Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Cell Death Dis ; 15(6): 414, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38871731

ABSTRACT

The repurposing of medications developed for central nervous system (CNS) disorders, possessing favorable safety profiles and blood-brain barrier permeability, represents a promising strategy for identifying new therapies to combat glioblastoma (GBM). In this study, we investigated the anti-GBM activity of specific antipsychotics and antidepressants in vitro and in vivo. Our results demonstrate that these compounds share a common mechanism of action in GBM, disrupting lysosomal function and subsequently inducing lysosomal membrane rupture and cell death. Notably, PTEN intact GBMs possess an increased sensitivity to these compounds. The inhibition of lysosomal function synergized with inhibitors targeting the EGFR-PI3K-Akt pathway, leading to an energetic and antioxidant collapse. These findings provide a foundation for the potential clinical application of CNS drugs in GBM treatment. Additionally, this work offers critical insights into the mechanisms and determinants of cytotoxicity for drugs currently undergoing clinical trials as repurposing agents for various cancers, including Fluoxetine, Sertraline, Thioridazine, Chlorpromazine, and Fluphenazine.


Subject(s)
Antipsychotic Agents , Glioblastoma , Lysosomes , PTEN Phosphohydrolase , Signal Transduction , Humans , Glioblastoma/drug therapy , Glioblastoma/pathology , Glioblastoma/metabolism , PTEN Phosphohydrolase/metabolism , Lysosomes/metabolism , Lysosomes/drug effects , Signal Transduction/drug effects , Antipsychotic Agents/pharmacology , Antipsychotic Agents/therapeutic use , Animals , Cell Line, Tumor , Mice , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Brain Neoplasms/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Mice, Nude , Drug Repositioning , Phosphatidylinositol 3-Kinases/metabolism , ErbB Receptors/metabolism , ErbB Receptors/antagonists & inhibitors , Chlorpromazine/pharmacology
2.
Sci Rep ; 14(1): 12363, 2024 05 29.
Article in English | MEDLINE | ID: mdl-38811596

ABSTRACT

Radiotherapy is the standard treatment for glioblastoma (GBM), but the overall survival rate for radiotherapy treated GBM patients is poor. The use of adjuvant and concomitant temozolomide (TMZ) improves the outcome; however, the effectiveness of this treatment varies according to MGMT levels. Herein, we evaluated whether MGMT expression affected the radioresponse of human GBM, GBM stem-like cells (GSCs), and melanoma. Our results indicated a correlation between MGMT promoter methylation status and MGMT expression. MGMT-producing cell lines ACPK1, GBMJ1, A375, and MM415 displayed enhanced radiosensitivity when MGMT was silenced using siRNA or when inhibited by lomeguatrib, whereas the OSU61, NSC11, WM852, and WM266-4 cell lines, which do not normally produce MGMT, displayed reduced radiosensitivity when MGMT was overexpressed. Mechanistically lomeguatrib prolonged radiation-induced γH2AX retention in MGMT-producing cells without specific cell cycle changes, suggesting that lomeguatrib-induced radiosensitization in these cells is due to radiation-induced DNA double-stranded break (DSB) repair inhibition. The DNA-DSB repair inhibition resulted in cell death via mitotic catastrophe in MGMT-producing cells. Overall, our results demonstrate that MGMT expression regulates radioresponse in GBM, GSC, and melanoma, implying a role for MGMT as a target for radiosensitization.


Subject(s)
DNA Modification Methylases , DNA Repair Enzymes , Glioblastoma , Melanoma , Radiation Tolerance , Tumor Suppressor Proteins , Humans , Glioblastoma/genetics , Glioblastoma/radiotherapy , Glioblastoma/metabolism , Glioblastoma/pathology , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , Melanoma/genetics , Melanoma/metabolism , Melanoma/pathology , Melanoma/radiotherapy , DNA Modification Methylases/metabolism , DNA Modification Methylases/genetics , Cell Line, Tumor , Radiation Tolerance/genetics , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/radiation effects , Neoplastic Stem Cells/pathology , Promoter Regions, Genetic , DNA Methylation , DNA Repair , DNA Breaks, Double-Stranded/radiation effects , Gene Expression Regulation, Neoplastic , Temozolomide/pharmacology , Brain Neoplasms/genetics , Brain Neoplasms/radiotherapy , Brain Neoplasms/pathology , Brain Neoplasms/metabolism , Purines
3.
iScience ; 27(1): 108650, 2024 Jan 19.
Article in English | MEDLINE | ID: mdl-38226170

ABSTRACT

Severe tumor heterogeneity drives the aggressive and treatment refractory nature of glioblastomas (GBMs). While limiting GBM heterogeneity offers promising therapeutic potential, the underlying mechanisms that regulate GBM plasticity remain poorly understood. We utilized 14 patient-derived and four commercially available cell lines to uncover miR-194-3p as a key epigenetic determinant of stemness and transcriptional subtype in GBM. We demonstrate that miR-194-3p degrades TAB2, an important mediator of NF-κB activity, decreasing NF-κB transcriptional activity. The loss in NF-κB activity following miR-194-3p overexpression or TAB2 silencing decreased expression of induced pluripotent stem cell (iPSC) genes, inhibited the oncogenic IL-6/STAT3 signaling axis, suppressed the mesenchymal transcriptional subtype in relation to the proneural subtype, and induced differentiation from the glioma stem cell (GSC) to monolayer (ML) phenotype. miR-194-3p/TAB2/NF-κB signaling axis acts as an epigenetic switch that regulates GBM plasticity and targeting this signaling axis represents a potential strategy to limit transcriptional heterogeneity in GBMs.

4.
Clin Cancer Res ; 27(2): 554-565, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33087331

ABSTRACT

PURPOSE: Concurrent gemcitabine and nab-paclitaxel treatment is one of the preferred chemotherapy regimens for metastatic and locally advanced pancreatic ductal adenocarcinoma (PDAC). Previous studies demonstrate that caveolin-1 (Cav-1) expression is critical for nab-paclitaxel uptake into tumors and correlates with response. Gemcitabine increases nab-paclitaxel uptake by increasing Cav-1 expression. Thus, we hypothesized that pretreatment with gemcitabine would further enhance the sensitivity of PDAC to nab-paclitaxel by increasing Cav-1 expression and nab-paclitaxel uptake. EXPERIMENTAL DESIGN: We investigated the sensitivity of different gemcitabine and nab-paclitaxel treatment regimens in a panel of PDAC cell lines and orthotopic xenograft models. The sensitivity of different treatment regimens was compared with the standard concurrent treatment. RESULTS: Pretreatment with gemcitabine before nab-paclitaxel increased Cav-1 and albumin uptake and significantly decreased proliferation and clonogenicity compared with concurrent treatment, which correlated with increased levels of apoptosis. Cav-1 silencing reduced the uptake of albumin, and therapeutic advantage was observed when cells were pretreated with gemcitabine prior to nab-paclitaxel. In addition, we observed that pretreatment with gemcitabine resulted in partial synchronization of cells in the G2-M-phase at the time of nab-paclitaxel treatment, providing another mechanism for the benefit of altered scheduling. In heterotopic and orthotopic xenograft models, the altered schedule of gemcitabine prior to nab-paclitaxel significantly delayed tumor growth compared with concurrent delivery without added toxicity. CONCLUSIONS: Pretreatment with gemcitabine significantly increased nab-paclitaxel uptake and correlated with an increased treatment efficacy and survival benefit in preclinical models, compared with standard concurrent treatment. These results justify preclinical and clinical testing of this altered scheduling combination.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Pancreatic Ductal/drug therapy , Pancreatic Neoplasms/drug therapy , Tumor Burden/drug effects , Xenograft Model Antitumor Assays/methods , Albumins/administration & dosage , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Caveolin 1/genetics , Caveolin 1/metabolism , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Drug Administration Schedule , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Mice, Inbred NOD , Mice, Knockout , Mice, Nude , Mice, SCID , Paclitaxel/administration & dosage , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Treatment Outcome , Tumor Burden/genetics , Gemcitabine
5.
EBioMedicine ; 36: 281-292, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30269995

ABSTRACT

BACKGROUND: The goal of this study is to identify and characterize treatment resistant tumor initiating cells (TRTICs) using orthotopic xenografts. METHODS: TRTICs were enriched from GBM cell lines using mouse xenografts treated with fractionated doses of radiation and temozolomide. TRTICs were characterized by neurosphere clonogenicity and self-renewal, serial xenotransplantation, differentiation potential, and mRNA & miRNA transcriptomic profiling. We use an unbiased approach to identify antigens encoding TRTIC and glioma stem cells (GSC) populations. Co-culture experiments of TRTIC and differentiated cells were conducted to evaluate the reliance of TRTIC differentiation on the secretome of differentiated cells. FINDINGS: TRTICs acquire stem-like gene expression signatures and increased side population staining resulting from the activation of multi-drug resistance genes. Genetic and functional characterization of TRTICs shows a striking resemblance with GSCs. TRTICs can differentiate towards specific progeny in the neural stem cell lineage. TRTIC-derived tumors display all the histological hallmarks of glioblastoma (GBM) and exhibit a miRNA-transcript and mRNA-transcriptomic profile associated with aggressiveness. We report that CD24+/CD44+ antigens are expressed in TRTICs and patient-derived GSCs. Double positive CD24+/CD44+ exhibit treatment resistance and enhanced tumorigenicity. Interestingly, co-culture experiments with TRTICs and differentiated cells indicated that the regulation of TRTIC differentiation could rely on the secretome in the tumor niche. INTERPRETATION: Radiation and temozolomide treatment enriches a population of cells that have increased iPSC gene expression. As few as 500 cells produced aggressive intracranial tumors resembling patient GBM. CD24+/CD44+ antigens are increased in TRTICs and patient-derived GSCs. The enrichment for TRTICs may result in part from the secretome of differentiated cells. FUND: NIH/NCI 1RC2CA148190, 1R01CA108633, 1R01CA188228, and The Ohio State University Comprehensive Cancer Center.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Lineage/genetics , Gene Expression Regulation, Neoplastic , Induced Pluripotent Stem Cells/metabolism , Neoplasm, Residual/genetics , Neoplastic Stem Cells/metabolism , Animals , Biomarkers , Brain Neoplasms/mortality , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Disease Models, Animal , Gene Expression Profiling , Humans , Mice , Neoplastic Stem Cells/pathology , Reproducibility of Results , Transcriptome , Xenograft Model Antitumor Assays
6.
Oncotarget ; 9(18): 14597-14607, 2018 Mar 06.
Article in English | MEDLINE | ID: mdl-29581866

ABSTRACT

Glioblastoma (GBM) is an aggressive, malignant brain tumor that inevitably develops resistance to conventional chemotherapy and radiation treatments. In order to identify signaling pathways involved in the development of radiation resistance, we performed mass spectrometry-based phospho-proteomic profiling of GBM cell lines and normal human astrocytes before and after radiation treatment. We found radiation induced phosphorylation of a number of proteins including calpastatin, specifically in GBM stem cells (GSCs). Herein, we focused on calpastatin, an endogenous inhibitor of calpain proteases. Radiation-induced phosphorylation of calpastatin at Ser-633 within the inhibitory domain was validated with a phospho-specific antibody. In order to test the functional significance of phosphorylated calpastatin, we utilized site-directed mutagenesis to generate phospho-inactive (Ser633Ala) and phospho-mimetic (Ser633Glu) mutant calpastatin. GBM cell lines stably expressing the mutant calpastatin showed that phosphorylation was necessary for radiation-induced calpain activation. We also showed that casein kinase 2, a pro-survival kinase overexpressed in many cancer types, phosphorylated calpastatin at Ser-633. Our results indicate that calpastatin phosphorylation promotes radiation resistance in GBM cells by increasing the activity of calpain proteases, which are known to promote survival and invasion in cancer.

7.
Mol Cancer Ther ; 17(2): 336-346, 2018 02.
Article in English | MEDLINE | ID: mdl-28838997

ABSTRACT

Treatment refractory glioblastoma (GBM) remains a major clinical problem globally, and targeted therapies in GBM have not been promising to date. The Cancer Genome Atlas integrative analysis of GBM reported the striking finding of genetic alterations in the p53 and PI3K pathways in more than 80% of GBMs. Given the role of these pathways in making cell-fate decisions and responding to genotoxic stress, we investigated the reliance of these two pathways in mediating radiation resistance. We selected a panel of GBM cell lines and glioma stem cells (GSC) with wild-type TP53 (p53-wt) and mutant TP53, mutations known to interfere with p53 functionality (p53-mt). Cell lines were treated with a brain permeable inhibitor of P-Akt (ser473), phosphatidylinositol ether lipid analogue (PIA), with and without radiation treatment. Sensitivity to treatment was measured using Annexin-V/PI flow cytometry and Western blot analysis for the markers of apoptotic signaling, alkaline COMET assay. All results were verified in p53 isogenic cell lines. p53-mt cell lines were selectively radiosensitized by PIA. This radiosensitization effect corresponded with an increase in DNA damage and a decrease in DNA-PKcs levels. TP53 silencing in p53-wt cells showed a similar response as the p53-mt cells. In addition, the radiosensitization effects of Akt inhibition were not observed in normal human astrocytes, suggesting that this treatment strategy could have limited off-target effects. We demonstrate that the inhibition of the PI3K/Akt pathway by PIA radiosensitizes p53-mt cells by antagonizing DNA repair. In principle, this strategy could provide a large therapeutic window for the treatment of TP53-mutant tumors. Mol Cancer Ther; 17(2); 336-46. ©2017 AACRSee all articles in this MCT Focus section, "Developmental Therapeutics in Radiation Oncology."


Subject(s)
DNA Repair/drug effects , Glioblastoma/genetics , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Animals , Cell Line, Tumor , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Radiation, Ionizing , Radiation-Sensitizing Agents/pharmacology , Signal Transduction , Tumor Suppressor Protein p53/metabolism
8.
Front Oncol ; 7: 257, 2017.
Article in English | MEDLINE | ID: mdl-29164057

ABSTRACT

The present most common image diagnostic tracer in clinical practice for glioma is 18F-fluorodeoxyglucose (FDG) positron emission tomography (PET) for brain tumors diagnosis and prognosis. PET is a promising molecular imaging technique, which provides real-time information on the metabolic behavior of the tracer. The diffusive nature of glioblastoma (GBM) and heterogeneity often make the radiographic detection by FDG-PET inaccurate, and there is no gold standard. FDG-PET often leads to several controversies in making clinical decisions due to their uptake by normal surrounding tissues, and pose a challenge in delineating treatment-induced necrosis, edema, inflammation, and pseudoprogression. Thus, it is imperative to find new criteria independent of conventional morphological diagnosis to demarcate normal and tumor tissues. We have provided proof of concept studies for 11C methionine-PET (MET-PET) imaging of gliomas, along with prognostic and diagnostic significance. MET-PET is not widely used in the United States, though clinical trials from Japan and Germany suggesting the diagnostic ability of MET-PET imaging are superior to FDG-PET imaging for brain tumors. A major impediment is the availability of the onsite cyclotron and isotopic carbon chemistry facilities. In this article, we have provided the scientific rationale and advantages of the use of MET-PET as GBM tracers. We extend our discussion on the expected pitfalls of using MET-PET and ways to overcome them by incorporating a translational component of profiling gene status in the methionine metabolic pathway. This translational correlative component to the MET-PET clinical trials can lead to a better understanding of the existing controversies and can enhance our knowledge for future randomization of GBM patients based on their tumor gene signatures to achieve better prognosis and treatment outcome.

9.
Cancer Res ; 77(21): 5925-5937, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28923854

ABSTRACT

Nab-paclitaxel, a nanoparticle conjugate of paclitaxel to human albumin, exhibits efficacy in pancreatic cancer, non-small cell lung cancer and breast cancer. However, there is a lack of predictive biomarkers to identify patients who might benefit most from its administration. This study addresses this gap in knowledge by identifying that caveolin-1 (Cav-1) is a candidate mechanism-based biomarker. Caveolae are small membrane invaginations important for transendothelial albumin uptake. Cav-1, the principal structural component of caveolae, is overexpressed in the cancers noted above that respond to nab-paclitaxel. Thus, we hypothesized that Cav-1 may be critical for albumin uptake in tumors and perhaps determine their response to this drug. Cav-1 protein levels correlated positively with nab-paclitaxel sensitivity. RNAi-mediated attenuation of Cav-1 expression reduced uptake of albumin and nab-paclitaxel in cancer cells and rendered them resistant to nab-paclitaxel-induced apoptosis. Conversely, Cav-1 overexpression enhanced sensitivity to nab-paclitaxel. Selection for cellular resistance to nab-paclitaxel in cell culture correlated with a loss of Cav-1 expression. In mouse xenograft models, cancer cells, where Cav-1 was attenuated, exhibited resistance to the antitumor effects of nab-paclitaxel therapy. Overall, our findings suggest Cav-1 as a predictive biomarker for the response to nab-paclitaxel and other albumin-based cancer therapeutic drugs. Cancer Res; 77(21); 5925-37. ©2017 AACR.


Subject(s)
Albumins/metabolism , Caveolae/metabolism , Endocytosis , Neoplasms/drug therapy , Paclitaxel/pharmacology , Albumins/pharmacology , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Caveolin 1/genetics , Caveolin 1/metabolism , Cell Line, Tumor , Humans , Immunoblotting , Male , Mice, Nude , Microscopy, Confocal , Neoplasms/genetics , Neoplasms/metabolism , RNA Interference , Xenograft Model Antitumor Assays
10.
Clin Cancer Res ; 23(9): 2325-2334, 2017 May 01.
Article in English | MEDLINE | ID: mdl-27810903

ABSTRACT

Purpose: To identify potential molecular hubs that regulate oncogenic kinases and target them to improve treatment outcomes for glioblastoma patients.Experimental Design: Data mining of The Cancer Genome Atlas datasets identified nicotinamide-N-methyl transferase (NNMT) as a prognostic marker for glioblastoma, an enzyme linked to the reorganization of the methylome. We tested our hypothesis that NNMT plays a crucial role by modulating protein methylation, leading to inactivation of tumor suppressors and activation of oncogenes. Further experiments were performed to understand the underlying biochemical mechanisms using glioblastoma patient samples, established, primary, and isogenic cells.Results: We demonstrate that NNMT outcompetes leucine carboxyl methyl transferase 1 (LCMT1) for methyl transfer from principal methyl donor SAM in biological systems. Inhibiting NNMT increased the availability of methyl groups for LCMT1 to methylate PP2A, resulting in the inhibition of oncogenic serine/threonine kinases (STK). Further, NNMT inhibition retained the radiosensitizer nicotinamide and enhanced radiation sensitivity. We have provided the biochemical rationale of how NNMT plays a vital role in inhibiting tumor suppressor PP2A while concomitantly activating STKs.Conclusions: We report the intricate novel mechanism in which NNMT inhibits tumor suppressor PP2A by reorganizing the methylome both at epigenome and proteome levels and concomitantly activating prosurvival STKs. In glioblastoma tumors with NNMT expression, activation of PP2A can be accomplished by FDA approved perphenazine (PPZ), which is currently used to treat mood disorders such as schizophrenia, bipolar disorder, etc. This study forms a foundation for further glioblastoma clinical trials using PPZ with standard of care treatment. Clin Cancer Res; 23(9); 2325-34. ©2016 AACR.


Subject(s)
Carboxylic Ester Hydrolases/genetics , Glioblastoma/drug therapy , Nicotinamide N-Methyltransferase/genetics , Perphenazine/administration & dosage , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Gene Silencing , Genes, Tumor Suppressor , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Methylation/drug effects , Mice , Nicotinamide N-Methyltransferase/antagonists & inhibitors , Protein O-Methyltransferase/genetics , Protein Serine-Threonine Kinases/genetics
11.
Aging (Albany NY) ; 8(7): 1540-70, 2016 07.
Article in English | MEDLINE | ID: mdl-27425845

ABSTRACT

Various stem cell niches of the brain have differential requirements for Cyclin A2. Cyclin A2 loss results in marked cerebellar dysmorphia, whereas forebrain growth is retarded during early embryonic development yet achieves normal size at birth. To understand the differential requirements of distinct brain regions for Cyclin A2, we utilized neuroanatomical, transgenic mouse, and mathematical modeling techniques to generate testable hypotheses that provide insight into how Cyclin A2 loss results in compensatory forebrain growth during late embryonic development. Using unbiased measurements of the forebrain stem cell niche, we parameterized a mathematical model whereby logistic growth instructs progenitor cells as to the cell-types of their progeny. Our data was consistent with prior findings that progenitors proliferate along an auto-inhibitory growth curve. The growth retardation inCCNA2-null brains corresponded to cell cycle lengthening, imposing a developmental delay. We hypothesized that Cyclin A2 regulates DNA repair and that CCNA2-null progenitors thus experienced lengthened cell cycle. We demonstrate that CCNA2-null progenitors suffer abnormal DNA repair, and implicate Cyclin A2 in double-strand break repair. Cyclin A2's DNA repair functions are conserved among cell lines, neural progenitors, and hippocampal neurons. We further demonstrate that neuronal CCNA2 ablation results in learning and memory deficits in aged mice.


Subject(s)
Aging/physiology , Brain/metabolism , Cell Cycle/genetics , Cyclin A2/metabolism , Neurons/metabolism , Animals , Behavior, Animal/physiology , Conditioning, Psychological/physiology , Cyclin A2/genetics , DNA Repair , Hand Strength/physiology , Mice , Mice, Transgenic , Models, Biological , Motor Skills/physiology , Social Behavior , Stem Cell Niche
12.
Clin Cancer Res ; 22(14): 3513-23, 2016 07 15.
Article in English | MEDLINE | ID: mdl-26936918

ABSTRACT

PURPOSE: We employed a metabolomics-based approach with the goal to better understand the molecular signatures of glioblastoma cells and tissues, with an aim toward identifying potential targetable biomarkers for developing more effective and novel therapies. EXPERIMENTAL DESIGN: We used liquid chromatography coupled with mass spectrometry (LC-MS/Q-TOF and LC-MS/QQQ) for the discovery and validation of metabolites from primary and established glioblastoma cells, glioblastoma tissues, and normal human astrocytes. RESULTS: We identified tryptophan, methionine, kynurenine, and 5-methylthioadenosine as differentially regulated metabolites (DRM) in glioblastoma cells compared with normal human astrocytes (NHAs). Unlike NHAs, glioblastoma cells depend on dietary methionine for proliferation, colony formation, survival, and to maintain a deregulated methylome (SAM:SAH ratio). In methylthioadenosine phosphorylase (MTAP)-deficient glioblastoma cells, expression of MTAP transgene did not alter methionine dependency, but compromised tumor growth in vivo We discovered that a lack of the kynurenine-metabolizing enzymes kynurenine monooxygenase and/or kynureninase promotes the accumulation of kynurenine, which triggers immune evasion in glioblastoma cells. In silico analysis of the identified DRMs mapped the activation of key oncogenic kinases that promotes tumorigenesis in glioblastoma. We validated this result by demonstrating that the exogenous addition of DRMs to glioblastoma cells in vitro results in oncogene activation as well as the simultaneous downregulation of Ser/Thr phosphatase PP2A. CONCLUSIONS: We have connected a four-metabolite signature, implicated in the methionine and kynurenine pathways, to the promotion and maintenance of glioblastoma. Together, our data suggest that these metabolites and their respective metabolic pathways serve as potential therapeutic targets for glioblastoma. Clin Cancer Res; 22(14); 3513-23. ©2016 AACR.


Subject(s)
Cell Proliferation/physiology , Glioblastoma/metabolism , Glioblastoma/pathology , Kynurenine/metabolism , Methionine/metabolism , Oncogenes/genetics , Astrocytes/metabolism , Astrocytes/pathology , Biomarkers, Tumor/metabolism , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Line , Chromatography, High Pressure Liquid/methods , Humans , Mass Spectrometry/methods , Metabolomics/methods , Purine-Nucleoside Phosphorylase/metabolism , Tryptophan/metabolism , Tumor Cells, Cultured
13.
Neuro Oncol ; 16(10): 1354-64, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24879047

ABSTRACT

BACKGROUNDS: Piperlongumine, a natural plant product, kills multiple cancer types with little effect on normal cells. Piperlongumine raises intracellular levels of reactive oxygen species (ROS), a phenomenon that may underlie the cancer-cell killing. Although these findings suggest that piperlongumine could be useful for treating cancers, the mechanism by which the drug selectively kills cancer cells remains unknown. METHODS: We treated multiple high-grade glioma (HGG) sphere cultures with piperlongumine and assessed its effects on ROS and cell-growth levels as well as changes in downstream signaling. We also examined the levels of putative piperlongumine targets and their roles in HGG cell growth. RESULTS: Piperlongumine treatment increased ROS levels and preferentially killed HGG cells with little effect in normal brain cells. Piperlongumine reportedly increases ROS levels after interactions with several redox regulators. We found that HGG cells expressed higher levels of the putative piperlongumine targets than did normal neural stem cells (NSCs). Furthermore, piperlongumine treatment in HGG cells, but not in normal NSCs, increased oxidative inactivation of peroxiredoxin 4 (PRDX4), an ROS-reducing enzyme that is overexpressed in HGGs and facilitates proper protein folding in the endoplasmic reticulum (ER). Moreover, piperlongumine exacerbated intracellular ER stress, an effect that was mimicked by suppressing PRDX4 expression. CONCLUSIONS: Our results reveal that the mechanism by which piperlongumine preferentially kills HGG cells involves PRDX4 inactivation, thereby inducing ER stress. Therefore, piperlongumine treatment could be considered as a novel therapeutic option for HGG treatment.


Subject(s)
Antineoplastic Agents/administration & dosage , Brain Neoplasms/drug therapy , Dioxolanes/administration & dosage , Endoplasmic Reticulum Stress/drug effects , Glioma/drug therapy , Peroxiredoxins/metabolism , Animals , Apoptosis/drug effects , Brain Neoplasms/metabolism , Brain Neoplasms/mortality , Databases, Factual , Glioma/metabolism , Glioma/mortality , Humans , Mice , Reactive Oxygen Species/metabolism , Survival Analysis , Tumor Cells, Cultured
14.
Clin Cancer Res ; 20(14): 3884-95, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24787670

ABSTRACT

PURPOSE: Alveolar rhabdomyosarcoma that harbors the PAX3-FOXO1 fusion gene (t-ARMS) is a common and lethal subtype of this childhood malignancy. Improvement in clinical outcomes in this disease is predicated upon the identification of novel therapeutic targets. EXPERIMENTAL DESIGN: Robust mouse models were used for in vivo analysis, and molecular studies were performed on xenografts treated in parallel. Two independent patient sets (n = 101 and 124) of clinically annotated tumor specimens were used for analysis of FANCD2 levels and its association with clinical and molecular characteristics and outcomes. RESULTS: Our xenograft studies reveal a selective suppression of FANCD2 by m-TOR kinase inhibition and radiosensitization of the t-ARMS line only. In the initial patient set, we show that FANCD2 transcript levels are prognostic in univariate analysis, and are significantly associated with metastatic disease and that the copresence of the translocation and high expression of FANCD2 is independently prognostic. We also demonstrate a significant and nonrandom enrichment of mTOR-associated genes that correlate with FANCD2 gene expression within the t-ARMS samples, but not within other cases. In the second patient set, we show that on a protein level, FANCD2 expression correlates with PAX3-FOXO1 fusion gene and is strongly associated with phospho-P70S6K expression in cases with the fusion gene. CONCLUSIONS: Our data demonstrate that FANCD2 may have a significant role in the radiation resistance and virulence of t-ARMS. Indirectly targeting this DNA repair protein, through mTOR inhibition, may represent a novel and selective treatment strategy.


Subject(s)
Biomarkers, Tumor/metabolism , Fanconi Anemia Complementation Group D2 Protein/physiology , Oncogene Proteins, Fusion/metabolism , Paired Box Transcription Factors/metabolism , Rhabdomyosarcoma, Alveolar/metabolism , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Chemoradiotherapy , Female , Humans , Mice, SCID , Morpholines/pharmacology , Radiation Tolerance , Rhabdomyosarcoma, Alveolar/drug therapy , Rhabdomyosarcoma, Alveolar/radiotherapy , Signal Transduction , Xenograft Model Antitumor Assays
15.
PLoS One ; 8(5): e64393, 2013.
Article in English | MEDLINE | ID: mdl-23696889

ABSTRACT

MicroRNAs regulate several aspects of tumorigenesis and cancer progression. Most cancer tissues are archived formalin-fixed and paraffin-embedded (FFPE). While microRNAs are a more stable form of RNA thought to withstand FFPE-processing and degradation there is only limited evidence for the latter assumption. We examined whether microRNA profiling can be successfully conducted on FFPE cancer tissues using SOLiD ligation based sequencing. Tissue storage times (2-9 years) appeared to not affect the number of detected microRNAs in FFPE samples compared to matched frozen samples (paired t-test p>0.7). Correlations of microRNA expression values were very high across microRNAs in a given sample (Pearson's r = 0.71-0.95). Higher variance of expression values among samples was associated with higher correlation coefficients between FFPE and frozen tissues. One of the FFPE samples in this study was degraded for unknown reasons with a peak read length of 17 nucleotides compared to 21 in all other samples. The number of detected microRNAs in this sample was within the range of microRNAs detected in all other samples. Ligation-based microRNA deep sequencing on FFPE cancer tissues is feasible and RNA degradation to the degree observed in our study appears to not affect the number of microRNAs that can be quantified.


Subject(s)
Cryopreservation/methods , Formaldehyde/chemistry , Gene Expression Profiling/methods , High-Throughput Nucleotide Sequencing/methods , MicroRNAs/genetics , Paraffin Embedding/methods , Tissue Fixation/methods , Adult , Aged , Female , Humans , In Vitro Techniques , Male , Middle Aged
16.
J Neurooncol ; 110(1): 37-48, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22875706

ABSTRACT

Glioblastomas are among the most aggressive human cancers, and prognosis remains poor despite presently available therapies. Angiogenesis is a hallmark of glioblastoma, and the resultant vascularity is associated with poor prognosis. The proteins that mediate angiogenesis, including vascular endothelial growth factor (VEGF) signaling proteins, have emerged as attractive targets for therapeutic development. Since VEGF receptor-2 (VEGFR-2) is thought to be the primary receptor mediating angiogenesis, direct inhibition of this receptor may produce an ideal therapeutic effect. In this context, we tested the therapeutic effect of CT322, a selective inhibitor of VEGFR-2. Using an intracranial murine xenograft model (U87-EGFRvIII-luciferase), we demonstrate that CT322 inhibited glioblastoma growth in vivo and prolonged survival. Of note, the anti-neoplastic effect of CT322 is augmented by the incorporation of temozolomide or temozolomide with radiation therapy. Immunohistochemical analysis of CT322 treated tumors revealed decreased CD31 staining, suggesting that the tumoricidal effect is mediated by inhibition of angiogenesis. These pre-clinical results provide the foundation to further understand long term response and tumor escape mechanisms to anti-angiogenic treatments on EGFR over-expressing glioblastomas.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Brain Neoplasms/therapy , Chemoradiotherapy/methods , Fibronectins/pharmacology , Glioma/therapy , Peptide Fragments/pharmacology , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Animals , Brain Neoplasms/pathology , Cell Line, Tumor , Dacarbazine/administration & dosage , Dacarbazine/analogs & derivatives , Glioma/pathology , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Temozolomide , Xenograft Model Antitumor Assays
17.
PLoS One ; 7(8): e42818, 2012.
Article in English | MEDLINE | ID: mdl-22916164

ABSTRACT

Glioblastoma multiforme (GBM), the most common and aggressive primary brain malignancy, is incurable despite the best combination of current cancer therapies. For the development of more effective therapies, discovery of novel candidate tumor drivers is urgently needed. Here, we report that peroxiredoxin 4 (PRDX4) is a putative tumor driver. PRDX4 levels were highly increased in a majority of human GBMs as well as in a mouse model of GBM. Reducing PRDX4 expression significantly decreased GBM cell growth and radiation resistance in vitro with increased levels of ROS, DNA damage, and apoptosis. In a syngenic orthotopic transplantation model, Prdx4 knockdown limited GBM infiltration and significantly prolonged mouse survival. These data suggest that PRDX4 can be a novel target for GBM therapies in the future.


Subject(s)
Apoptosis , Cell Division , Glioblastoma/pathology , Peroxiredoxins/metabolism , Animals , DNA Damage , Disease Models, Animal , Glioblastoma/metabolism , Humans , Mice , Reactive Oxygen Species/metabolism , Up-Regulation
18.
Neuro Oncol ; 14(4): 405-15, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22351749

ABSTRACT

FTY720 is a sphingosine analogue that down regulates expression of sphingosine-1-phosphate receptors and causes apoptosis of multiple tumor cell types, including glioma cells. This study examined the effect of FTY720 on brain tumor stem cells (BTSCs) derived from human glioblastoma (GBM) tissue. FTY720 treatment of BTSCs led to rapid inactivation of ERK MAP kinase, leading to upregulation of the BH3-only protein Bim and apoptosis. In combination with temozolomide (TMZ), the current standard chemotherapeutic agent for GBM, FTY720 synergistically induced BTSC apoptosis. FTY720 also slowed growth of intracranial xenograft tumors in nude mice and augmented the therapeutic effect of TMZ, leading to enhanced survival. Furthermore, the combination of FTY720 and TMZ decreased the invasiveness of BTSCs in mouse brains. FTY720 is known to cross the blood-brain barrier and recently received Food and Drug Administration approval for treatment of relapsing multiple sclerosis. Thus, FTY720 is an excellent potential therapeutic agent for treatment of GBM.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis Regulatory Proteins/drug effects , Extracellular Signal-Regulated MAP Kinases/drug effects , Membrane Proteins/drug effects , Neoplastic Stem Cells/drug effects , Propylene Glycols/pharmacology , Proto-Oncogene Proteins/drug effects , Receptors, Lysosphingolipid/drug effects , Sphingosine/analogs & derivatives , Animals , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Bcl-2-Like Protein 11 , Brain Neoplasms/drug therapy , Dacarbazine/administration & dosage , Dacarbazine/analogs & derivatives , Fingolimod Hydrochloride , Glioblastoma/drug therapy , Humans , Induction Chemotherapy , Membrane Proteins/metabolism , Mice , Mice, Nude , Propylene Glycols/therapeutic use , Proto-Oncogene Proteins/metabolism , Sphingosine/pharmacology , Sphingosine/therapeutic use , Temozolomide , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
19.
J Inorg Biochem ; 106(1): 32-42, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22112837

ABSTRACT

Gold(III) compounds have been recognized as anticancer agents due to their structural and electronic similarities with currently employed platinum(II) species. An added benefit to gold(III) agents is the ability to overcome cisplatin resistance. This work identified four gold(III) compounds, [Au(Phen)Cl(2)]PF(6), [Au(DPQ)Cl(2)]PF(6), [Au(DPPZ)Cl(2)]PF(6), and [Au(DPQC)Cl(2)]PF(6), (Phen = 1,10-phenanthroline, DPQ = dipyrido[3,2-d:2',3'-f]quinoxaline, DPPZ = dipyrido[3,2-a:2',3'-c] phenazine, DPQC = dipyrido[3,2-d:2',3'-f] cyclohexyl quinoxaline) that exhibited anticancer activity in both cisplatin sensitive and cisplatin resistant ovarian cancer cells. Two of these compounds, [Au(DPQ)Cl(2)]PF(6) (AQ) and [Au(DPPZ)Cl(2)]PF(6) (AZ), displayed exceptional anticancer activity and were the focus of more intensive mechanistic study. At the molecular level, AQ and AZ formed DNA adducts, generated free radicals, and upregulated pro-apoptotic signaling molecules (p53, caspases, PARP, death effectors). Taken together, these two novel gold(III) polypyridyl complexes exhibit potent antitumor activity in cisplatin resistant cancer cells. These activities may be mediated, in part, by the activation of apoptotic signaling.


Subject(s)
Antineoplastic Agents/chemistry , Cisplatin/chemistry , Gold/chemistry , Organometallic Compounds/chemistry , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Blotting, Western , Caspases/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Circular Dichroism , Cisplatin/pharmacology , DNA Adducts/chemistry , DNA, Neoplasm/chemistry , Drug Resistance, Neoplasm/drug effects , Female , Humans , Inhibitory Concentration 50 , Molecular Mimicry , Molecular Structure , Organometallic Compounds/pharmacology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Poly(ADP-ribose) Polymerases/metabolism , Reactive Oxygen Species/metabolism , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...