Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 15 de 15
1.
Neuro Oncol ; 2024 Feb 16.
Article En | MEDLINE | ID: mdl-38366847

BACKGROUND: Precision treatment of glioblastoma is increasingly focused on molecular subtyping, with the mesenchymal subtype particularly resistant to temozolomide. Here, we aim to develop a targeted therapy for temozolomide resensitization in the mesenchymal subtype. METHODS: We integrated kinomic profiles and kinase inhibitor screens from patient-derived proneural and mesenchymal glioma-propagating cells public clinical datasets to identify key protein kinases implicated in temozolomide resistance. RNAseq, apoptosis assays and comet assays were used to examine the role of p38MAPK signaling and adaptive chemoresistance in mesenchymal cells. The efficacy of dual p38MAPK and MEK/ERK inhibition using ralimetinib (selective orally active p38MAPK inhibitor; phase I/II for glioblastoma) and binimetinib (approved MEK1/2 inhibitor for melanoma; phase II for high-grade glioma) in primary and recurrent mesenchymal tumors was evaluated using an intracranial patient-derived tumor xenograft model, focusing on survival analysis. RESULTS: Our transcriptomic-kinomic integrative analysis revealed p38MAPK as the prime target whose gene signature enables patient stratification based on their molecular subtypes and provides prognostic value. Repurposed p38MAPK inhibitors synergize favourably with temozolomide to promote intracellular retention of temozolomide and exacerbate DNA damage. Mesenchymal cells exhibit adaptive chemoresistance to p38MAPK inhibition through a pH-/calcium-mediated MEK/ERK pathway. Dual p38MAPK and MEK inhibition effectively maintains temozolomide sensitivity in primary and recurrent intracranial mesenchymal glioblastoma xenografts. CONCLUSION: Temozolomide resistance in mesenchymal glioblastoma is associated with p38MAPK activation. Adaptive chemoresistance in p38MAPK-resistant cells is mediated by MEK/ERK signaling. Adjuvant therapy with dual p38MAPK and MEK inhibition prolongs temozolomide sensitivity, which can be developed into a precision therapy for the mesenchymal subtype.

2.
Pharmacol Res ; 182: 106308, 2022 08.
Article En | MEDLINE | ID: mdl-35714825

This review describes recent technological advances applied to glioblastoma (GBM), a brain tumor with dismal prognosis. International consortial efforts suggest the presence of molecular subtypes within histologically identical GBM tumors. This emphasizes that future treatment decisions should no longer be made based solely on morphological analyses, but must now take into consideration such molecular and cellular heterogeneity. The use of single-cell technologies has advanced our understanding and assignation of functional subtypes revealing therapeutic vulnerabilities. Our team has developed stratification approaches in the past few years, and we have been able to identify patient cohorts enriched for various signaling pathways. Importantly, our Glioportal brain tumor resource has been established under the National Neuroscience Institute Tissue Bank in 2021. This resource offers preclinical capability to validate working hypotheses established from patient clinical datasets. This review highlights recent developments with the ultimate goal of assigning functional meaning to molecular subtypes, revealing therapeutic vulnerabilities.


Brain Neoplasms , Glioblastoma , Brain Neoplasms/metabolism , Glioblastoma/metabolism , Humans , Molecular Targeted Therapy , Precision Medicine , Prognosis
3.
Dev Cell ; 56(20): 2841-2855.e8, 2021 10 25.
Article En | MEDLINE | ID: mdl-34559979

Glioblastoma are heterogeneous tumors composed of highly invasive and highly proliferative clones. Heterogeneity in invasiveness could emerge from discrete biophysical properties linked to specific molecular expression. We identified clones of patient-derived glioma propagating cells that were either highly proliferative or highly invasive and compared their cellular architecture, migratory, and biophysical properties. We discovered that invasiveness was linked to cellular fitness. The most invasive cells were stiffer, developed higher mechanical forces on the substrate, and moved stochastically. The mechano-chemical-induced expression of the formin FMN1 conferred invasive strength that was confirmed in patient samples. Moreover, FMN1 expression was also linked to motility in other cancer and normal cell lines, and its ectopic expression increased fitness parameters. Mechanistically, FMN1 acts from the microtubule lattice and promotes a robust mechanical cohesion, leading to highly invasive motility.


Cell Movement/physiology , Formins/metabolism , Glioblastoma/metabolism , Neoplasm Invasiveness/pathology , Brain Neoplasms/pathology , Cell Line, Tumor , Fetal Proteins/metabolism , Glioblastoma/pathology , Humans , Microfilament Proteins/metabolism
4.
Sci Adv ; 7(18)2021 04.
Article En | MEDLINE | ID: mdl-33931443

Molecular profiling of the most aggressive brain tumor glioblastoma (GBM) on the basis of gene expression, DNA methylation, and genomic variations advances both cancer research and clinical diagnosis. The enhancer architectures and regulatory circuitries governing tumor-intrinsic transcriptional diversity and subtype identity are still elusive. Here, by mapping H3K27ac deposition, we analyze the active regulatory landscapes across 95 GBM biopsies, 12 normal brain tissues, and 38 cell line counterparts. Analyses of differentially regulated enhancers and super-enhancers uncovered previously unrecognized layers of intertumor heterogeneity. Integrative analysis of variant enhancer loci and transcriptome identified topographies of transcriptional enhancers and core regulatory circuitries in four molecular subtypes of primary tumors: AC1-mesenchymal, AC1-classical, AC2-proneural, and AC3-proneural. Moreover, this study reveals core oncogenic dependency on super-enhancer-driven transcriptional factors, long noncoding RNAs, and druggable targets in GBM. Through profiling of transcriptional enhancers, we provide clinically relevant insights into molecular classification, pathogenesis, and therapeutic intervention of GBM.


Brain Neoplasms , Glioblastoma , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Chromatin/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Glioblastoma/pathology , Humans
5.
Theranostics ; 11(11): 5127-5142, 2021.
Article En | MEDLINE | ID: mdl-33859738

Hypoxic microenvironment is a hallmark of solid tumors, especially glioblastoma. The strong reliance of glioma-propagating cells (GPCs) on hypoxia-induced survival advantages is potentially exploitable for drug development. Methods: To identify key signaling pathways for hypoxia adaptation by patient-derived GPCs, we performed a kinase inhibitor profiling by screening 188 small molecule inhibitors against 130 different kinases in normoxia and hypoxia. Potential kinase candidates were prioritized for in vitro and in vivo investigations using a ranking algorithm that integrated information from the kinome connectivity network and estimated patients' survival based on expression status. Results: Hypoxic drug screen highlighted extensive modifications of kinomic landscape and a crucial functionality of c-MET-PI3K. c-MET inhibitors diminished phosphorylation of c-MET and PI3K in GPCs subjected to hypoxia, suggesting its role in the hypoxic adaptation of GPCs. Mechanistically, the inhibition of c-MET and PI3K impaired antioxidant defense, leading to oxidative catastrophe and apoptosis. Repurposed c-MET inhibitors PF04217903 and tivantinib exhibited hypoxic-dependent drug synergism with temozolomide, resulting in reduced tumor load and growth of GPC xenografts. Detailed analysis of bulk and single-cell glioblastoma transcriptomes associates the cellular subpopulation over-expressing c-MET with inflamed, hypoxic, metastatic, and stem-like phenotypes. Conclusions: Thus, our "bench to bedside (the use of patient-derived GPCs and xenografts for basic research) and back (validation with independent glioblastoma transcriptome databases)" analysis unravels the novel therapeutic indications of c-MET and PI3K/Akt inhibitors for the treatment of glioblastoma, and potentially other cancers, in the hypoxic tumor microenvironment.


Glioma/genetics , Hypoxia/genetics , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-met/genetics , Animals , Antioxidants/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Cell Line, Tumor , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioma/drug therapy , Humans , Hypoxia/drug therapy , Male , Mice , Phosphorylation/drug effects , Phosphorylation/genetics , Pyrazines/pharmacology , Pyrrolidinones/pharmacology , Quinolines/pharmacology , Signal Transduction/drug effects , Signal Transduction/genetics , Temozolomide/pharmacology , Transcriptome/drug effects , Transcriptome/genetics , Triazoles/pharmacology
6.
Nat Commun ; 10(1): 3601, 2019 08 09.
Article En | MEDLINE | ID: mdl-31399589

Intratumoral heterogeneity is a hallmark of glioblastoma (GBM) tumors, thought to negatively influence therapeutic outcome. Previous studies showed that mesenchymal tumors have a worse outcome than the proneural subtype. Here we focus on STAT3 as its activation precedes the proneural-mesenchymal transition. We first establish a STAT3 gene signature that stratifies GBM patients into STAT3-high and -low cohorts. STAT3 inhibitor treatment selectively mitigates STAT3-high cell viability and tumorigenicity in orthotopic mouse xenograft models. We show the mechanism underlying resistance in STAT3-low cells by combining STAT3 signature analysis with kinome screen data on STAT3 inhibitor-treated cells. This allows us to draw connections between kinases affected by STAT3 inhibitors, their associated transcription factors and target genes. We demonstrate that dual inhibition of IGF-1R and STAT3 sensitizes STAT3-low cells and improves survival in mice. Our study underscores the importance of serially profiling tumors so as to accurately target individuals who may demonstrate molecular subtype switching.


Genetic Predisposition to Disease/genetics , Glioblastoma/drug therapy , Glioblastoma/genetics , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Animals , Cell Survival , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Synergism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/genetics , Gene Knockdown Techniques , Humans , Imidazoles/pharmacology , Insulin-Like Growth Factor Binding Protein 2/genetics , Insulin-Like Growth Factor Binding Protein 2/metabolism , Mice , Pyrazines/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Receptor, IGF Type 1/antagonists & inhibitors , Receptor, IGF Type 1/genetics , STAT3 Transcription Factor/antagonists & inhibitors , Temozolomide/pharmacology , Xenograft Model Antitumor Assays
7.
Anal Chem ; 90(17): 10249-10255, 2018 09 04.
Article En | MEDLINE | ID: mdl-30070837

We report the development and implementation of an epi-detected spectral-focusing hyperspectral stimulated Raman scattering (SRS) imaging technique for label-free biomolecular subtyping of glioblastomas (GBMs). The hyperspectral SRS imaging technique developed generates SRS image stacks (from 2800 to 3020 cm-1 at 7 cm-1 intervals) within 30 s through controlling the time delay between the chirped pump and Stokes beams. SRS images at representative Raman shifts (e.g., 2845, 2885, and 2935 cm-1) delineate the biochemical variations and morphological differences between proneural and mesenchymal subtypes of GBMs. Multivariate curve resolution (MCR) analysis on hyperspectral SRS images enables the quantification of major biomolecule distributions in mesenchymal and proneural GBMs. Further principal component analysis (PCA) and linear discriminant analysis (LDA) together with leave-one SRS spectrum-out, cross-validation (LOOCV) yields a diagnostic sensitivity of 96.7% (29/30) and specificity of 88.9% (28/36) for differentiation between mesenchymal and proneural subtypes of GBMs. This study shows great potential of applying hyperspectral SRS imaging technique developed for rapid, label-free molecular subtyping of GBMs in neurosurgery.


Brain Neoplasms/classification , Glioblastoma/classification , Nonlinear Optical Microscopy/methods , Spectrum Analysis, Raman/methods , Humans , Multivariate Analysis , Principal Component Analysis
8.
Proc Natl Acad Sci U S A ; 115(22): E5086-E5095, 2018 05 29.
Article En | MEDLINE | ID: mdl-29764999

Competitive BET bromodomain inhibitors (BBIs) targeting BET proteins (BRD2, BRD3, BRD4, and BRDT) show promising preclinical activities against brain cancers. However, the BET protein-dependent glioblastoma (GBM)-promoting transcriptional network remains elusive. Here, with mechanistic exploration of a next-generation chemical degrader of BET proteins (dBET6), we reveal a profound and consistent impact of BET proteins on E2F1- dependent transcriptional program in both differentiated GBM cells and brain tumor-initiating cells. dBET6 treatment drastically reduces BET protein genomic occupancy, RNA-Pol2 activity, and permissive chromatin marks. Subsequently, dBET6 represses the proliferation, self-renewal, and tumorigenic ability of GBM cells. Moreover, dBET6-induced degradation of BET proteins exerts superior antiproliferation effects compared to conventional BBIs and overcomes both intrinsic and acquired resistance to BBIs in GBM cells. Our study reveals crucial functions of BET proteins and provides the rationale and therapeutic merits of targeted degradation of BET proteins in GBM.


Antineoplastic Agents/pharmacology , E2F1 Transcription Factor , Glioblastoma , Protein Serine-Threonine Kinases , RNA-Binding Proteins , Cell Cycle Proteins , Cell Line, Tumor , Drug Delivery Systems , E2F1 Transcription Factor/antagonists & inhibitors , E2F1 Transcription Factor/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/metabolism , Protein Domains , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism
9.
Mol Biol Cell ; 27(8): 1246-61, 2016 Apr 15.
Article En | MEDLINE | ID: mdl-26912794

Glioblastomas are extremely aggressive brain tumors with highly invasive properties. Brain linear tracks such as blood vessel walls constitute their main invasive routes. Here we analyze rat C6 and patient-derived glioma cell motility in vitro using micropatterned linear tracks to mimic blood vessels. On laminin-coated tracks (3-10 µm), these cells used an efficient saltatory mode of migration similar to their in vivo migration. This saltatory migration was also observed on larger tracks (50-400 µm in width) at high cell densities. In these cases, the mechanical constraints imposed by neighboring cells triggered this efficient mode of migration, resulting in the formation of remarkable antiparallel streams of cells along the tracks. This motility involved microtubule-dependent polarization, contractile actin bundles and dynamic paxillin-containing adhesions in the leading process and in the tail. Glioma linear migration was dramatically reduced by inhibiting formins but, surprisingly, accelerated by inhibiting Arp2/3. Protein expression and phenotypic analysis indicated that the formin FHOD3 played a role in this motility but not mDia1 or mDia2. We propose that glioma migration under confinement on laminin relies on formins, including FHOD3, but not Arp2/3 and that the low level of adhesion allows rapid antiparallel migration.


Brain Neoplasms/pathology , Cell Migration Assays/methods , Glioma/pathology , Microfilament Proteins/metabolism , Muscle Proteins/metabolism , Actin-Related Protein 2-3 Complex/metabolism , Animals , Biomechanical Phenomena , Brain Neoplasms/blood supply , Cell Adhesion , Cell Count , Cell Movement , Formins , Glioblastoma/pathology , Glioma/blood supply , Humans , Laminin/metabolism , Microfilament Proteins/genetics , Muscle Proteins/genetics , Paxillin/metabolism , Rats , Tumor Cells, Cultured
10.
J Natl Cancer Inst ; 108(2)2016 Feb.
Article En | MEDLINE | ID: mdl-26547933

BACKGROUND: Cell surface sialylation is associated with tumor cell invasiveness in many cancers. Glioblastoma is the most malignant primary brain tumor and is highly infiltrative. ST3GAL1 sialyltransferase gene is amplified in a subclass of glioblastomas, and its role in tumor cell self-renewal remains unexplored. METHODS: Self-renewal of patient glioma cells was evaluated using clonogenic, viability, and invasiveness assays. ST3GAL1 was identified from differentially expressed genes in Peanut Agglutinin-stained cells and validated in REMBRANDT (n = 390) and Gravendeel (n = 276) clinical databases. Gene set enrichment analysis revealed upstream processes. TGFß signaling on ST3GAL1 transcription was assessed using chromatin immunoprecipitation. Transcriptome analysis of ST3GAL1 knockdown cells was done to identify downstream pathways. A constitutively active FoxM1 mutant lacking critical anaphase-promoting complex/cyclosome ([APC/C]-Cdh1) binding sites was used to evaluate ST3Gal1-mediated regulation of FoxM1 protein. Finally, the prognostic role of ST3Gal1 was determined using an orthotopic xenograft model (3 mice groups comprising nontargeting and 2 clones of ST3GAL1 knockdown in NNI-11 [8 per group] and NNI-21 [6 per group]), and the correlation with patient clinical information. All statistical tests on patients' data were two-sided; other P values below are one-sided. RESULTS: High ST3GAL1 expression defines an invasive subfraction with self-renewal capacity; its loss of function prolongs survival in a mouse model established from mesenchymal NNI-11 (P < .001; groups of 8 in 3 arms: nontargeting, C1, and C2 clones of ST3GAL1 knockdown). ST3GAL1 transcriptomic program stratifies patient survival (hazard ratio [HR] = 2.47, 95% confidence interval [CI] = 1.72 to 3.55, REMBRANDT P = 1.92 x 10⁻8; HR = 2.89, 95% CI = 1.94 to 4.30, Gravendeel P = 1.05 x 10⁻¹¹), independent of age and histology, and associates with higher tumor grade and T2 volume (P = 1.46 x 10⁻4). TGFß signaling, elevated in mesenchymal patients, correlates with high ST3GAL1 (REMBRANDT gliomacor = 0.31, P = 2.29 x 10⁻¹°; Gravendeel gliomacor = 0.50, P = 3.63 x 10⁻²°). The transcriptomic program upon ST3GAL1 knockdown enriches for mitotic cell cycle processes. FoxM1 was identified as a statistically significantly modulated gene (P = 2.25 x 10⁻5) and mediates ST3Gal1 signaling via the (APC/C)-Cdh1 complex. CONCLUSIONS: The ST3GAL1-associated transcriptomic program portends poor prognosis in glioma patients and enriches for higher tumor grades of the mesenchymal molecular classification. We show that ST3Gal1-regulated self-renewal traits are crucial to the sustenance of glioblastoma multiforme growth.


Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Glioblastoma/metabolism , Glioblastoma/pathology , Sialyltransferases/metabolism , Transcriptome , Transforming Growth Factor beta/metabolism , Animals , Cell Proliferation , Cell Survival , Chromatin Immunoprecipitation , Forkhead Box Protein M1 , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Heterografts , Humans , Kaplan-Meier Estimate , Mice , Neoplasm Invasiveness , Prognosis , Signal Transduction , Tumor Stem Cell Assay , Up-Regulation , beta-Galactoside alpha-2,3-Sialyltransferase
11.
Antioxid Redox Signal ; 19(18): 2261-79, 2013 Dec 20.
Article En | MEDLINE | ID: mdl-23477542

AIMS: We explore the role of an elevated O2(-):H2O2 ratio as a prosurvival signal in glioma-propagating cells (GPCs). We hypothesize that depleting this ratio sensitizes GPCs to apoptotic triggers. RESULTS: We observed that an elevated O2(-):H2O2 ratio conferred enhanced resistance in GPCs, and depletion of this ratio by pharmacological and genetic methods sensitized cells to apoptotic triggers. We established the reactive oxygen species (ROS) Index as a quantitative measure of a normalized O2(-):H2O2 ratio and determined its utility in predicting chemosensitivity. Importantly, mice implanted with GPCs of a reduced ROS Index demonstrated extended survival. Analysis of tumor sections revealed effective targeting of complementarity determinant 133 (CD133)- and nestin-expressing neural precursors. Further, we established the Connectivity Map to interrogate a gene signature derived from a varied ROS Index for the patterns of association with individual patient gene expression in four clinical databases. We showed that patients with a reduced ROS Index demonstrate better survival. These data provide clinical evidence for the viability of our O2(-):H2O2-mediated chemosensitivity profiles. INNOVATION AND CONCLUSION: Gliomas are notoriously recurrent and highly infiltrative, and have been shown to arise from stem-like cells. We implicate an elevated O2(-):H2O2 ratio as a prosurvival signal in GPC self-renewal and proliferation. The ROS Index provides quantification of O2(-):H2O2-mediated chemosensitivity, an advancement in a previously qualitative field. Intriguingly, glioma patients with a reduced ROS Index correlate with longer survival and the Proneural molecular classification, a feature frequently associated with tumors of better prognosis. These data emphasize the feasibility of manipulating the O2(-):H2O2 ratio as a therapeutic strategy.


Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Drug Resistance, Neoplasm/drug effects , Glioma/drug therapy , Glioma/metabolism , Reactive Oxygen Species/metabolism , Brain Neoplasms/diagnosis , Glioma/diagnosis , Glioma/pathology , Humans , Hydrogen Peroxide/metabolism , Oxygen/metabolism , Prognosis , Survival Analysis , Treatment Outcome
12.
J Biomol Screen ; 17(9): 1136-50, 2012 Oct.
Article En | MEDLINE | ID: mdl-22927677

Gliomas are the most devastating of primary adult malignant brain tumors. These tumors are highly infiltrative and can arise from cells with extensive self-renewal capability and chemoresistance, frequently termed glioma-propagating cells (GPCs). GPCs are thus the plausible culprits of tumor recurrence. Treatment strategies that eradicate GPCs will greatly improve disease outcome. Such findings support the use of GPCs as in vitro cellular systems for small-molecule screening. However, the nuances in using GPCs as a cellular screening platform are not trivial. These slow-growing cells are typically cultured as suspension, spheroid structures in serum-free condition supplemented with growth factors. Consequently, replenishment of growth factors throughout the screening period must occur to maintain cells in their undifferentiated state, as the more lineage-committed, differentiated cells are less tumorigenic. We present a case study of a small-molecule screen conducted with GPCs and explain how unique sphere activity assays were implemented to distinguish drug efficacies against the long-term, self-renewing fraction, as opposed to transient-amplifying progenitors, the latter of which are detected in conventional viability assays. We identified Polo-like kinase 1 as a regulator of GPC survival. Finally, we leveraged on public glioma databases to illustrate GPC contribution to disease progression and patient survival outcome.


Brain Neoplasms/enzymology , Cell Cycle Proteins/metabolism , Drug Screening Assays, Antitumor/methods , Glioma/enzymology , Neoplastic Stem Cells/drug effects , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Animals , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Cell Cycle Proteins/analysis , Cell Cycle Proteins/antagonists & inhibitors , Cell Proliferation/drug effects , Cell Survival , Computational Biology , Databases, Genetic , Disease Progression , Glioma/mortality , Glioma/pathology , Humans , Inhibitory Concentration 50 , Mice , Microarray Analysis , Neoplastic Stem Cells/enzymology , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/analysis , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/analysis , Proto-Oncogene Proteins/antagonists & inhibitors , Pteridines/pharmacology , Small Molecule Libraries/pharmacology , Spheroids, Cellular/drug effects , Spheroids, Cellular/enzymology , Tumor Cells, Cultured , Polo-Like Kinase 1
13.
Cancer Res ; 72(10): 2543-53, 2012 May 15.
Article En | MEDLINE | ID: mdl-22431710

Mutations in the parkin gene, which encodes a ubiquitin ligase, are a major genetic cause of parkinsonism. Interestingly, parkin also plays a role in cancer as a putative tumor suppressor, and the gene is frequently targeted by deletion and inactivation in human malignant tumors. Here, we investigated a potential tumor suppressor role for parkin in gliomas. We found that parkin expression was dramatically reduced in glioma cells. Restoration of parkin expression promoted G(1) phase cell-cycle arrest and mitigated the proliferation rate of glioma cells in vitro and in vivo. Notably, parkin-expressing glioma cells showed a reduction in levels of cyclin D1, but not cyclin E, and a selective downregulation of Akt serine-473 phosphorylation and VEGF receptor levels. In accordance, cells derived from a parkin-null mouse model exhibited increased levels of cyclin D1, VEGF receptor, and Akt phosphorylation, and divided significantly faster when compared with wild-type cells, with suppression of these changes following parkin reintroduction. Clinically, analysis of parkin pathway activation was predictive for the survival outcome of patients with glioma. Taken together, our study provides mechanistic insight into the tumor suppressor function of parkin in brain tumors and suggests that measurement of parkin pathway activation may be used clinically as a prognostic tool in patients with brain tumor.


Brain Neoplasms/metabolism , Genes, Tumor Suppressor , Glioma/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Cyclin D1/metabolism , Down-Regulation , Glioma/genetics , Glioma/mortality , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Prognosis , Ubiquitin-Protein Ligases/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
14.
Front Biosci (Schol Ed) ; 3(2): 698-708, 2011 01 01.
Article En | MEDLINE | ID: mdl-21196406

Glioblastoma multiforme (GBM) represents the most devastating adult brain tumor. GBM follows a hierarchical development in oncogenesis, with a sub-population of cells - brain tumor stem cells (BTSCs), exhibiting tumor-initiating potential. BTSCs possess extensive self-renewal capability and can repopulate the entire tumor mass. They are resistant to conventional therapies, suggesting that they are the likely candidates of tumor recurrence. Their eradication is thus important for an effective cure. Previous works showed that human-derived BTSCs could be stably maintained for 10-15 passages in serum-free condition, and gene expression and karyotypic hallmarks similar to the primary tumors were preserved. However, primary cells have been shown to sustain additional karyotypic aberrations owing to the harsh conditions of extended in vitro serial passage. Several investigators have proposed passaging these cells in xenograft models. A limitation of such an approach is the inability to return to identical passages for experimental repetitions, or the unavailability of suitably-aged mice for implantation. We have devised a method to cryopreserve BTSCs and that important characteristics were maintained, establishing a repository for drug screening endeavors.


Brain Neoplasms/pathology , Cryopreservation/methods , Glioblastoma/pathology , Neoplastic Stem Cells/pathology , Animals , Antineoplastic Agents/isolation & purification , Antineoplastic Agents/pharmacology , Drug Screening Assays, Antitumor , Humans , Mice , Neoplastic Stem Cells/drug effects
15.
Stem Cells ; 27(1): 29-39, 2009 Jan.
Article En | MEDLINE | ID: mdl-18845764

Cancer stem cells have been shown to initiate and sustain tumor growth. In many instances, clinical material is limited, compounded by a lack of methods to preserve such cells at convenient time points. Although brain tumor-initiating cells grown in a spheroid manner have been shown to maintain their integrity through serial transplantation in immune-compromised animals, practically, it is not always possible to have access to animals of suitable ages to continuously maintain these cells. We therefore explored vitrification as a cryopreservation technique for brain tumor-initiating cells. Tumor neurospheres were derived from five patients with glioblastoma multiforme (GBM). Cryopreservation in 90% serum and 10% dimethyl sulfoxide yielded greatest viability and could be explored in future studies. Vitrification yielded cells that maintained self-renewal and multipotentiality properties. Karyotypic analyses confirmed the presence of GBM hallmarks. Upon implantation into NOD/SCID mice, our vitrified cells reformed glioma masses that could be serially transplanted. Transcriptome analysis showed that the vitrified and nonvitrified samples in either the stem-like or differentiated states clustered together, providing evidence that vitrification does not change the genotype of frozen cells. Upon induction of differentiation, the transcriptomes of vitrified cells associated with the original primary tumors, indicating that tumor stem-like cells are a genetically distinct population from the differentiated mass, underscoring the importance of working with the relevant tumor-initiating population. Our results demonstrate that vitrification of brain tumor-initiating cells preserves the biological phenotype and genetic profiles of the cells. This should facilitate the establishment of a repository of tumor-initiating cells for subsequent experimental designs.


Cryopreservation , Glioblastoma/pathology , Neurons/pathology , Spheroids, Cellular/pathology , AC133 Antigen , Animals , Antigens, CD/metabolism , Cell Aggregation , Cell Differentiation , Cell Proliferation , Cell Shape , Cell Survival , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Glycoproteins/metabolism , Humans , Karyotyping , Mice , Mice, SCID , Multipotent Stem Cells/pathology , Neoplastic Stem Cells/pathology , Peptides/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
...