ß signaling have been developed that showed potent antitumor activity in preclinical models. A number of agents are currently evaluated in early clinical studies in glioma patients. Available results of these studies are highlighted and a perspective on the promise of TGF-ß-targeted therapy is given.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/drug therapy , Glioma/drug therapy , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/metabolism , Antineoplastic Agents/therapeutic use , Brain Neoplasms/metabolism , Humans , Signal Transduction/physiology
19.
J Neurooncol ; 107(3): 487-501, 2012 May.
Article in English | MEDLINE | ID: mdl-22249692

ABSTRACT

Glioma stem-like cells (GSCs) may be the initiating cells in glioblastoma (GBM) and contribute to the resistance of these tumors to conventional therapies. Development of novel chemotherapeutic agents and treatment approaches against GBM, especially those specifically targeting GSCs are thus necessary. In the present study, we found that a novel Janus kinase 2/Signal Transducer and Activator of Transcription 3 (JAK2/STAT3) pathway inhibitor (WP1193) significantly decreased the proliferation of established glioma cell lines in vitro and inhibit the growth of glioma in vivo. To test the efficacy of WP1193 against GSCs, we then administrated WP1193 to GSCs isolated and expanded from multiple human GBM tumors. We revealed that WP1193 suppressed phosphorylation of JAK2 and STAT3 with high potency and demonstrated a dose-dependent inhibition of proliferation and neurosphere formation of GSCs. These effects were at least due in part to G1 arrest associated with down-regulation of cyclin D1 and up-regulation of p21( Cip1/Waf-1 ). Furthermore, WP1193 exposure decreased expression of stem cell markers including CD133 and c-myc, and induced cell death in GSCs through apoptosis. Taken together, our data indicate that WP1193 is a potent small molecule inhibitor of the JAK2/STAT3 pathway that shows promise as a therapeutic agent against GBM by targeting GSCs.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cyanoacrylates/pharmacology , Glioblastoma/drug therapy , Neoplastic Stem Cells/drug effects , Pyridines/pharmacology , Animals , Blotting, Western , Flow Cytometry , Glioblastoma/metabolism , Humans , Janus Kinase 2/metabolism , Male , Mice , Mice, Nude , Neoplastic Stem Cells/metabolism , Reverse Transcriptase Polymerase Chain Reaction , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
20.
Genes Dev ; 25(24): 2594-609, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-22190458

ABSTRACT

Recent molecular classification of glioblastoma (GBM) has shown that patients with a mesenchymal (MES) gene expression signature exhibit poor overall survival and treatment resistance. Using regulatory network analysis of available expression microarray data sets of GBM, including The Cancer Genome Atlas (TCGA), we identified the transcriptional coactivator with PDZ-binding motif (TAZ), to be highly associated with the MES network. TAZ expression was lower in proneural (PN) GBMs and lower-grade gliomas, which correlated with CpG island hypermethylation of the TAZ promoter compared with MES GBMs. Silencing of TAZ in MES glioma stem cells (GSCs) decreased expression of MES markers, invasion, self-renewal, and tumor formation. Conversely, overexpression of TAZ in PN GSCs as well as murine neural stem cells (NSCs) induced MES marker expression and aberrant osteoblastic and chondrocytic differentiation in a TEAD-dependent fashion. Using chromatin immunoprecipitation (ChIP), we show that TAZ is directly recruited to a majority of MES gene promoters in a complex with TEAD2. The coexpression of TAZ, but not a mutated form of TAZ that lacks TEAD binding, with platelet-derived growth factor-B (PDGF-B) resulted in high-grade tumors with MES features in a murine model of glioma. Our studies uncover a direct role for TAZ and TEAD in driving the MES differentiation of malignant glioma.


Subject(s)
Brain Neoplasms/physiopathology , Glioma/physiopathology , Mesenchymal Stem Cells/cytology , Neoplastic Stem Cells/cytology , Transcription Factors/metabolism , Acyltransferases , Animals , Cell Line, Tumor , Cells, Cultured , DNA-Binding Proteins/metabolism , Epigenomics , Gene Expression Regulation, Neoplastic , Humans , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, SCID , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , TEA Domain Transcription Factors , Transcription Factors/genetics , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...