Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 78
Filter
1.
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
2.
Cancer Cell Int ; 22(1): 389, 2022 Dec 08.
Article in English | MEDLINE | ID: mdl-36482431

ABSTRACT

BACKGROUND: The invasive nature of GBM combined with the diversity of brain microenvironments creates the potential for a topographic heterogeneity in GBM radioresponse. Investigating the mechanisms responsible for a microenvironment-induced differential GBM response to radiation may provide insights into the molecules and processes mediating GBM radioresistance. METHODS: Using a model system in which human GBM stem-like cells implanted into the right striatum of nude mice migrate throughout the right hemisphere (RH) to the olfactory bulb (OB), the radiation-induced DNA damage response was evaluated in each location according to γH2AX and 53BP1 foci and cell cycle phase distribution as determined by flow cytometry and immunohistochemistry. RNAseq was used to compare transcriptomes of tumor cells growing in the OB and the RH. Protein expression and neuron-tumor interaction were defined by immunohistochemistry and confocal microscopy. RESULTS: After irradiation, there was a more rapid dispersal of γH2AX and 53BP1 foci in the OB versus in the RH, indicative of increased double strand break repair capacity in the OB and consistent with the OB providing a radioprotective niche. With respect to the cell cycle, by 6 h after irradiation there was a significant loss of mitotic tumor cells in both locations suggesting a similar activation of the G2/M checkpoint. However, by 24 h post-irradiation there was an accumulation of G2 phase cells in the OB, which continued out to at least 96 h. Transcriptome analysis showed that tumor cells in the OB had higher expression levels of DNA repair genes involved in non-homologous end joining and genes related to the spindle assembly checkpoint. Tumor cells in the OB were also found to have an increased frequency of soma-soma contact with neurons. CONCLUSION: GBM cells that have migrated to the OB have an increased capacity to repair radiation-induced double strand breaks and altered cell cycle regulation. These results correspond to an upregulation of genes involved in DNA damage repair and cell cycle control. Because the murine OB provides a source of radioresistant tumor cells not evident in other experimental systems, it may serve as a model for investigating the mechanisms mediating GBM radioresistance.

3.
Mol Cancer Ther ; 21(9): 1406-1414, 2022 09 06.
Article in English | MEDLINE | ID: mdl-35732578

ABSTRACT

A fundamental component of cellular radioresponse is the translational control of gene expression. Because a critical regulator of translational control is the eukaryotic translation initiation factor 4F (eIF4F) cap binding complex, we investigated whether eIF4A, the RNA helicase component of eIF4F, can serve as a target for radiosensitization. Knockdown of eIF4A using siRNA reduced translational efficiency, as determined from polysome profiles, and enhanced tumor cell radiosensitivity as determined by clonogenic survival. The increased radiosensitivity was accompanied by a delayed dispersion of radiation-induced γH2AX foci, suggestive of an inhibition of DNA double-strand break repair. Studies were then extended to (-)-SDS-1-021, a pharmacologic inhibitor of eIF4A. Treatment of cells with the rocaglate (-)-SDS-1-021 resulted in a decrease in translational efficiency as well as protein synthesis. (-)-SDS-1-021 treatment also enhanced the radiosensitivity of tumor cell lines. This (-)-SDS-1-021-induced radiosensitization was accompanied by a delay in radiation-induced γH2AX foci dispersal, consistent with a causative role for the inhibition of double-strand break repair. In contrast, although (-)-SDS-1-021 inhibited translation and protein synthesis in a normal fibroblast cell line, it had no effect on radiosensitivity of normal cells. Subcutaneous xenografts were then used to evaluate the in vivo response to (-)-SDS-1-021 and radiation. Treatment of mice bearing subcutaneous xenografts with (-)-SDS-1-021 decreased tumor translational efficiency as determined by polysome profiles. Although (-)-SDS-1-021 treatment alone had no effect on tumor growth, it significantly enhanced the radiation-induced growth delay. These results suggest that eIF4A is a tumor-selective target for radiosensitization.


Subject(s)
Eukaryotic Initiation Factor-4F , Neoplasms , Radiation Tolerance , Animals , Cell Line, Tumor , DNA Breaks, Double-Stranded , Eukaryotic Initiation Factor-4F/antagonists & inhibitors , Humans , Mice , Neoplasms/radiotherapy , Xenograft Model Antitumor Assays
4.
Sci Rep ; 12(1): 4059, 2022 03 08.
Article in English | MEDLINE | ID: mdl-35260696

ABSTRACT

Increased ribosome biogenesis is a distinguishing feature of cancer cells, and small molecule inhibitors of ribosome biogenesis are currently in clinical trials as single agent therapy. It has been previously shown that inhibiting ribosome biogenesis through the inhibition of nuclear export of ribosomal subunits sensitizes tumor cells to radiotherapy. In this study, the radiosensitizing potential of CX-5461, a small molecule inhibitor of RNA polymerase I, was tested. Radiosensitization was measured by clonogenic survival assay in a panel of four tumor cell lines derived from three different tumor types commonly treated with radiation. 50 nM CX-5461 radiosensitized PANC-1, U251, HeLa, and PSN1 cells with dose enhancement factors in the range of 1.2-1.3. However, 50 nM CX-5461 was not sufficient to inhibit 45S transcription alone or in combination with radiation. The mechanism of cell death with the combination of CX-5461 and radiation occurred through mitotic catastrophe and not apoptosis. CX-5461 inhibited the repair and/or enhanced the initial levels of radiation-induced DNA double strand breaks. Understanding the mechanism of CX-5461-induced radiosensitization should be of value in the potential application of the CX-5461/radiotherapy combination in cancer treatment.


Subject(s)
Benzothiazoles , Naphthyridines , RNA Polymerase I , Radiation-Sensitizing Agents , Apoptosis , Benzothiazoles/pharmacology , Cell Line, Tumor , DNA Breaks, Double-Stranded , DNA Damage , Humans , Naphthyridines/pharmacology , RNA Polymerase I/antagonists & inhibitors , Radiation-Sensitizing Agents/pharmacology
5.
Int J Mol Sci ; 22(19)2021 Oct 01.
Article in English | MEDLINE | ID: mdl-34639005

ABSTRACT

Towards improving the efficacy of radiotherapy, one approach is to target the molecules and processes mediating cellular radioresponse. Along these lines, translational control of gene expression has been established as a fundamental component of cellular radioresponse, which suggests that the molecules participating in this process (i.e., the translational machinery) can serve as determinants of radiosensitivity. Moreover, the proteins comprising the translational machinery are often overexpressed in tumor cells suggesting the potential for tumor specific radiosensitization. Studies to date have shown that inhibiting proteins involved in translation initiation, the rate-limiting step in translation, specifically the three members of the eIF4F cap binding complex eIF4E, eIF4G, and eIF4A as well as the cap binding regulatory kinases mTOR and Mnk1/2, results in the radiosensitization of tumor cells. Because ribosomes are required for translation initiation, inhibiting ribosome biogenesis also appears to be a strategy for radiosensitization. In general, the radiosensitization induced by targeting the translation initiation machinery involves inhibition of DNA repair, which appears to be the consequence of a reduced expression of proteins critical to radioresponse. The availability of clinically relevant inhibitors of this component of the translational machinery suggests opportunities to extend this approach to radiosensitization to patient care.


Subject(s)
Biomarkers, Tumor , Neoplasms/genetics , Peptide Chain Initiation, Translational/radiation effects , Protein Biosynthesis/radiation effects , Radiation Tolerance/genetics , Animals , Eukaryotic Initiation Factors/metabolism , Gene Expression Regulation, Neoplastic , Humans , Neoplasms/metabolism , Neoplasms/radiotherapy , Protein Processing, Post-Translational , Radiotherapy , Ribosomes/metabolism , Signal Transduction
6.
Mol Cancer Ther ; 20(9): 1672-1679, 2021 09.
Article in English | MEDLINE | ID: mdl-34158343

ABSTRACT

AZD0530, a potent small-molecule inhibitor of the Src kinase family, is an anticancer drug used in the treatment of various cancers. In the case of glioblastoma (GBM), where resistance to radiotherapy frequently occurs, Src kinase is known as one of the molecules responsible for imparting radioresistance to GBM. Thus, we evaluated the effect of AZD0530 on the radiosensitivity of human GBM cells and human glioblastoma stem-like cells (GSCs). We show that Src activity of GBM and GSC is increased by radiation and inhibited by AZD0530, and using clonogenic assays, AZD0530 enhances the radiosensitivity of GBM and GSCs. Also, AZD0530 induced a prolongation of radiation-induced γH2AX without specific cell cycle and mitotic index changes, suggesting that AZD0530-induced radiosensitization in GBM cells and GSCs results from the inhibition of DNA repair. In addition, AZD0530 was shown to inhibit the radiation-induced EGFR/PI3K/AKT pathway, which is known to promote and regulate radioresistance and survival of GBM cells by radiation. Finally, mice bearing orthotopic xenografts initiated from GBM cells were then used to evaluate the in vivo response to AZD0530 and radiation. The combination of AZD0530 and radiation showed the longest median survival compared with any single modality. Thus, these results show that AZD0530 enhances the radiosensitivity of GBM cells and GSCs and suggest the possibility of AZD0530 as a clinical radiosensitizer for treatment of GBM.


Subject(s)
Benzodioxoles/pharmacology , Gene Expression Regulation, Neoplastic , Glioblastoma/radiotherapy , Neoplastic Stem Cells/radiation effects , Quinazolines/pharmacology , Radiation Tolerance/drug effects , Radiation-Sensitizing Agents/pharmacology , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Cell Cycle , Cell Proliferation , Female , Glioblastoma/drug therapy , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Mice , Mice, Nude , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
7.
Methods Mol Biol ; 2269: 37-47, 2021.
Article in English | MEDLINE | ID: mdl-33687670

ABSTRACT

Ionizing radiation is a critical component of glioblastoma (GBM) therapy. Recent data have implicated glioblastoma stem-like cells (GSCs) as determinants of GBM development, maintenance, and treatment response. Understanding the response of GSCs to radiation should thus provide insight into the development of improved GBM treatment strategies. Towards this end, in vitro techniques for the analysis of GSC radiosensitivity are an essential starting point. One such method, the clonogenic survival assay has been adapted to assessing the intrinsic radiosensitivity of GSCs and is described here. As an alternative method, the limiting dilution assay is presented for defining the radiosensitivity of GSC lines that do not form colonies or only grow as neurospheres. In addition to these cellular strategies, we describe γH2AX foci analysis, which provides a surrogate marker for radiosensitivity at the molecular level. Taken together, the in vitro methods presented here provide tools for defining intrinsic radiosensitivity of GSCs and for testing agents that may enhance GBM radioresponse.


Subject(s)
Biomarkers, Tumor , Genetic Loci , Glioblastoma , Histones , Neoplasm Proteins , Neoplastic Stem Cells , Radiation Tolerance , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/pathology , Glioblastoma/radiotherapy , Histones/genetics , Histones/metabolism , Humans , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology
8.
Cancers (Basel) ; 12(11)2020 Nov 04.
Article in English | MEDLINE | ID: mdl-33158243

ABSTRACT

Radiation therapy is a mainstay in the standard of care for glioblastoma (GBM), thus inhibiting the DNA damage response (DDR) is a major strategy to improve radiation response and therapeutic outcomes. Small interfering RNA (siRNA) therapy holds immeasurable potential for the treatment of GBM, however delivery of the siRNA payload remains the largest obstacle for clinical implementation. Here we demonstrate the effectiveness of the novel nanomaterial, ECO (1-aminoethylimino[bis(N-oleoylcysteinylaminoethyl) propionamide]), to deliver siRNA targeting DDR proteins ataxia telangiectasia mutated and DNA-dependent protein kinase (DNApk-cs) for the radiosensitzation of GBM in vitro and in vivo. ECO nanoparticles (NPs) were shown to efficiently deliver siRNA and silence target protein expression in glioma (U251) and glioma stem cell lines (NSC11, GBMJ1). Importantly, ECO NPs displayed no cytotoxicity and minimal silencing of genes in normal astrocytes. Treatment with ECO/siRNA NPs and radiation resulted in the prolonged presence of γH2AX foci, indicators of DNA damage, and increased radiosensitivity in all tumor cell lines. In vivo, intratumoral injection of ECO/siDNApk-cs NPs with radiation resulted in a significant increase in survival compared with injection of NPs alone. These data suggest the ECO nanomaterial can effectively deliver siRNA to more selectively target and radiosensitize tumor cells to improve therapeutic outcomes in GBM.

9.
J Neurooncol ; 149(3): 383-390, 2020 Sep.
Article in English | MEDLINE | ID: mdl-33057920

ABSTRACT

PURPOSE: Glioblastoma (GBM) is characterized by extensive clonal diversity suggesting the presence of tumor cells with varying degrees of treatment sensitivity. Radiotherapy is an integral part of glioblastoma treatment. Whether GBMs are comprised of spatially distinct cellular populations with uniform or varying degrees of radiosensitivity has not been established. METHODS: Spatially distinct regions of three GBMs (J3, J7 and J14) were resected and unique cell lines were derived from each region. DNA from cell lines, corresponding tumor fragments, and patient blood was extracted for whole exome sequencing. Variants, clonal composition, and functional implications were compared and analyzed with superFreq and IPA. Limiting dilution assays were performed on cell lines to measure intrinsic radiosensitivity. RESULTS: Based on WES, cell lines generated from different regions of the same tumor were more closely correlated with their tumor of origin than the other GBMs. Variant and clonal composition comparisons showed that cell lines from distinct tumors displayed increasing levels of ITH with J3 and J14 having the lowest and highest, respectively. The radiosensitivities of the cell lines generated from the J3 tumor were similar as were those generated from the J7 tumor. However, the radiosensitivities of the 2 cell lines generated from the J14 tumor (J14T3 and J14T6) were significantly different with J14T6 being more sensitive than J14T3. CONCLUSION: Data suggest a tumor dependent ITH in radiosensitivity. The existence of ITH in radiosensitivity may impact not only the initial therapeutic response but also the effectiveness of retreatment protocols.


Subject(s)
Biomarkers, Tumor/genetics , Exome Sequencing/methods , Gene Expression Regulation, Neoplastic/radiation effects , Glioblastoma/pathology , Mutation , Radiation Tolerance , Glioblastoma/genetics , Glioblastoma/radiotherapy , Humans , Prognosis , Tumor Cells, Cultured
10.
Int J Radiat Oncol Biol Phys ; 107(1): 194-201, 2020 05 01.
Article in English | MEDLINE | ID: mdl-31987963

ABSTRACT

PURPOSE: The various microenvironments that exist within the brain combined with the invasive nature of glioblastoma (GBM) creates the potential for a topographic influence on tumor cell radiosensitivity. The aim of this study was to determine whether specific brain microenvironments differentially influence tumor cell radioresponse. METHODS AND MATERIALS: GBM stem-like cells were implanted into the right striatum of nude mice. To measure radiosensitivity, proliferation status of individual tumor cells was determined according to the incorporation of 5-chloro-2'-deoxyuridine delivered at 4, 12, and 20 days after brain irradiation. As an additional measure of radiosensitivity, the percentage of human cells in the right hemisphere and the olfactory bulb were defined using digital droplet polymerase chain reaction. Targeted gene expression profiling was accomplished using NanoString analysis. RESULTS: Tumor cells were detected throughout the striatum, corpus callosum, and olfactory bulb. After an initial loss of proliferating tumor cells in the corpus callosum and striatum after irradiation, there was only a minor recovery by 20 days. In contrast, the proliferation of tumor cells located in the olfactory bulb began to recover at 4 days and returned to unirradiated levels by day 12 postirradiation. The percentage of human cells in the right hemisphere and the olfactory bulb after irradiation also suggested that the tumor cells in the olfactory bulb were relatively radioresistant. Gene expression profiling identified consistent differences between tumor cells residing in the olfactory bulb and those in the right hemisphere. CONCLUSIONS: These results suggest that the olfactory bulb provides a radioresistant niche for GBM cells.


Subject(s)
Glioblastoma/pathology , Olfactory Bulb/pathology , Olfactory Bulb/radiation effects , Radiation Tolerance , Stem Cell Niche/radiation effects , Animals , Mice , Tumor Microenvironment/radiation effects
11.
Cancer Res ; 79(23): 6032-6043, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31615806

ABSTRACT

A consequence of the intratumor heterogeneity (ITH) of glioblastoma (GBM) is the susceptibility to treatment-driven evolution. To determine the potential of radiotherapy to influence GBM evolution, we used orthotopic xenografts initiated from CD133+ GBM stem-like cells (GSC). Toward this end, orthotopic xenografts grown in nude mice were exposed to a fractionated radiation protocol, which resulted in a significant increase in animal survival. Brain tumors from control and irradiated mice were then collected at morbidity and compared in terms of growth pattern, clonal diversity, and genomic architecture. In mice that received fractionated radiation, tumors were less invasive, with more clearly demarcated borders and tumor core hypercellularity as compared with controls, suggesting a fundamental change in tumor biology. Viral integration site analysis indicated a reduction in clonal diversity in the irradiated tumors, implying a decrease in ITH. Changes in clonal diversity were not detected after irradiation of GSCs in vitro, suggesting that the radiation-induced reduction in ITH was dependent on the brain microenvironment. Whole-exome sequencing revealed differences in mutation patterns between control and irradiated tumors, which included modifications in the presence and clonality of driver mutations associated with GBM. Moreover, changes in the distribution of mutations as a function of subpopulation size between control and irradiated tumors were consistent with subclone expansion and contraction, that is, subpopulation evolution. Taken together, these results indicate that radiation drives the evolution of the GSC-initiated orthotopic xenografts and suggest that radiation-driven evolution may have therapeutic implications for recurrent GBM. SIGNIFICANCE: Radiation drives the evolution of glioblastoma orthotopic xenografts; when translated to the clinic, this may have therapeutic implications for recurrent tumors.


Subject(s)
Brain Neoplasms/radiotherapy , Evolution, Molecular , Genetic Heterogeneity/radiation effects , Glioblastoma/radiotherapy , Neoplastic Stem Cells/radiation effects , Animals , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , DNA Mutational Analysis , Female , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Mice , Mutation/radiation effects , Neoplastic Stem Cells/pathology , Radiation Tolerance/genetics , Tumor Microenvironment/genetics , Tumor Microenvironment/radiation effects , Exome Sequencing , Xenograft Model Antitumor Assays
12.
Neurooncol Pract ; 5(4): 246-250, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30402263

ABSTRACT

BACKGROUND: Valproic acid (VPA) is an antiepileptic agent with histone deacetylase inhibitor activity shown to enhance overall survival and progression free survival in patients with newly diagnosed glioblastoma (GBM). This reports on the late toxicity of the VPA/radiotherapy (RT)/temozolomide (TMZ) combination in the long-term survivors of a phase 2 study evaluating this regimen. METHODS: 37 patients with newly diagnosed GBM were initially enrolled on this trial and received combination therapy. VPA/RT/TMZ related late toxicities were evaluated in the 6 patients that lived greater than 3 years using the Cancer Therapy and Evaluation Program Common Toxicity Criteria (CTC) Version 4.0 for toxicity and adverse event reporting as well as the RTOG/EORTC Radiation Morbidity Scoring Scheme. RESULTS: The median duration of follow-up for these 6 patients was 69.5m. In this cohort, the median OS was 73.8m (60.8-103.8m) and median PFS was 53.1m (37.3 - 103.8m). The most common late toxicity of VPA in conjunction with RT/TMZ were the CTC classifications of neurological, pain, and blood/ bone marrow toxicity and most were grade 1/2. There were only two grade 3/4 toxicities. CONCLUSIONS: The addition of VPA to concurrent RT/TMZ in patients with newly diagnosed GBM was well tolerated with little late toxicity. Additionally, VPA may result in improved outcomes as compared to historical data and merits further study.

13.
Mol Cancer Ther ; 17(8): 1717-1726, 2018 08.
Article in English | MEDLINE | ID: mdl-29866745

ABSTRACT

Analysis of the radiation-induced translatome of glioblastoma stem-like cells (GSC) identified an interacting network in which XPO1 serves as a major hub protein. To determine whether this nuclear export protein provides a target for radiosensitization, we defined the effects of clinically relevant XPO1 inhibitor selinexor on the radiosensitivity of glioblastoma cells. As determined by clonogenic survival analysis, selinexor enhanced the radiosensitivity of GSCs but not normal fibroblast cell lines. On the basis of γH2AX foci and neutral comet analyses, selinexor inhibited the repair of radiation-induced DNA double-strand breaks in GSCs, suggesting that the selinexor-induced radiosensitization is mediated by an inhibition of DNA repair. Consistent with a role for XPO1 in the nuclear to cytoplasm export of rRNA, selinexor reduced 5S and 18S rRNA nuclear export in GSCs, which was accompanied by a decrease in gene translation efficiency, as determined from polysome profiles, as well as in protein synthesis. In contrast, rRNA nuclear export and protein synthesis were not reduced in normal cells treated with selinexor. Orthotopic xenografts initiated from a GSC line were then used to define the in vivo response to selinexor and radiation. Treatment of mice bearing orthotopic xenografts with selinexor decreased tumor translational efficiency as determined from polysome profiles. Although selinexor treatment alone had no effect on the survival of mice with brain tumors, it significantly enhanced the radiation-induced prolongation of survival. These results indicate that selinexor enhances the radiosensitivity of glioblastoma cells and suggest that this effect involves the global inhibition of gene translation. Mol Cancer Ther; 17(8); 1717-26. ©2018 AACR.


Subject(s)
Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Hydrazines/therapeutic use , Radiation Tolerance/drug effects , Triazoles/therapeutic use , Animals , Brain Neoplasms/pathology , Female , Glioblastoma/pathology , Humans , Hydrazines/pharmacology , Mice , Mice, Nude , Triazoles/pharmacology
14.
Mol Cancer Ther ; 17(6): 1207-1216, 2018 06.
Article in English | MEDLINE | ID: mdl-29549168

ABSTRACT

Radiotherapy is a primary treatment modality for glioblastomas (GBM). Because DNA-PKcs is a critical factor in the repair of radiation-induced double strand breaks (DSB), this study evaluated the potential of VX-984, a new DNA-PKcs inhibitor, to enhance the radiosensitivity of GBM cells. Treatment of the established GBM cell line U251 and the GBM stem-like cell (GSC) line NSC11 with VX-984 under in vitro conditions resulted in a concentration-dependent inhibition of radiation-induced DNA-PKcs phosphorylation. In a similar concentration-dependent manner, VX-984 treatment enhanced the radiosensitivity of each GBM cell line as defined by clonogenic analysis. As determined by γH2AX expression and neutral comet analyses, VX-984 inhibited the repair of radiation-induced DNA double-strand break in U251 and NSC11 GBM cells, suggesting that the VX-984-induced radiosensitization is mediated by an inhibition of DNA repair. Extending these results to an in vivo model, treatment of mice with VX-984 inhibited radiation-induced DNA-PKcs phosphorylation in orthotopic brain tumor xenografts, indicating that this compound crosses the blood-brain tumor barrier at sufficient concentrations. For mice bearing U251 or NSC11 brain tumors, VX-984 treatment alone had no significant effect on overall survival; radiation alone increased survival. The survival of mice receiving the combination protocol was significantly increased as compared with control and as compared with radiation alone. These results indicate that VX-984 enhances the radiosensitivity of brain tumor xenografts and suggest that it may be of benefit in the therapeutic management of GBM. Mol Cancer Ther; 17(6); 1207-16. ©2018 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , DNA-Activated Protein Kinase/antagonists & inhibitors , Glioblastoma/metabolism , Protein Kinase Inhibitors/pharmacology , Radiation Tolerance/drug effects , Radiation-Sensitizing Agents/pharmacology , Animals , Cell Line, Tumor , DNA-Activated Protein Kinase/metabolism , Disease Models, Animal , Female , Glioblastoma/pathology , Histones/metabolism , Humans , Mice , Phosphorylation , Xenograft Model Antitumor Assays
15.
Mol Cancer Ther ; 17(5): 1070-1078, 2018 05.
Article in English | MEDLINE | ID: mdl-29483212

ABSTRACT

The processes mediating the repair of DNA double-strand breaks (DSB) are critical determinants of radiosensitivity and provide a source of potential targets for tumor radiosensitization. Among the events required for efficient DSB repair are a variety of post-translational histone modifications, including methylation. Because trimethylation of histone H3 on lysine 27 (H3K27me3) has been associated with chromatin condensation, which can influence DSB repair, we determined the effects of radiation on H3K27me3 levels in tumor and normal cell lines. Irradiation of tumor cells resulted in a rapid loss of H3K27me3, which was prevented by the siRNA-mediated knockdown of the H3K27 demethylase UTX. Knockdown of UTX also enhanced the radiosensitivity of each tumor cell line. Treatment of tumor cells with the H3K27 demethylase inhibitor GSKJ4 immediately before irradiation prevented the radiation-induced decrease in H3K27me3 and enhanced radiosensitivity. As determined by neutral comet analysis and γH2AX expression, this GSKJ4 treatment protocol inhibited the repair of radiation-induced DSBs. Consistent with in vitro results, treatment of mice bearing leg tumor xenografts with GSKJ4 significantly enhance radiation-induce tumor growth delay. In contrast with results generated from tumor cell lines, radiation had no effect on H3K27me3 levels in normal fibroblast cell lines and GSKJ4 did not enhance their radiosensitivity. These data suggest that H3K27me3 demethylation contributes to DSB repair in tumor cells and that UTX, the demethylase responsible, provides a target for selective tumor cell radiosensitization. Mol Cancer Ther; 17(5); 1070-8. ©2018 AACR.


Subject(s)
Histone Demethylases/metabolism , Histones/metabolism , Lysine/metabolism , Nuclear Proteins/metabolism , Radiation Tolerance/radiation effects , A549 Cells , Benzazepines/pharmacology , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Cell Survival/radiation effects , DNA Breaks, Double-Stranded/drug effects , DNA Breaks, Double-Stranded/radiation effects , DNA Repair/drug effects , DNA Repair/radiation effects , Histone Demethylases/antagonists & inhibitors , Histone Demethylases/genetics , Humans , Methylation/drug effects , Methylation/radiation effects , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Pyrimidines/pharmacology , RNA Interference , Radiation Tolerance/drug effects , Radiation Tolerance/genetics
16.
Oncotarget ; 9(1): 691-705, 2018 Jan 02.
Article in English | MEDLINE | ID: mdl-29416646

ABSTRACT

Alternative splicing is a critical event in the posttranscriptional regulation of gene expression. To investigate whether this process influences radiation-induced gene expression we defined the effects of ionizing radiation on the generation of alternative transcripts in total cellular mRNA (the transcriptome) and polysome-bound mRNA (the translatome) of the human glioblastoma stem-like cell line NSC11. For these studies, RNA-Seq profiles from control and irradiated cells were compared using the program SpliceSeq to identify transcripts and splice variations induced by radiation. As compared to the transcriptome (total RNA) of untreated cells, the radiation-induced transcriptome contained 92 splice events suggesting that radiation induced alternative splicing. As compared to the translatome (polysome-bound RNA) of untreated cells, the radiation-induced translatome contained 280 splice events of which only 24 were overlapping with the radiation-induced transcriptome. These results suggest that radiation not only modifies alternative splicing of precursor mRNA, but also results in the selective association of existing mRNA isoforms with polysomes. Comparison of radiation-induced alternative transcripts to radiation-induced gene expression in total RNA revealed little overlap (about 3%). In contrast, in the radiation-induced translatome, about 38% of the induced alternative transcripts corresponded to genes whose expression level was affected in the translatome. This study suggests that whereas radiation induces alternate splicing, the alternative transcripts present at the time of irradiation may play a role in the radiation-induced translational control of gene expression and thus cellular radioresponse.

17.
Translation (Austin) ; 5(1): e1265703, 2017.
Article in English | MEDLINE | ID: mdl-28702276

ABSTRACT

Radiation-induced gene expression has long been hypothesized to protect against cell death. Defining this process would provide not only insight into the mechanisms mediating cell survival after radiation exposure, but also a novel source of targets for radiosensitization. However, whereas the radiation-induced gene expression profiles using total cellular mRNA have been generated for cell lines as well as normal tissues, with few exception, the changes in mRNA do not correlate with changes in the corresponding protein. The traditional approach to profiling gene expression, i.e., using total cellular RNA, does not take into account posttranscriptional regulation. In this review, we describe the use of gene expression profiling of polysome-bound RNA to establish that radiation modifies gene expression via translational control. Because changes in polysome-bound mRNA correlate with changes in protein, analysis of the translational profiles provides a unique data set for investigating the mechanisms mediating cellular radioresponse.

18.
Oncotarget ; 7(47): 77365-77377, 2016 Nov 22.
Article in English | MEDLINE | ID: mdl-27764801

ABSTRACT

Glioblastoma multiforme (GBM) continues to be the most frequently diagnosed and lethal primary brain tumor. Adjuvant chemo-radiotherapy remains the standard of care following surgical resection. In this study, using reverse phase protein arrays (RPPAs), we assessed the biological effects of radiation on signaling pathways to identify potential radiosensitizing molecular targets. We identified subsets of proteins with clearly concordant/discordant behavior between irradiated and non-irradiated GBM cells in vitro and in vivo. Moreover, we observed high expression of Forkhead box protein M1 (FOXM1) in irradiated GBM cells both in vitro and in vivo. Recent evidence of FOXM1 as a master regulator of metastasis and its important role in maintaining neural, progenitor, and GBM stem cells, intrigued us to validate it as a radiosensitizing target. Here we show that FOXM1 inhibition radiosensitizes GBM cells by abrogating genes associated with cell cycle progression and DNA repair, suggesting its role in cellular response to radiation. Further, we demonstrate that radiation induced stimulation of FOXM1 expression is dependent on STAT3 activation. Co-immunoprecipitation and co-localization assays revealed physical interaction of FOXM1 with phosphorylated STAT3 under radiation treatment. In conclusion, we hypothesize that FOXM1 regulates radioresistance via STAT3 in GBM cells. We also, show GBM patients with high FOXM1 expression have poor prognosis. Collectively our observations might open novel opportunities for targeting FOXM1 for effective GBM therapy.


Subject(s)
Brain Neoplasms/metabolism , Forkhead Box Protein M1/metabolism , Glioblastoma/metabolism , Radiation Tolerance , STAT3 Transcription Factor/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/mortality , Brain Neoplasms/radiotherapy , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Line, Tumor , DNA Breaks, Double-Stranded , DNA Repair , Forkhead Box Protein M1/genetics , Glioblastoma/genetics , Glioblastoma/mortality , Glioblastoma/radiotherapy , Homologous Recombination , Humans , Kaplan-Meier Estimate , Mitosis/drug effects , Peptides/pharmacology , Prognosis , Protein Binding , Protein Transport , Proteome , Proteomics/methods , RNA Interference , RNA, Small Interfering/genetics , Radiation Tolerance/genetics , STAT3 Transcription Factor/genetics
19.
Cancer Res ; 76(10): 3078-87, 2016 05 15.
Article in English | MEDLINE | ID: mdl-27005284

ABSTRACT

Changes in polysome-bound mRNA (translatome) are correlated closely with changes in the proteome in cells. Therefore, to better understand the processes mediating the response of glioblastoma to ionizing radiation (IR), we used polysome profiling to define the IR-induced translatomes of a set of human glioblastoma stem-like cell (GSC) lines. Although cell line specificity accounted for the largest proportion of genes within each translatome, there were also genes that were common to the GSC lines. In particular, analyses of the IR-induced common translatome identified components of the DNA damage response, consistent with a role for the translational control of gene expression in cellular radioresponse. Moreover, translatome analyses suggested that IR enhanced cap-dependent translation processes, an effect corroborated by the finding of increased eIF4F-cap complex formation detected after irradiation in all GSC lines. Translatome analyses also predicted that Golgi function was affected by IR. Accordingly, Golgi dispersal was detected after irradiation of each of the GSC lines. In addition to the common responses seen, translatome analyses predicted cell line-specific changes in mitochondria, as substantiated by changes in mitochondrial mass and DNA content. Together, these results suggest that analysis of radiation-induced translatomes can provide new molecular insights concerning the radiation response of cancer cells. More specifically, they suggest that the translational control of gene expression may provide a source of molecular targets for glioblastoma radiosensitization. Cancer Res; 76(10); 3078-87. ©2016 AACR.


Subject(s)
Glioblastoma/pathology , Golgi Apparatus/metabolism , Mitochondria/metabolism , Neoplastic Stem Cells/pathology , Polyribosomes/metabolism , Protein Biosynthesis/radiation effects , Fluorescent Antibody Technique , Gene Expression Profiling , Glioblastoma/genetics , Glioblastoma/radiotherapy , Golgi Apparatus/genetics , Golgi Apparatus/radiation effects , Humans , Mitochondria/genetics , Mitochondria/radiation effects , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Oligonucleotide Array Sequence Analysis , Polyribosomes/genetics , Polyribosomes/radiation effects , RNA, Messenger/genetics , Radiation, Ionizing , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
20.
Transl Cancer Res ; 5(Suppl 6): S1029-S1032, 2016 Nov.
Article in English | MEDLINE | ID: mdl-32999862
SELECTION OF CITATIONS
SEARCH DETAIL
...