Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 33
2.
Cell Rep ; 28(8): 2064-2079.e11, 2019 08 20.
Article En | MEDLINE | ID: mdl-31433983

Identifying cellular programs that drive cancers to be stem-like and treatment resistant is critical to improving outcomes in patients. Here, we demonstrate that constitutive extracellular signal-regulated kinase 1/2 (ERK1/2) activation sustains a stem-like state in glioblastoma (GBM), the most common primary malignant brain tumor. Pharmacological inhibition of ERK1/2 activation restores neurogenesis during murine astrocytoma formation, inducing neuronal differentiation in tumorspheres. Constitutive ERK1/2 activation globally regulates miRNA expression in murine and human GBMs, while neuronal differentiation of GBM tumorspheres following the inhibition of ERK1/2 activation requires the functional expression of miR-124 and the depletion of its target gene SOX9. Overexpression of miR124 depletes SOX9 in vivo and promotes a stem-like-to-neuronal transition, with reduced tumorigenicity and increased radiation sensitivity. Providing a rationale for reports demonstrating miR-124-induced abrogation of GBM aggressiveness, we conclude that reversal of an ERK1/2-miR-124-SOX9 axis induces a neuronal phenotype and that enforcing neuronal differentiation represents a therapeutic strategy to improve outcomes in GBM.


Brain Neoplasms/pathology , Cell Differentiation , Glioblastoma/pathology , MAP Kinase Signaling System , MicroRNAs/metabolism , Neurons/pathology , SOX9 Transcription Factor/metabolism , Animals , Astrocytoma/genetics , Astrocytoma/pathology , Benzamides/pharmacology , Brain Neoplasms/genetics , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Diphenylamine/analogs & derivatives , Diphenylamine/pharmacology , Disease Progression , Female , Glioblastoma/genetics , Humans , MAP Kinase Signaling System/drug effects , Male , Mice, Nude , MicroRNAs/genetics , Neoplasm Invasiveness , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Neurogenesis/drug effects , Neurons/drug effects , Neurons/metabolism , Phenotype , Protein Kinase Inhibitors/pharmacology , Radiation Tolerance/drug effects
3.
Exp Cell Res ; 378(1): 76-86, 2019 05 01.
Article En | MEDLINE | ID: mdl-30844389

Slow-cycling and treatment-resistant cancer cells escape therapy, providing a rationale for regrowth and recurrence in patients. Much interest has focused on identifying the properties of slow-cycling tumor cells in glioblastoma (GBM), the most common and lethal primary brain tumor. Despite aggressive ionizing radiation (IR) and treatment with the alkylating agent temozolomide (TMZ), GBM patients invariably relapse and ultimately succumb to the disease. In patient biopsies, we demonstrated that GBM cells expressing the proliferation markers Ki67 and MCM2 displayed a larger cell volume compared to rare slow-cycling tumor cells. In optimized density gradients, we isolated a minor fraction of slow-cycling GBM cells in patient biopsies and tumorsphere cultures. Transcriptional profiling, self-renewal, and tumorigenicity assays reflected the slow-cycling state of high-density GBM cells (HDGCs) compared to the tumor bulk of low-density GBM cells (LDGCs). Slow-cycling HDGCs enriched for stem cell antigens proliferated a few days after isolation to generate LDGCs. Both in vitro and in vivo, we demonstrated that HDGCs show increased treatment-resistance to IR and TMZ treatment compared to LDGCs. In conclusion, density gradients represent a non-marker based approach to isolate slow-cycling and treatment-resistant GBM cells across GBM subgroups.


Brain Neoplasms/pathology , Cell Self Renewal , Glioblastoma/pathology , Neoplastic Stem Cells/pathology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Brain Neoplasms/drug therapy , Brain Neoplasms/radiotherapy , Cell Proliferation , Drug Resistance, Neoplasm , Glioblastoma/drug therapy , Glioblastoma/radiotherapy , Humans , Ki-67 Antigen/genetics , Ki-67 Antigen/metabolism , Mice , Mice, Nude , Minichromosome Maintenance Complex Component 2/genetics , Minichromosome Maintenance Complex Component 2/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Radiation Tolerance , Temozolomide/pharmacology , Temozolomide/therapeutic use , Transcriptome , Tumor Cells, Cultured
4.
Neuron ; 100(4): 763-765, 2018 11 21.
Article En | MEDLINE | ID: mdl-30465758

Mechanical cues regulate neuronal function and reactivity of glial cells, the origin of gliomas. In this issue of Neuron, Chen et al. (2018) uncover a feedforward loop mediated by the mechanosensitive ion channel Piezo1 and tissue stiffness that drives glioma aggression.


Aggression , Glioma , Brain , Humans , Ion Channels , Neurons
5.
Cell Death Dis ; 9(10): 1010, 2018 09 27.
Article En | MEDLINE | ID: mdl-30262908

The weak immunogenicity of gliomas presents a barrier for effective immunotherapy. Na/H exchanger isoform 1 (NHE1) maintains alkaline intracellular pH (pHi) of glioma cells and acidic microenvironment. In addition, NHE1 is expressed in tumor-associated microglia and tumor-associated macrophages (TAMs) and involved in protumoral communications between glioma and TAMs. Therefore, we hypothesize that NHE1 plays a role in developing tumor resistance and immunosuppressive tumor microenvironment. In this study, we investigated the efficacy of pharmacological inhibition of NHE1 on combinatorial therapies. Here we show that temozolomide (TMZ) treatment stimulates NHE1 protein expression in two intracranial syngeneic mouse glioma models (SB28, GL26). Pharmacological inhibition of NHE1 potentiated the cytotoxic effects of TMZ, leading to reduced tumor growth and increased median survival of mice. Blockade of NHE1 stimulated proinflammatory activation of TAM and increased cytotoxic T cell infiltration into tumors. Combining TMZ, anti-PD-1 antibody treatment with NHE1 blockade significantly prolonged the median survival in the mouse glioma model. These results demonstrate that pharmacological inhibition of NHE1 protein presents a new strategy for potentiating TMZ-induced cytotoxicity and increasing tumor immunogenicity for immunotherapy to improve glioma therapy.


Glioma/drug therapy , Glioma/metabolism , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/metabolism , Sodium-Hydrogen Exchanger 1/metabolism , Temozolomide/pharmacology , Animals , Antibodies/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Female , Immunotherapy/methods , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Microglia/drug effects , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/metabolism , Tumor Microenvironment/drug effects
6.
Transl Oncol ; 11(4): 941-949, 2018 Aug.
Article En | MEDLINE | ID: mdl-29883968

The goal of this research was to elucidate the relationship between WHO 2016 molecular classifications of newly diagnosed, nonenhancing lower grade gliomas (LrGG), tissue sample histopathology, and magnetic resonance (MR) parameters derived from diffusion, perfusion, and 1H spectroscopic imaging from the tissue sample locations and the entire tumor. A total of 135 patients were scanned prior to initial surgery, with tumor cellularity scores obtained from 88 image-guided tissue samples. MR parameters were obtained from corresponding sample locations, and histograms of normalized MR parameters within the T2 fluid-attenuated inversion recovery lesion were analyzed in order to evaluate differences between subgroups. For tissue samples, higher tumor scores were related to increased normalized apparent diffusion coefficient (nADC), lower fractional anisotropy (nFA), lower cerebral blood volume (nCBV), higher choline (nCho), and lower N-acetylaspartate (nNAA). Within the T2 lesion, higher tumor grade was associated with higher nADC, lower nFA, and higher Cho to NAA index. Pathological analysis confirmed that diffusion and metabolic parameters increased and perfusion decreased with tumor cellularity. This information can be used to select targets for tissue sampling and to aid in making decisions about treating residual disease.

7.
Cancer Cell ; 33(5): 874-889.e7, 2018 05 14.
Article En | MEDLINE | ID: mdl-29681511

Gliomas comprise heterogeneous malignant glial and stromal cells. While blood vessel co-option is a potential mechanism to escape anti-angiogenic therapy, the relevance of glial phenotype in this process is unclear. We show that Olig2+ oligodendrocyte precursor-like glioma cells invade by single-cell vessel co-option and preserve the blood-brain barrier (BBB). Conversely, Olig2-negative glioma cells form dense perivascular collections and promote angiogenesis and BBB breakdown, leading to innate immune cell activation. Experimentally, Olig2 promotes Wnt7b expression, a finding that correlates in human glioma profiling. Targeted Wnt7a/7b deletion or pharmacologic Wnt inhibition blocks Olig2+ glioma single-cell vessel co-option and enhances responses to temozolomide. Finally, Olig2 and Wnt7 become upregulated after anti-VEGF treatment in preclinical models and patients. Thus, glial-encoded pathways regulate distinct glioma-vascular microenvironmental interactions.


Brain Neoplasms/blood supply , Glioma/blood supply , Oligodendrocyte Transcription Factor 2/metabolism , Oligodendroglia/microbiology , Wnt Proteins/metabolism , Animals , Bevacizumab/pharmacology , Blood-Brain Barrier/metabolism , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Glioma/drug therapy , Glioma/metabolism , Humans , Mice , Neoplasm Transplantation , Oligodendrocyte Transcription Factor 2/genetics , Temozolomide/pharmacology , Tumor Cells, Cultured , Tumor Microenvironment , Wnt Proteins/genetics , Wnt Signaling Pathway/drug effects
8.
Mol Cancer Res ; 16(5): 777-790, 2018 05.
Article En | MEDLINE | ID: mdl-29431617

Interstitial fluid pressure (IFP) presents a barrier to drug uptake in solid tumors, including the aggressive primary brain tumor glioblastoma (GBM). It remains unclear how fluid dynamics impacts tumor progression and can be targeted therapeutically. To address this issue, a novel telemetry-based approach was developed to measure changes in IFP during progression of GBM xenografts. Antisecretory factor (AF) is an endogenous protein that displays antisecretory effects in animals and patients. Here, endogenous induction of AF protein or exogenous administration of AF peptide reduced IFP and increased drug uptake in GBM xenografts. AF inhibited cell volume regulation of GBM cells, an effect that was phenocopied in vitro by the sodium-potassium-chloride cotransporter 1 (SLC12A2/NKCC1) inhibitor bumetanide. As a result, AF induced apoptosis and increased survival in GBM models. In vitro, the ability of AF to reduce GBM cell proliferation was phenocopied by bumetanide and NKCC1 knockdown. Next, AF's ability to sensitize GBM cells to the alkylating agent temozolomide, standard of care in GBM patients, was evaluated. Importantly, combination of AF induction and temozolomide treatment blocked regrowth in GBM xenografts. Thus, AF-mediated inhibition of cell volume regulation represents a novel strategy to increase drug uptake and improve outcome in GBM. Mol Cancer Res; 16(5); 777-90. ©2018 AACR.


Glioblastoma/therapy , Animals , Cell Line, Tumor , Cell Proliferation , Cell Size , Disease Progression , Glioblastoma/pathology , Humans , Mice , Mice, Nude
9.
Mol Cancer Res ; 15(11): 1623-1633, 2017 11.
Article En | MEDLINE | ID: mdl-28778876

Glioblastoma (GBM) is the most common primary malignant brain tumor of adults and confers a poor prognosis due, in part, to diffuse invasion of tumor cells. Heparan sulfate (HS) glycosaminoglycans, present on the cell surface and in the extracellular matrix, regulate cell signaling pathways and cell-microenvironment interactions. In GBM, the expression of HS glycosaminoglycans and the enzymes that regulate their function are altered, but the actual HS content and structure are unknown. However, inhibition of HS glycosaminoglycan function is emerging as a promising therapeutic strategy for some cancers. In this study, we use liquid chromatography-mass spectrometry analysis to demonstrate differences in HS disaccharide content and structure across four patient-derived tumorsphere lines (GBM1, 5, 6, 43) and between two murine tumorsphere lines derived from murine GBM with enrichment of mesenchymal and proneural gene expression (mMES and mPN, respectively) markers. In GBM, the heterogeneous HS content and structure across patient-derived tumorsphere lines suggested diverse functions in the GBM tumor microenvironment. In GBM5 and mPN, elevated expression of sulfatase 2 (SULF2), an extracellular enzyme that alters ligand binding to HS, was associated with low trisulfated HS disaccharides, a substrate of SULF2. In contrast, other primary tumorsphere lines had elevated expression of the HS-modifying enzyme heparanase (HPSE). Using gene editing strategies to inhibit HPSE, a role for HPSE in promoting tumor cell adhesion and invasion was identified. These studies characterize the heterogeneity in HS glycosaminoglycan content and structure across GBM and reveal their role in tumor cell invasion.Implications: HS-interacting factors promote GBM invasion and are potential therapeutic targets. Mol Cancer Res; 15(11); 1623-33. ©2017 AACR.


Brain Neoplasms/metabolism , Glioblastoma/metabolism , Heparitin Sulfate/chemistry , Heparitin Sulfate/metabolism , Animals , Brain Neoplasms/chemistry , Cell Line, Tumor , Chromatography, Liquid , Gene Editing , Glioblastoma/chemistry , Glucuronidase/genetics , Humans , Mass Spectrometry , Mice , Neoplasm Invasiveness , Neoplasm Transplantation , Signal Transduction , Sulfatases , Sulfotransferases/metabolism , Tumor Microenvironment
10.
Radiat Res ; 188(4): 443-454, 2017 10.
Article En | MEDLINE | ID: mdl-28777696

Cranial radiation severely affects brain health and function, including glial cell production and myelination. Recent studies indicate that voluntary exercise has beneficial effects on oligodendrogenesis and myelination. Here, we hypothesized that voluntary running would increase oligodendrocyte numbers in the corpus callosum after irradiation of the juvenile mouse brain. The brains of C57Bl/6J male mice were 6 Gy irradiated on postnatal day 9 during the main gliogenic developmental phase, resulting in a loss of oligodendrocyte precursor cells. Upon adulthood, the mice were injected with bromodeoxyuridine and allowed to exercise on a running wheel for four weeks. Cell proliferation and survival, Ascl1+ oligodendrocyte precursor and Olig2+ oligodendrocyte cell numbers as well as CC1+ mature oligodendrocytes were quantified using immunohistology. Radiation induced a reduction in the number of Olig2+ oligodendrocytes by nearly 50% without affecting production or survival of new Olig2+ cells. Ascl1+ cells earlier in the oligodendroglial cell lineage were also profoundly affected, with numbers reduced by half. By three weeks of age, Olig2+ cell numbers had not recovered, and this was paralleled by a volumetric loss in the corpus callosum. The deficiency of Olig2+ oligodendrocytes persisted into adulthood. Additionally, the depletion of Ascl1+ progenitor cells was irreversible, and was even more pronounced at 12 weeks postirradiation compared to day 2 postirradiation. Furthermore, the overall number of CC1+ mature oligodendrocytes decreased by 28%. The depletion of Olig2+ cells in irradiated animals was reversed by 4 weeks of voluntary exercise. Moreover, voluntary exercise also increased the number of Ascl1+ progenitor cells in irradiated animals. Taken together, these results demonstrate that exercise in adulthood significantly ameliorates the profound and long-lasting effects of moderate exposure to immature oligodendrocytes during postnatal development.


Brain/cytology , Brain/radiation effects , Oligodendroglia/cytology , Physical Conditioning, Animal/physiology , Animals , Cell Count , Cell Proliferation/radiation effects , Corpus Callosum/cytology , Corpus Callosum/radiation effects , Dose-Response Relationship, Radiation , Male , Mice , Mice, Inbred C57BL , Neural Stem Cells/cytology , Organ Size/radiation effects , Time Factors , White Matter/cytology , White Matter/radiation effects
11.
Neuro Oncol ; 19(2): 153-161, 2017 02 01.
Article En | MEDLINE | ID: mdl-27282398

High-grade gliomas in children are different from those that arise in adults. Recent collaborative molecular analyses of these rare cancers have revealed previously unappreciated connections among chromatin regulation, developmental signaling, and tumorigenesis. As we begin to unravel the unique developmental origins and distinct biological drivers of this heterogeneous group of tumors, clinical trials need to keep pace. It is important to avoid therapeutic strategies developed purely using data obtained from studies on adult glioblastoma. This approach has resulted in repetitive trials and ineffective treatments being applied to these children, with limited improvement in clinical outcome. The authors of this perspective, comprising biology and clinical expertise in the disease, recently convened to discuss the most effective ways to translate the emerging molecular insights into patient benefit. This article reviews our current understanding of pediatric high-grade glioma and suggests approaches for innovative clinical management.


Brain Neoplasms/pathology , Cell Transformation, Neoplastic/pathology , Glioma/pathology , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Child , Glioma/genetics , Glioma/metabolism , Humans , Neoplasm Grading , Prognosis
12.
Nat Cell Biol ; 18(12): 1336-1345, 2016 Dec.
Article En | MEDLINE | ID: mdl-27820599

Increased overall survival for patients with glioma brain tumours is associated with mutations in the metabolic regulator isocitrate dehydrogenase 1 (IDH1). Gliomas develop within a mechanically challenged microenvironment that is characterized by a dense extracellular matrix (ECM) that compromises vascular integrity to induce hypoxia and activate HIF1α. We found that glioma aggression and patient prognosis correlate with HIF1α levels and the stiffness of a tenascin C (TNC)-enriched ECM. Gain- and loss-of-function xenograft manipulations demonstrated that a mutant IDH1 restricts glioma aggression by reducing HIF1α-dependent TNC expression to decrease ECM stiffness and mechanosignalling. Recurrent IDH1-mutant patient gliomas had a stiffer TNC-enriched ECM that our studies attributed to reduced miR-203 suppression of HIF1α and TNC mediated via a tension-dependent positive feedback loop. Thus, our work suggests that elevated ECM stiffness can independently foster glioblastoma aggression and contribute to glioblastoma recurrence via bypassing the protective activity of IDH1 mutational status.


Brain Neoplasms/pathology , Feedback, Physiological , Glioblastoma/metabolism , Glioblastoma/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Isocitrate Dehydrogenase/metabolism , Tenascin/metabolism , Brain Neoplasms/metabolism , Cell Line, Tumor , Extracellular Matrix/metabolism , Fluorescent Antibody Technique , Humans , Isocitrate Dehydrogenase/genetics , Mechanotransduction, Cellular , MicroRNAs/metabolism , Mutation/genetics , Neoplasm Invasiveness , Signal Transduction , Xenograft Model Antitumor Assays
13.
Oncotarget ; 7(43): 69173-69187, 2016 Oct 25.
Article En | MEDLINE | ID: mdl-27732951

Glioblastoma multiforme (GBM) carries a poor prognosis and continues to lack effective treatments. Glioblastoma stem cells (GSCs) drive tumor formation, invasion, and drug resistance and, as such, are the focus of studies to identify new therapies for disease control. Here, we identify the involvement of IKK and NF-κB signaling in the maintenance of GSCs. Inhibition of this pathway impairs self-renewal as analyzed in tumorsphere formation and GBM expansion as analyzed in brain slice culture. Interestingly, both the canonical and non-canonical branches of the NF-κB pathway are shown to contribute to this phenotype. One source of NF-κB activation in GBM involves the TGF-ß/TAK1 signaling axis. Together, our results demonstrate a role for the NF-κB pathway in GSCs and provide a mechanistic basis for its potential as a therapeutic target in glioblastoma.


Cell Self Renewal , I-kappa B Kinase/metabolism , NF-kappa B/metabolism , Neoplastic Stem Cells/metabolism , Signal Transduction , Animals , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , I-kappa B Kinase/genetics , MAP Kinase Kinase Kinases/metabolism , NF-kappa B/genetics , RNA Interference , Rats , Spheroids, Cellular/metabolism , Tissue Culture Techniques , Transforming Growth Factor beta/metabolism
14.
Cancer Res ; 75(20): 4302-11, 2015 Oct 15.
Article En | MEDLINE | ID: mdl-26282165

High grade gliomas (HGG) are classified into four subgroups based on transcriptional signatures and phenotypic characteristics. In particular, the proneural-to-mesenchymal transition (PMT) is associated with increased malignancy, poor prognosis, and disease recurrence, but the underlying causes of PMT are still unclear. In this study, we investigated whether radiotherapy promotes PMT using a genetically engineered mouse model of proneural HGG. We found that cranial ionizing radiation induced robust and durable PMT in tumors. Additionally, we isolated primary proneural HGG cells from mouse and human tumors and demonstrate that radiation induced a sustained cell-intrinsic mesenchymal transition associated with increased invasiveness and resistance to the alkylating agent temozolomide. Expectedly, irradiation-induced PMT was also associated with activation of the STAT3 transcription factor, and the combination of STAT3 blockade using JAK2 inhibitors with radiation abrogated the mesenchymal transition and extended survival of mice. Taken together, our data suggest that clinical JAK2 inhibitors should be tested in conjunction with radiation in patients with proneural HGG as a new strategy for blocking the emergence of therapy-resistant mesenchymal tumors at relapse.


Glioma/metabolism , Glioma/pathology , STAT3 Transcription Factor/antagonists & inhibitors , Allografts , Animals , Biomarkers , Disease Models, Animal , Disease Progression , Gene Expression Regulation, Neoplastic , Glioma/drug therapy , Glioma/genetics , Glioma/radiotherapy , Humans , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/metabolism , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Mesenchymal Stem Cells/radiation effects , Mice , Mice, Knockout , Neoplasm Grading , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Radiation , STAT3 Transcription Factor/metabolism
15.
Cell Tissue Res ; 359(1): 225-54, 2015 Jan.
Article En | MEDLINE | ID: mdl-25416506

Neurogenesis, the generation of new neurons, is deregulated in neural stem cell (NSC)- and progenitor-derived murine models of malignant medulloblastoma and glioma, the most common brain tumors of children and adults, respectively. Molecular characterization of human malignant brain tumors, and in particular brain tumor stem cells (BTSCs), has identified neurodevelopmental transcription factors, microRNAs, and epigenetic factors known to inhibit neuronal and glial differentiation. We are starting to understand how these factors are regulated by the major oncogenic drivers in malignant brain tumors. In this review, we will focus on the molecular switches that block normal neuronal differentiation and induce brain tumor formation. Genetic or pharmacological manipulation of these switches in BTSCs has been shown to restore the ability of tumor cells to differentiate. We will discuss potential brain tumor therapies that will promote differentiation in order to reduce treatment resistance, suppress tumor growth, and prevent recurrence in patients.


Brain Neoplasms/pathology , Cell Differentiation , Animals , Brain Neoplasms/genetics , Carcinogenesis/pathology , Cell Proliferation , Epigenesis, Genetic , Humans , Neurogenesis
16.
Mol Cancer Ther ; 14(2): 419-28, 2015 Feb.
Article En | MEDLINE | ID: mdl-25522764

Glioblastoma (GBM) is the most common malignant primary brain tumor. Radiotherapy fails to eliminate subpopulations of stem-like tumor-propagating cells (TPC), resulting in tumor regrowth. To identify kinases that promote TPC self-renewal rather than increasing proliferation in human GBM cultures, we screened a library of 54 nonselective tool compounds and determined their kinase inhibitor profiles in vitro. Most compounds inhibited aurora kinase (AURK) activity and blocked TPC self-renewal, while inducing GBM cell polynucleation and apoptosis. To prevent regrowth by TPCs, we used a priming dose of radiation followed by incubation with the pan-AURK inhibitor VX680 to block self-renewal and induce apoptosis in GBM cultures. In mice xenografted with human GBM cells, radiotherapy followed by VX680 treatment resulted in reduced tumor growth and increased survival relative to either monotherapy alone or VX680 treatment before radiation. Our results indicate that AURK inhibition, subsequent to radiation, may enhance the efficacy of radiotherapy by targeting radioresistant TPCs in human GBMs.


Aurora Kinases/antagonists & inhibitors , Brain Neoplasms/drug therapy , Brain Neoplasms/radiotherapy , Glioblastoma/drug therapy , Glioblastoma/radiotherapy , Neoplastic Stem Cells/pathology , Protein Kinase Inhibitors/therapeutic use , Animals , Apoptosis/drug effects , Aurora Kinases/metabolism , Biomarkers, Tumor/metabolism , Brain Neoplasms/pathology , Cell Cycle Checkpoints/drug effects , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Glioblastoma/pathology , Histones/metabolism , Humans , Membrane Glycoproteins/metabolism , Mice, Nude , Neoplastic Stem Cells/drug effects , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Radiation Tolerance/drug effects , Xenograft Model Antitumor Assays
17.
Adv Cancer Res ; 121: 1-65, 2014.
Article En | MEDLINE | ID: mdl-24889528

Glioma is the most common primary malignant brain tumor and arises throughout the central nervous system. Recent focus on stem-like glioma cells has implicated neural stem cells (NSCs), a minor precursor population restricted to germinal zones, as a potential source of gliomas. In this review, we focus on the relationship between oligodendrocyte progenitor cells (OPCs), the largest population of cycling glial progenitors in the postnatal brain, and gliomagenesis. OPCs can give rise to gliomas, with signaling pathways associated with NSCs also playing key roles during OPC lineage development. Gliomas can also undergo a switch from progenitor- to stem-like phenotype after therapy, consistent with an OPC-origin even for stem-like gliomas. Future in-depth studies of OPC biology may shed light on the etiology of OPC-derived gliomas and reveal new therapeutic avenues.


Cell Transformation, Neoplastic , Glioma/pathology , Neural Stem Cells/physiology , Neuroglia/physiology , Animals , Cell Differentiation/genetics , Cell Lineage/physiology , Cell Transformation, Neoplastic/pathology , Glioma/genetics , Glioma/therapy , Humans , Molecular Targeted Therapy , Neural Stem Cells/pathology , Neuroglia/pathology
18.
Exp Neurol ; 260: 56-68, 2014 Oct.
Article En | MEDLINE | ID: mdl-23376224

Brain tumors have frequently been associated with a neural stem cell (NSC) origin and contain stem-like tumor cells, so-called brain tumor stem cells (BTSCs) that share many features with normal NSCs. A stem cell state of BTSCs confers resistance to radiotherapy and treatment with alkylating agents. It is also a hallmark of aggressive brain tumors and is maintained by transcriptional networks that are also active in embryonic stem cells. Advances in reprogramming of somatic cells into induced pluripotent stem (iPS) cells have further identified genes that drive stemness. In this review, we will highlight the possible drivers of stemness in medulloblastoma and glioma, the most frequent types of primary malignant brain cancer in children and adults, respectively. Signals that drive expansion of developmentally defined neural precursor cells are also active in corresponding brain tumors. Transcriptomal subgroups of human medulloblastoma and glioma match features of NSCs but also more restricted progenitors. Lessons from genetically-engineered mouse (GEM) models show that temporally and regionally defined NSCs can give rise to distinct subgroups of medulloblastoma and glioma. We will further discuss how acquisition of stem cell features may drive brain tumorigenesis from a non-NSC origin. Genetic alterations, signaling pathways, and therapy-induced changes in the tumor microenvironment can drive reprogramming networks and induce stemness in brain tumors. Finally, we propose a model where dysregulation of microRNAs (miRNAs) that normally provide barriers against reprogramming plays an integral role in promoting stemness in brain tumors.


Cell Differentiation/physiology , Cell Lineage , Cell Transformation, Neoplastic/genetics , Neural Stem Cells/cytology , Signal Transduction/physiology , Stem Cells/cytology , Animals , Humans
19.
Proc Natl Acad Sci U S A ; 110(16): E1480-9, 2013 Apr 16.
Article En | MEDLINE | ID: mdl-23542378

Based on clinical presentation, glioblastoma (GBM) is stratified into primary and secondary types. The protein 53 (p53) pathway is functionally incapacitated in most GBMs by distinctive type-specific mechanisms. To model human gliomagenesis, we used a GFAP-HRas(V12) mouse model crossed into the p53ER(TAM) background, such that either one or both copies of endogenous p53 is replaced by a conditional p53ER(TAM) allele. The p53ER(TAM) protein can be toggled reversibly in vivo between wild-type and inactive conformations by administration or withdrawal of 4-hydroxytamoxifen (4-OHT), respectively. Surprisingly, gliomas that develop in GFAP-HRas(V12);p53(+/KI) mice abrogate the p53 pathway by mutating p19(ARF)/MDM2 while retaining wild-type p53 allele. Consequently, such tumors are unaffected by restoration of their p53ER(TAM) allele. By contrast, gliomas arising in GFAP-HRas(V12);p53(KI/KI) mice develop in the absence of functional p53. Such tumors retain a functional p19(ARF)/MDM2-signaling pathway, and restoration of p53ER(TAM) allele triggers p53-tumor-suppressor activity. Congruently, growth inhibition upon normalization of mutant p53 by a small molecule, Prima-1, in human GBM cultures also requires p14(ARF)/MDM2 functionality. Notably, the antitumoral efficacy of p53 restoration in tumor-bearing GFAP-HRas(V12);p53(KI/KI) animals depends on the duration and frequency of p53 restoration. Thus, intermittent exposure to p53ER(TAM) activity mitigated the selective pressure to inactivate the p19(ARF)/MDM2/p53 pathway as a means of resistance, extending progression-free survival. Our results suggest that intermittent dosing regimes of drugs that restore wild-type tumor-suppressor function onto mutant, inactive p53 proteins will prove to be more efficacious than traditional chronic dosing by similarly reducing adaptive resistance.


Disease Models, Animal , Glioblastoma/drug therapy , Glioblastoma/physiopathology , Signal Transduction/physiology , Tamoxifen/analogs & derivatives , Tumor Suppressor Protein p53/metabolism , Animals , Base Sequence , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p16/genetics , DNA Primers/genetics , Fluorescent Antibody Technique , Glioblastoma/metabolism , Histological Techniques , Humans , Immunoblotting , Kaplan-Meier Estimate , Mice , Mice, Transgenic , Molecular Sequence Data , Mutation/genetics , Proto-Oncogene Proteins c-mdm2/genetics , Sequence Analysis, DNA , Signal Transduction/genetics , Tamoxifen/pharmacology , Tamoxifen/therapeutic use
20.
Cancer Cell ; 21(5): 601-613, 2012 May 15.
Article En | MEDLINE | ID: mdl-22624711

The proto-oncogene MYCN is mis-expressed in various types of human brain tumors. To clarify how developmental and regional differences influence transformation, we transduced wild-type or mutationally stabilized murine N-myc(T58A) into neural stem cells (NSCs) from perinatal murine cerebellum, brain stem, and forebrain. Transplantation of N-myc(WT) NSCs was insufficient for tumor formation. N-myc(T58A) cerebellar and brain stem NSCs generated medulloblastoma/primitive neuroectodermal tumors, whereas forebrain NSCs developed diffuse glioma. Expression analyses distinguished tumors generated from these different regions, with tumors from embryonic versus postnatal cerebellar NSCs demonstrating Sonic Hedgehog (SHH) dependence and SHH independence, respectively. These differences were regulated in part by the transcription factor SOX9, activated in the SHH subclass of human medulloblastoma. Our results demonstrate context-dependent transformation of NSCs in response to a common oncogenic signal.


Brain Neoplasms/metabolism , Cell Lineage , Cell Transformation, Neoplastic/metabolism , Neural Stem Cells/metabolism , Nuclear Proteins/metabolism , Oncogene Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Animals , Biomarkers/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Brain Stem/embryology , Brain Stem/metabolism , Cell Differentiation , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Cerebellar Neoplasms/metabolism , Cerebellar Neoplasms/pathology , Cerebellum/embryology , Cerebellum/metabolism , Female , Gestational Age , Glioma/metabolism , Glioma/pathology , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Humans , Kruppel-Like Transcription Factors/metabolism , Medulloblastoma/metabolism , Medulloblastoma/pathology , Mice , Mice, Nude , Mice, Transgenic , Mutation , N-Myc Proto-Oncogene Protein , Neural Stem Cells/pathology , Neuroectodermal Tumors, Primitive/metabolism , Neuroectodermal Tumors, Primitive/pathology , Nuclear Proteins/genetics , Oncogene Proteins/genetics , Prosencephalon/embryology , Prosencephalon/metabolism , Proto-Oncogene Mas , Proto-Oncogene Proteins/genetics , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Signal Transduction , Spheroids, Cellular , Time Factors , Transduction, Genetic , Zinc Finger Protein Gli2
...