116 results on '"Tofilon, PJ"'
Search Results
2. Heterogeneity in radiation sensitivity within human primary tumour cell cultures as detected by the SCE assay.
- Author
-
Tofilon, PJ, Vines, CM, Meyn, RE, Wike, J, Brock, WA, Tofilon, P J, Vines, C M, Meyn, R E, and Brock, W A
- Published
- 1989
- Full Text
- View/download PDF
3. The effects of N-methylformamide on artificial and spontaneous metastases from a murine hepatocarcinoma.
- Author
-
Tofilon, PJ, Vines, CM, Milas, L, Tofilon, P J, and Vines, C M
- Published
- 1987
- Full Text
- View/download PDF
4. RESPONSE: re: enhancement of tumor response to gamma-radiation by an inhibitor of cyclooxygenase-2 enzyme.
- Author
-
Milas, L, Mason, KA, and Tofilon, PJ
- Published
- 2000
- Full Text
- View/download PDF
5. Targeting PRMT5 enhances the radiosensitivity of tumor cells grown in vitro and in vivo.
- Author
-
Degorre C, Lohard S, Bobrek CN, Rawal KN, Kuhn S, and Tofilon PJ
- Subjects
- Animals, Humans, Cell Line, Tumor, Mice, DNA Repair, Cell Proliferation radiation effects, Xenograft Model Antitumor Assays, DNA Breaks, Double-Stranded radiation effects, Mice, Nude, Protein-Arginine N-Methyltransferases metabolism, Protein-Arginine N-Methyltransferases genetics, Protein-Arginine N-Methyltransferases antagonists & inhibitors, Radiation Tolerance drug effects, Radiation Tolerance genetics
- Abstract
PRMT5 is a widely expressed arginine methyltransferase that regulates processes involved in tumor cell proliferation and survival. In the study described here, we investigated whether PRMT5 provides a target for tumor radiosensitization. Knockdown of PRMT5 using siRNA enhanced the radiosensitivity of a panel of cell lines corresponding to tumor types typically treated with radiotherapy. To extend these studies to an experimental therapeutic setting, the PRMT5 inhibitor LLY-283 was used. Exposure of the tumor cell lines to LLY-283 decreased PRMT5 activity and enhanced their radiosensitivity. This increase in radiosensitivity was accompanied by an inhibition of DNA double-strand break repair as determined by γH2AX foci and neutral comet analyses. For a normal fibroblast cell line, although LLY-283 reduced PRMT5 activity, it had no effect on their radiosensitivity. Transcriptome analysis of U251 cells showed that LLY-283 treatment reduced the expression of genes and altered the mRNA splicing pattern of genes involved in the DNA damage response. Subcutaneous xenografts were then used to evaluate the in vivo response to LLY-283 and radiation. Treatment of mice with LLY-283 decreased tumor PRMT5 activity and significantly enhanced the radiation-induced growth delay. These results suggest that PRMT5 is a tumor selective target for radiosensitization., (© 2024. This is a U.S. Government work and not under copyright protection in the US; foreign copyright protection may apply.)
- Published
- 2024
- Full Text
- View/download PDF
6. MGMT inhibition regulates radioresponse in GBM, GSC, and melanoma.
- Author
-
Yun HS, Kramp TR, Palanichamy K, Tofilon PJ, and Camphausen K
- Subjects
- Humans, Cell Line, Tumor, 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, 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, Radiation Tolerance genetics
- 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., (© 2024. This is a U.S. Government work and not under copyright protection in the US; foreign copyright protection may apply.)
- Published
- 2024
- Full Text
- View/download PDF
7. Glioblastoma cells have increased capacity to repair radiation-induced DNA damage after migration to the olfactory bulb.
- Author
-
Degorre C, Sutton IC, Lehman SL, Shankavaram UT, Camphausen K, and Tofilon PJ
- 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., (© 2022. This is a U.S. Government work and not under copyright protection in the US; foreign copyright protection may apply.)
- Published
- 2022
- Full Text
- View/download PDF
8. Inhibition of the Translation Initiation Factor eIF4A Enhances Tumor Cell Radiosensitivity.
- Author
-
Lehman SL, Wechsler T, Schwartz K, Brown LE, Porco JA, Devine WG, Pelletier J, Shankavaram UT, Camphausen K, and Tofilon PJ
- Subjects
- Animals, Cell Line, Tumor, DNA Breaks, Double-Stranded, Humans, Mice, Xenograft Model Antitumor Assays, Eukaryotic Initiation Factor-4F antagonists & inhibitors, Neoplasms radiotherapy, Radiation Tolerance
- 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., (©2022 American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
9. CX-5461 induces radiosensitization through modification of the DNA damage response and not inhibition of RNA polymerase I.
- Author
-
Lehman SL, Schwartz KR, Maheshwari S, Camphausen K, and Tofilon PJ
- Subjects
- Apoptosis, Cell Line, Tumor, DNA Breaks, Double-Stranded, DNA Damage, Humans, Benzothiazoles pharmacology, Naphthyridines pharmacology, RNA Polymerase I antagonists & inhibitors, Radiation-Sensitizing Agents pharmacology
- 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., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
10. Translation Initiation Machinery as a Tumor Selective Target for Radiosensitization.
- Author
-
Lehman SL, Wilson ED, Camphausen K, and Tofilon PJ
- Subjects
- Animals, Eukaryotic Initiation Factors metabolism, Gene Expression Regulation, Neoplastic, Humans, Neoplasms metabolism, Neoplasms radiotherapy, Protein Processing, Post-Translational, Radiotherapy, Ribosomes metabolism, Signal Transduction, Biomarkers, Tumor, Neoplasms genetics, Peptide Chain Initiation, Translational radiation effects, Protein Biosynthesis radiation effects, Radiation Tolerance genetics
- 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.
- Published
- 2021
- Full Text
- View/download PDF
11. The Radiosensitizing Effect of AZD0530 in Glioblastoma and Glioblastoma Stem-Like Cells.
- Author
-
Yun HS, Lee J, Kil WJ, Kramp TR, Tofilon PJ, and Camphausen K
- Subjects
- 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, Benzodioxoles pharmacology, Gene Expression Regulation, Neoplastic, Glioblastoma radiotherapy, Neoplastic Stem Cells radiation effects, Quinazolines pharmacology, Radiation Tolerance drug effects, Radiation-Sensitizing Agents pharmacology
- 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., (©2021 American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
12. Improving Radiation Response in Glioblastoma Using ECO/siRNA Nanoparticles Targeting DNA Damage Repair.
- Author
-
Lee JA, Ayat N, Sun Z, Tofilon PJ, Lu ZR, and Camphausen K
- 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.
- Published
- 2020
- Full Text
- View/download PDF
13. Radiation Drives the Evolution of Orthotopic Xenografts Initiated from Glioblastoma Stem-like Cells.
- Author
-
McAbee JH, Rath BH, Valdez K, Young DL, Wu X, Shankavaram UT, Camphausen K, and Tofilon PJ
- Subjects
- 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, Brain Neoplasms radiotherapy, Evolution, Molecular, Genetic Heterogeneity radiation effects, Glioblastoma radiotherapy, Neoplastic Stem Cells radiation effects
- 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., (©2019 American Association for Cancer Research.)- Published
- 2019
- Full Text
- View/download PDF
14. Late toxicity in long-term survivors from a phase 2 study of concurrent radiation therapy, temozolomide and valproic acid for newly diagnosed glioblastoma.
- Author
-
Krauze AV, Mackey M, Rowe L, Chang MG, Holdford DJ, Cooley T, Shih J, Tofilon PJ, and Camphausen K
- 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.
- Published
- 2018
- Full Text
- View/download PDF
15. The XPO1 Inhibitor Selinexor Inhibits Translation and Enhances the Radiosensitivity of Glioblastoma Cells Grown In Vitro and In Vivo .
- Author
-
Wahba A, Rath BH, O'Neill JW, Camphausen K, and Tofilon PJ
- Subjects
- Animals, Brain Neoplasms pathology, Female, Glioblastoma pathology, Humans, Hydrazines pharmacology, Mice, Mice, Nude, Triazoles pharmacology, Brain Neoplasms drug therapy, Glioblastoma drug therapy, Hydrazines therapeutic use, Radiation Tolerance drug effects, Triazoles therapeutic use
- 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 ., (©2018 American Association for Cancer Research.)
- Published
- 2018
- Full Text
- View/download PDF
16. The DNA-PK Inhibitor VX-984 Enhances the Radiosensitivity of Glioblastoma Cells Grown In Vitro and as Orthotopic Xenografts.
- Author
-
Timme CR, Rath BH, O'Neill JW, Camphausen K, and Tofilon PJ
- Subjects
- Animals, Cell Line, Tumor, DNA-Activated Protein Kinase metabolism, Disease Models, Animal, Female, Glioblastoma pathology, Histones metabolism, Humans, Mice, Phosphorylation, Xenograft Model Antitumor Assays, Antineoplastic Agents pharmacology, DNA-Activated Protein Kinase antagonists & inhibitors, Glioblastoma metabolism, Protein Kinase Inhibitors pharmacology, Radiation Tolerance drug effects, Radiation-Sensitizing Agents pharmacology
- 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 ., (©2018 American Association for Cancer Research.)
- Published
- 2018
- Full Text
- View/download PDF
17. Inhibition of the Histone H3K27 Demethylase UTX Enhances Tumor Cell Radiosensitivity.
- Author
-
Rath BH, Waung I, Camphausen K, and Tofilon PJ
- Subjects
- 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, Histone Demethylases metabolism, Histones metabolism, Lysine metabolism, Nuclear Proteins metabolism, Radiation Tolerance radiation effects
- 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 ., (©2018 American Association for Cancer Research.)
- Published
- 2018
- Full Text
- View/download PDF
18. Radiation-induced alternative transcripts as detected in total and polysome-bound mRNA.
- Author
-
Wahba A, Ryan MC, Shankavaram UT, Camphausen K, and Tofilon PJ
- 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., Competing Interests: CONFLICTS OF INTEREST None.
- Published
- 2017
- Full Text
- View/download PDF
19. Radiation-induced translational control of gene expression.
- Author
-
Wahba A, Lehman SL, and Tofilon PJ
- 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.
- Published
- 2016
- Full Text
- View/download PDF
20. FOXM1 and STAT3 interaction confers radioresistance in glioblastoma cells.
- Author
-
Maachani UB, Shankavaram U, Kramp T, Tofilon PJ, Camphausen K, and Tandle AT
- Subjects
- 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, STAT3 Transcription Factor genetics, Brain Neoplasms metabolism, Forkhead Box Protein M1 metabolism, Glioblastoma metabolism, Radiation Tolerance genetics, STAT3 Transcription Factor metabolism
- 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.
- Published
- 2016
- Full Text
- View/download PDF
21. Glioblastoma radiosensitization by pimozide.
- Author
-
Rath BH, Camphausen K, and Tofilon PJ
- Abstract
Competing Interests: Conflicts of Interest: The authors have no conflicts of interest to declare.
- Published
- 2016
- Full Text
- View/download PDF
22. Polysome Profiling Links Translational Control to the Radioresponse of Glioblastoma Stem-like Cells.
- Author
-
Wahba A, Rath BH, Bisht K, Camphausen K, and Tofilon PJ
- Subjects
- 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, Glioblastoma pathology, Golgi Apparatus metabolism, Mitochondria metabolism, Neoplastic Stem Cells pathology, Polyribosomes metabolism, Protein Biosynthesis radiation effects
- 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., (©2016 American Association for Cancer Research.)
- Published
- 2016
- Full Text
- View/download PDF
23. Coculture with astrocytes reduces the radiosensitivity of glioblastoma stem-like cells and identifies additional targets for radiosensitization.
- Author
-
Rath BH, Wahba A, Camphausen K, and Tofilon PJ
- Subjects
- Animals, Brain Neoplasms genetics, Cell Line, Tumor, Cluster Analysis, Coculture Techniques, Cytokines metabolism, Disease Models, Animal, Dose-Response Relationship, Radiation, Female, Gene Expression Profiling, Glioblastoma genetics, Histones metabolism, Humans, Mice, STAT3 Transcription Factor metabolism, Xenograft Model Antitumor Assays, Astrocytes metabolism, Astrocytes radiation effects, Brain Neoplasms metabolism, Glioblastoma metabolism, Neoplastic Stem Cells metabolism, Neoplastic Stem Cells radiation effects, Radiation Tolerance genetics
- Abstract
Toward developing a model system for investigating the role of the microenvironment in the radioresistance of glioblastoma (GBM), human glioblastoma stem-like cells (GSCs) were grown in coculture with human astrocytes. Using a trans-well assay, survival analyses showed that astrocytes significantly decreased the radiosensitivity of GSCs compared to standard culture conditions. In addition, when irradiated in coculture, the initial level of radiation-induced γH2AX foci in GSCs was reduced and foci dispersal was enhanced suggesting that the presence of astrocytes influenced the induction and repair of DNA double-strand breaks. These data indicate that astrocytes can decrease the radiosensitivity of GSCs in vitro via a paracrine-based mechanism and further support a role for the microenvironment as a determinant of GBM radioresponse. Chemokine profiling of coculture media identified a number of bioactive molecules not present under standard culture conditions. The gene expression profiles of GSCs grown in coculture were significantly different as compared to GSCs grown alone. These analyses were consistent with an astrocyte-mediated modification in GSC phenotype and, moreover, suggested a number of potential targets for GSC radiosensitization that were unique to coculture conditions. Along these lines, STAT3 was activated in GSCs grown with astrocytes; the JAK/STAT3 inhibitor WP1066 enhanced the radiosensitivity of GSCs under coculture conditions and when grown as orthotopic xenografts. Further, this coculture system may also provide an approach for identifying additional targets for GBM radiosensitization., (© 2015 The Authors. Cancer Medicine published by John Wiley & Sons Ltd.)
- Published
- 2015
- Full Text
- View/download PDF
24. The ATP-competitive mTOR inhibitor INK128 enhances in vitro and in vivo radiosensitivity of pancreatic carcinoma cells.
- Author
-
Hayman TJ, Wahba A, Rath BH, Bae H, Kramp T, Shankavaram UT, Camphausen K, and Tofilon PJ
- Subjects
- Adenosine Triphosphate antagonists & inhibitors, Adenosine Triphosphate physiology, Animals, Cell Line, Tumor, Cell Survival drug effects, Cell Survival radiation effects, DNA Breaks, Double-Stranded, Female, Humans, Mice, Mice, Nude, Protein Biosynthesis radiation effects, Radiation Tolerance drug effects, TOR Serine-Threonine Kinases metabolism, Xenograft Model Antitumor Assays, Pancreatic Neoplasms, Benzoxazoles pharmacology, Pancreatic Neoplasms radiotherapy, Pyrimidines pharmacology, Radiation-Sensitizing Agents pharmacology, TOR Serine-Threonine Kinases antagonists & inhibitors
- Abstract
Purpose: Radiotherapy remains a primary treatment modality for pancreatic carcinoma, a tumor characterized by aberrant mTOR activity. Given the regulatory role of mTOR in gene translation, in this study, we defined the effects of the clinically relevant, ATP-competitive mTOR inhibitor, INK128 on the radiosensitivity of pancreatic carcinoma cell lines., Experimental Design: Clonogenic survival was used to determine the effects of INK128 on in vitro radiosensitivity of three pancreatic carcinoma cell lines and a normal fibroblast cell line with mTOR activity defined using immunoblots. DNA double-strand breaks were evaluated according to γH2AX foci. The influence of INK128 on radiation-induced gene translation was determined by microarray analysis of polysome-bound mRNA. Leg tumor xenografts grown from pancreatic carcinoma cells were evaluated for mTOR activity, eIF4F cap complex formation, and tumor growth delay., Results: INK128, while inhibiting mTOR activity in each of the cell lines, enhanced the in vitro radiosensitivity of the pancreatic carcinoma cells but had no effect on normal fibroblasts. The dispersal of radiation-induced γH2AX foci was inhibited in pancreatic carcinoma cells by INK128 as were radiation-induced changes in gene translation. Treatment of mice with INK128 resulted in an inhibition of mTOR activity as well as cap complex formation in tumor xenografts. Whereas INK128 alone had no effect of tumor growth rate, it enhanced the tumor growth delay induced by single and fractionated doses of radiation., Conclusion: These results indicate that mTOR inhibition induced by INK128 enhances the radiosensitivity of pancreatic carcinoma cells and suggest that this effect involves the inhibition of DNA repair.
- Published
- 2014
- Full Text
- View/download PDF
25. The mTORC1/mTORC2 inhibitor AZD2014 enhances the radiosensitivity of glioblastoma stem-like cells.
- Author
-
Kahn J, Hayman TJ, Jamal M, Rath BH, Kramp T, Camphausen K, and Tofilon PJ
- Subjects
- Animals, Apoptosis drug effects, Apoptosis radiation effects, Benzamides, Brain Neoplasms pathology, Brain Neoplasms radiotherapy, Cell Cycle drug effects, Cell Cycle radiation effects, Cell Proliferation drug effects, Cell Proliferation radiation effects, DNA Breaks, Double-Stranded drug effects, DNA Breaks, Double-Stranded radiation effects, DNA Repair drug effects, DNA Repair radiation effects, Female, Fluorescent Antibody Technique, Glioblastoma pathology, Glioblastoma radiotherapy, Histones metabolism, Humans, Mechanistic Target of Rapamycin Complex 1, Mechanistic Target of Rapamycin Complex 2, Mice, Mice, Nude, Neoplastic Stem Cells drug effects, Neoplastic Stem Cells radiation effects, Protein Kinase Inhibitors pharmacology, Pyrimidines, Tumor Cells, Cultured, X-Ray Therapy, Xenograft Model Antitumor Assays, Brain Neoplasms drug therapy, Glioblastoma drug therapy, Morpholines pharmacology, Multiprotein Complexes antagonists & inhibitors, Neoplastic Stem Cells pathology, Radiation-Sensitizing Agents pharmacology, TOR Serine-Threonine Kinases antagonists & inhibitors
- Abstract
Background: The mammalian target of rapamycin (mTOR) has been suggested as a target for radiosensitization. Given that radiotherapy is a primary treatment modality for glioblastoma (GBM) and that mTOR is often dysregulated in GBM, the goal of this study was to determine the effects of AZD2014, a dual mTORC1/2 inhibitor, on the radiosensitivity of GBM stem-like cells (GSCs)., Methods: mTORC1 and mTORC2 activities were defined by immunoblot analysis. The effects of this mTOR inhibitor on the in vitro radiosensitivity of GSCs were determined using a clonogenic assay. DNA double strand breaks were evaluated according to γH2AX foci. Orthotopic xenografts initiated from GSCs were used to define the in vivo response to AZD2014 and radiation., Results: Exposure of GSCs to AZD2014 resulted in the inhibition of mTORC1 and 2 activities. Based on clonogenic survival analysis, addition of AZD2014 to culture media 1 hour before irradiation enhanced the radiosensitivity of CD133+ and CD15+ GSC cell lines. Whereas AZD2014 treatment had no effect on the initial level of γH2AX foci, the dispersal of radiation-induced γH2AX foci was significantly delayed. Finally, the combination of AZD2014 and radiation delivered to mice bearing GSC-initiated orthotopic xenografts significantly prolonged survival as compared with the individual treatments., Conclusions: These data indicate that AZD2014 enhances the radiosensitivity of GSCs both in vitro and under orthotopic in vivo conditions and suggest that this effect involves an inhibition of DNA repair. Moreover, these results suggest that this dual mTORC1/2 inhibitor may be a radiosensitizer applicable to GBM therapy.
- Published
- 2014
- Full Text
- View/download PDF
26. Ionizing radiation and glioblastoma exosomes: implications in tumor biology and cell migration.
- Author
-
Arscott WT, Tandle AT, Zhao S, Shabason JE, Gordon IK, Schlaff CD, Zhang G, Tofilon PJ, and Camphausen KA
- Abstract
Exosomes are nanometer-sized lipid vesicles released ubiquitously by cells, which have been shown to have a normal physiological role, as well as influence the tumor microenvironment and aid metastasis. Recent studies highlight the ability of exosomes to convey tumor-suppressive and oncogenic mRNAs, microRNAs, and proteins to a receiving cell, subsequently activating downstream signaling pathways and influencing cellular phenotype. Here, we show that radiation increases the abundance of exosomes released by glioblastoma cells and normal astrocytes. Exosomes derived from irradiated cells enhanced the migration of recipient cells, and their molecular profiling revealed an abundance of molecules related to signaling pathways important for cell migration. In particular, connective tissue growth factor (CTGF) mRNA and insulin-like growth factor binding protein 2 (IGFBP2) protein levels were elevated, and coculture of nonirradiated cells with exosomes isolated from irradiated cells increased CTGF protein expression in the recipient cells. Additionally, these exosomes enhanced the activation of neurotrophic tyrosine kinase receptor type 1 (TrkA), focal adhesion kinase, Paxillin, and proto-oncogene tyrosine-protein kinase Src (Src) in recipient cells, molecules involved in cell migration. Collectively, our data suggest that radiation influences exosome abundance, specifically alters their molecular composition, and on uptake, promotes a migratory phenotype.
- Published
- 2013
- Full Text
- View/download PDF
27. Competitive but Not Allosteric mTOR Kinase Inhibition Enhances Tumor Cell Radiosensitivity.
- Author
-
Hayman TJ, Kramp T, Kahn J, Jamal M, Camphausen K, and Tofilon PJ
- Abstract
The mechanistic target of rapamycin (mTOR) is a critical kinase in the regulation of gene translation and has been suggested as a potential target for radiosensitization. The goal of this study was to compare the radiosensitizing activities of the allosteric mTOR inhibitor rapamycin with that of the competitive mTOR inhibitor PP242. On the basis of immunoblot analyses, whereas rapamycin only partially inhibited mTOR complex 1 (mTORC1) activity and had no effect on mTOR complex 2 (mTORC2), PP242 inhibited the activity of both mTOR-containing complexes. Irradiation alone had no effect on mTORC1 or mTORC2 activity. Clonogenic survival was used to define the effects of the mTOR inhibitors on in vitro radiosensitivity. In the two tumor cell lines evaluated, PP242 treatment 1 hour before irradiation increased radiosensitivity, whereas rapamycin had no effect. Addition of PP242 after irradiation also enhanced the radiosensitivity of both tumor lines. To investigate the mechanism of radiosensitization, the induction and repair of DNA double-strand breaks were evaluated according γH2AX foci. PP242 exposure did not influence the initial level of γH2AX foci after irradiation but did significantly delay the dispersal of radiation-induced γH2AX foci. In contrast to the tumor cell lines, the radiosensitivity of a normal human fibroblast cell line was not influenced by PP242. Finally, PP242 administration to mice bearing U251 xenografts enhanced radiation-induced tumor growth delay. These results indicate that in a preclinical tumor model PP242 enhances tumor cell radiosensitivity both in vitro and in vivo and suggest that this effect involves an inhibition of DNA repair.
- Published
- 2013
- Full Text
- View/download PDF
28. Astrocytes enhance the invasion potential of glioblastoma stem-like cells.
- Author
-
Rath BH, Fair JM, Jamal M, Camphausen K, and Tofilon PJ
- Subjects
- AC133 Antigen, Antigens, CD metabolism, Astrocytes cytology, Astrocytes metabolism, Brain Neoplasms metabolism, Cell Differentiation, Cell Line, Tumor, Culture Media, Conditioned metabolism, Culture Media, Conditioned pharmacology, Glioblastoma metabolism, Glycoproteins metabolism, Humans, Microscopy, Electron, Scanning Transmission, Neoplastic Stem Cells metabolism, Peptides metabolism, Transcriptome, Tumor Microenvironment, Brain Neoplasms pathology, Glioblastoma pathology, Neoplasm Invasiveness pathology, Neoplastic Stem Cells pathology
- Abstract
Glioblastomas (GBMs) are characterized as highly invasive; the contribution of GBM stem-like cells (GSCs) to the invasive phenotype, however, has not been completely defined. Towards this end, we have defined the invasion potential of CD133+ GSCs and their differentiated CD133- counterparts grown under standard in vitro conditions and in co-culture with astrocytes. Using a trans-well assay, astrocytes or astrocyte conditioned media in the bottom chamber significantly increased the invasion of GSCs yet had no effect on CD133- cells. In addition, a monolayer invasion assay showed that the GSCs invaded farther into an astrocyte monolayer than their differentiated progeny. Gene expression profiles were generated from two GSC lines grown in trans-well culture with astrocytes in the bottom chamber or directly in contact with astrocyte monolayers. In each co-culture model, genes whose expression was commonly increased in both GSC lines involved cell movement and included a number of genes that have been previously associated with tumor cell invasion. Similar gene expression modifications were not detected in CD133- cells co-cultured under the same conditions with astrocytes. Finally, evaluation of the secretome of astrocytes grown in monolayer identified a number of chemokines and cytokines associated with tumor cell invasion. These data suggest that astrocytes enhance the invasion of CD133+ GSCs and provide additional support for a critical role of brain microenvironment in the regulation of GBM biology.
- Published
- 2013
- Full Text
- View/download PDF
29. Preclinical models in radiation oncology.
- Author
-
Kahn J, Tofilon PJ, and Camphausen K
- Subjects
- Animals, Humans, Neoplasms drug therapy, Antineoplastic Agents therapeutic use, Neoplasms radiotherapy, Radiation Oncology, Xenograft Model Antitumor Assays
- Abstract
As the incidence of cancer continues to rise, the use of radiotherapy has emerged as a leading treatment modality. Preclinical models in radiation oncology are essential tools for cancer research and therapeutics. Various model systems have been used to test radiation therapy, including in vitro cell culture assays as well as in vivo ectopic and orthotopic xenograft models. This review aims to describe such models, their advantages and disadvantages, particularly as they have been employed in the discovery of molecular targets for tumor radiosensitization. Ultimately, any model system must be judged by its utility in developing more effective cancer therapies, which is in turn dependent on its ability to simulate the biology of tumors as they exist in situ. Although every model has its limitations, each has played a significant role in preclinical testing. Continued advances in preclinical models will allow for the identification and application of targets for radiation in the clinic.
- Published
- 2012
- Full Text
- View/download PDF
30. Translation initiation factor eIF4E is a target for tumor cell radiosensitization.
- Author
-
Hayman TJ, Williams ES, Jamal M, Shankavaram UT, Camphausen K, and Tofilon PJ
- Subjects
- Cell Line, Tumor, DNA Breaks, Double-Stranded, DNA Repair, DNA, Neoplasm genetics, Gene Knockdown Techniques, Humans, Neoplasms genetics, Neoplasms metabolism, Nucleocytoplasmic Transport Proteins deficiency, Nucleocytoplasmic Transport Proteins genetics, RNA, Small Interfering administration & dosage, RNA, Small Interfering genetics, Radiation Tolerance, Transfection, Neoplasms radiotherapy, Nucleocytoplasmic Transport Proteins metabolism
- Abstract
A core component in the cellular response to radiation occurs at the level of translational control of gene expression. Because a critical element in translation control is the availability of the initiation factor eIF4E, which selectively enhances the cap-dependent translation of mRNAs, we investigated a regulatory role for eIF4E in cellular radiosensitivity. eIF4E silencing enhanced the radiosensitivity of tumor cell lines but not normal cells. Similarly, pharmacologic inhibition of eIF4E with ribavirin also enhanced tumor cell radiosensitivity. eIF4E attenuation did not affect cell-cycle phase distribution or radiation-induced apoptosis, but it delayed the dispersion of radiation-induced γH2AX foci and increased the frequency of radiation-induced mitotic catastrophe. Radiation did not affect 4E-BP1 phosphorylation or cap-complex formation but it increased eIF4E binding to more than 1,000 unique transcripts including many implicated in DNA replication, recombination, and repair. Taken together, our findings suggest that eIF4E represents a logical therapeutic target to increase tumor cell radiosensitivity., (©2012 AACR)
- Published
- 2012
- Full Text
- View/download PDF
31. Post-radiation increase in VEGF enhances glioma cell motility in vitro.
- Author
-
Kil WJ, Tofilon PJ, and Camphausen K
- Subjects
- Blotting, Western, Brain Neoplasms metabolism, Brain Neoplasms pathology, Brain Neoplasms radiotherapy, Cell Adhesion, Cell Proliferation, Culture Media, Conditioned pharmacology, Glioma radiotherapy, Humans, In Vitro Techniques, RNA, Messenger genetics, Real-Time Polymerase Chain Reaction, Tumor Cells, Cultured, Vascular Endothelial Growth Factor A antagonists & inhibitors, Vascular Endothelial Growth Factor A genetics, Vascular Endothelial Growth Factor Receptor-2 genetics, Cell Movement, Glioma metabolism, Glioma pathology, Vascular Endothelial Growth Factor A metabolism, Vascular Endothelial Growth Factor Receptor-2 metabolism
- Abstract
Background: Glioblastoma multiforme (GBM) is among the most lethal of all human tumors, with frequent local recurrences after radiation therapy (RT). The mechanism accounting for such a recurrence pattern is unclear. It has classically been attributed to local recurrence of treatment-resistant cells. However, accumulating evidence suggests that additional mechanisms exist that involve the migration of tumor or tumor stem cells from other brain regions to tumor bed. VEGFs are well-known mitogens and can be up-regulated after RT. Here, we examine the effect of irradiation-induced VEGF on glioma cell motility., Materials and Methods: U251 and LN18 cell lines were used to generate irradiated-conditioned medium (IR-CM). At 72 h after irradiation, the supernatants were harvested. VEGF level in IR-CM was quantified by ELISA, and expression levels for VEGF mRNA were detected by RT-PCR. In vitro cancer cell motility was measured in chambers coated with/without Matrigel and IR-CM as a cell motility enhancer and a VEGF antibody as a neutralizer of VEGF bioactivity. Immunoblots were performed to evaluate the activity of cell motility-related kinases. Proliferation of GBM cells after treatment was measured by flow cytometry., Results: Irradiation increased the level of VEGF mRNA that was mitigated by pre-RT exposure to Actinomycin D. U251 glioma cell motility (migration and invasion) was enhanced by adding IR-CM to un-irradiated cells (174.9 ± 11.4% and 334.2 ± 46% of control, respectively). When we added VEGF antibody to IR-CM, this enhanced cell motility was negated (110.3 ± 12.0% and 105.7 ± 14.0% of control, respectively). Immunoblot analysis revealed that IR-CM increased phosphorylation of VEGF receptor-2 (VEGFR2) secondary to an increase in VEGF, with a concomitant increase of phosphorylation of the downstream targets (Src and FAK). Increased phosphorylation was mitigated by adding VEGF antibody to IR-CM. There was no difference in the mitotic index of GBM cells treated with and without IR-CM and VEGF., Conclusions: These results indicate that cell motility can be enhanced by conditioned medium from irradiated cells in vitro through stimulation of VEGFR2 signaling pathways and suggest that this effect involves the secretion of radiation-induced VEGF, leading to an increase in glioma cell motility.
- Published
- 2012
- Full Text
- View/download PDF
32. The brain microenvironment preferentially enhances the radioresistance of CD133(+) glioblastoma stem-like cells.
- Author
-
Jamal M, Rath BH, Tsang PS, Camphausen K, and Tofilon PJ
- Subjects
- AC133 Antigen, Animals, Brain metabolism, Brain Neoplasms metabolism, Cell Line, Tumor, Genes, Reporter, Glioblastoma metabolism, Histones metabolism, Humans, Intracellular Signaling Peptides and Proteins metabolism, Luciferases, Firefly biosynthesis, Luciferases, Firefly genetics, Male, Mice, Mice, Nude, Neoplasm Transplantation, Neoplastic Stem Cells radiation effects, Neoplastic Stem Cells transplantation, Tumor Burden radiation effects, Tumor Suppressor p53-Binding Protein 1, Antigens, CD metabolism, Brain pathology, Brain Neoplasms pathology, Glioblastoma pathology, Glycoproteins metabolism, Neoplastic Stem Cells metabolism, Peptides metabolism, Radiation Tolerance, Tumor Microenvironment
- Abstract
Brain tumor xenografts initiated from glioblastoma (GBM) CD133(+) tumor stem-like cells (TSCs) are composed of TSC and non-TSC subpopulations, simulating the phenotypic heterogeneity of GBMs in situ. Given that the discrepancies between the radiosensitivity of GBM cells in vitro and the treatment response of patients suggest a role for the microenvironment in GBM radioresistance, we compared the response of TSCs and non-TSCs irradiated under in vitro and orthotopic conditions. As a measure of radioresponse determined at the individual cell level, γH2AX and 53BP1 foci were quantified in CD133(+) cells and their differentiated (CD133(-)) progeny. Under in vitro conditions, no difference was detected between CD133(+) and CD133(-) cells in foci induction or dispersal after irradiation. However, irradiation of orthotopic xenografts initiated from TSCs resulted in the induction of fewer γH2AX and 53BP1 foci in CD133(+) cells compared to their CD133(-) counterparts within the same tumor. Xenograft irradiation resulted in a tumor growth delay of approximately 7 days with a corresponding increase in the percentage of CD133(+) cells at 7 days after radiation, which persisted to the onset of neurologic symptoms. These results suggest that, although the radioresponse of TSCs and non-TSCs does not differ under in vitro growth conditions, CD133(+) cells are relatively radioresistant under intracerebral growth conditions. Whereas these findings are consistent with the suspected role for TSCs as a determinant of GBM radioresistance, these data also illustrate the dependence of the cellular radioresistance on the brain microenvironment.
- Published
- 2012
- Full Text
- View/download PDF
33. Grand rounds at the National Institutes of Health: HDAC inhibitors as radiation modifiers, from bench to clinic.
- Author
-
Shabason JE, Tofilon PJ, and Camphausen K
- Subjects
- Clinical Trials as Topic, Humans, National Institutes of Health (U.S.), Teaching Rounds, United States, Antineoplastic Agents therapeutic use, Brain Neoplasms drug therapy, Glioblastoma drug therapy, Histone Deacetylase Inhibitors therapeutic use
- Abstract
Glioblastoma multiforme (GBM) is the most common and aggressive malignant brain tumour. Patients afflicted with this disease unfortunately have a very poor prognosis, and fewer than 5% of patients survive for 5 years from the time of diagnosis. Therefore, improved therapies to treat this disease are sorely needed. One such class of drugs that have generated great enthusiasm for the treatment of numerous malignancies, including GBM, is histone deacetylase (HDAC) inhibitors. Pre-clinical data have demonstrated the efficacy of various HDAC inhibitors as anticancer agents, with the greatest effects shown when HDAC inhibitors are used in combination with other therapies. As a result of encouraging pre-clinical data, numerous HDAC inhibitors are under investigation in clinical trials, either as monotherapies or in conjunction with other treatments such as chemotherapy, biologic therapy or radiation therapy. In fact, two actively studied HDAC inhibitors, vorinostat and depsipeptide, were recently approved for the treatment of refractory cutaneous T cell lymphoma. In this review, we first present a patient with GBM, and then discuss the pathogenesis, epidemiology and current treatment options of GBM. Finally, we examine the translation of pre-clinical studies that have demonstrated HDAC inhibitors as potent radiosensitizers in in vitro and in vivo models, to a phase II clinical trial combining the HDAC inhibitor, valproic acid, along with temozolomide and radiation therapy for the treatment of GBM., (Journal of Cellular and Molecular Medicine © 2011 Foundation for Cellular and Molecular Medicine/Blackwell Publishing Ltd No claim to US government works.)
- Published
- 2011
- Full Text
- View/download PDF
34. Radiosensitization of glioma cells by modulation of Met signalling with the hepatocyte growth factor neutralizing antibody, AMG102.
- Author
-
Buchanan IM, Scott T, Tandle AT, Burgan WE, Burgess TL, Tofilon PJ, and Camphausen K
- Subjects
- Animals, Antibodies, Monoclonal, Humanized, Cell Death drug effects, Cell Death radiation effects, Cell Line, Tumor, Cell Proliferation drug effects, Cell Proliferation radiation effects, DNA Damage, Glioma pathology, Humans, Mice, Mice, Nude, Radiation Tolerance radiation effects, Radiation, Ionizing, Signal Transduction radiation effects, Antibodies, Monoclonal pharmacology, Antibodies, Neutralizing pharmacology, Glioma metabolism, Hepatocyte Growth Factor immunology, Proto-Oncogene Proteins c-met metabolism, Radiation Tolerance drug effects, Signal Transduction drug effects
- Abstract
The hepatocyte growth factor (HGF)/Met signalling pathway is up-regulated in many cancers, with downstream mediators playing a role in DNA double strand break repair. Previous studies have shown increased radiosensitization of tumours through modulation of Met signalling by genetic methods. We investigated the effects of the anti-HGF monoclonal antibody, AMG102, on the response to ionizing radiation in a model of glioblastoma multiforme in vitro and in vivo. Radiosensitivity was evaluated in vitro in the U-87 MG human glioma cell line. Met activation was measured by Western blot, and the effect on survival following radiation was evaluated by clonogenic assay. Mechanism of cell death was evaluated by apoptosis and mitotic catastrophe assays. DNA damage was quantitated by γH2AX foci and neutral comet assay. Growth kinetics of subcutaneous tumours was used to assess the effects of AMG102 on in vivo tumour radiosensitivity. AMG102 inhibited Met activation after irradiation. An enhancement of radiation cell killing was shown with no toxicity using drug alone. Retention of γH2AX foci at 6 and 24 hrs following the drug/radiation combination indicated an inhibition of DNA repair following radiation, and comet assay confirmed DNA damage persisting over the same duration. At 48 and 72 hrs following radiation, a significant increase of cells undergoing mitotic catastrophe was seen in the drug/radiation treated cells. Growth of subcutaneous tumours was slowed in combination treated mice, with an effect that was greater than additive for each modality individually. Modulation of Met signalling with AMG102 may prove a novel radiation sensitizing strategy. Our data indicate that DNA repair processes downstream of Met are impaired leading to increased cell death through mitotic catastrophe., (Journal of Cellular and Molecular Medicine © 2011 Foundation for Cellular and Molecular Medicine/Blackwell Publishing Ltd No claim to original US government works.)
- Published
- 2011
- Full Text
- View/download PDF
35. Microenvironmental regulation of glioblastoma radioresponse.
- Author
-
Jamal M, Rath BH, Williams ES, Camphausen K, and Tofilon PJ
- Subjects
- Animals, Brain Neoplasms genetics, Brain Neoplasms pathology, Cluster Analysis, Gene Expression Profiling, Gene Expression Regulation, Neoplastic physiology, Glioblastoma genetics, Glioblastoma pathology, Humans, Male, Mice, Mice, Nude, Microarray Analysis, Tumor Cells, Cultured, Tumor Microenvironment genetics, Xenograft Model Antitumor Assays, Brain Neoplasms radiotherapy, Glioblastoma radiotherapy, Radiation Tolerance genetics, Radiation Tolerance physiology, Tumor Microenvironment physiology
- Abstract
Purpose: Brain tumor xenografts initiated from human glioblastoma (GBM) stem-like cells (TSC) simulate the biological characteristics of GBMs in situ. Therefore, to determine whether the brain microenvironment affects the intrinsic radiosensitivity of GBM cells, we compared the radioresponse of GBM TSCs grown in vitro and as brain tumor xenografts., Experimental Design: As indicators of DNA double-strand breaks (DSB), γH2AX, and 53BP1 foci were defined after irradiation of 2 GBM TSC lines grown in vitro and as orthotopic xenografts in nude mice. Microarray analysis was conducted to compare gene expression patterns under each growth condition., Results: Dispersal of radiation-induced γH2AX and 53BP1 foci was faster in the tumor cells grown as orthotopic xenografts compared with cells irradiated in vitro. In addition, cells irradiated in vivo were approximately 3-fold less susceptible to foci induction as compared with cells grown in vitro. Microarray analysis revealed a significant number of genes whose expression was commonly affected in the 2 GBM models by orthotopic growth conditions. Consistent with the decrease in sensitivity to foci induction, genes related to reactive oxygen species (ROS) metabolism were expressed at higher levels in the brain tumor xenografts., Conclusion: γH2AX and 53BP1 foci analyses indicate that GBM cells irradiated within orthotopic xenografts have a greater capacity to repair DSBs and are less susceptible to their induction than tumor cells irradiated under in vitro growth conditions. Because DSB induction and repair are critical determinants of radiosensitivity, these results imply that the brain microenvironment contributes to GBM radioresistance., (©2010 AACR.)
- Published
- 2010
- Full Text
- View/download PDF
36. Activation of the unfolded protein response contributes toward the antitumor activity of vorinostat.
- Author
-
Kahali S, Sarcar B, Fang B, Williams ES, Koomen JM, Tofilon PJ, and Chinnaiyan P
- Subjects
- Acetylation drug effects, Blotting, Western, Cell Line, Tumor, Endoplasmic Reticulum Chaperone BiP, Enzyme Inhibitors pharmacology, Heat-Shock Proteins metabolism, Histone Acetyltransferases metabolism, Humans, Lysine metabolism, Phosphorylation drug effects, RNA Interference, Vorinostat, eIF-2 Kinase genetics, eIF-2 Kinase metabolism, Antineoplastic Agents pharmacology, Histone Acetyltransferases antagonists & inhibitors, Hydroxamic Acids pharmacology, Unfolded Protein Response drug effects
- Abstract
Histone deacetylase (HDAC) inhibitors represent an emerging class of anticancer agents progressing through clinical trials. Although their primary target is thought to involve acetylation of core histones, several nonhistone substrates have been identified, including heat shock protein (HSP) 90, which may contribute towards their antitumor activity. Glucose-regulated protein 78 (GRP78) is a member of the HSP family of molecular chaperones and plays a central role in regulating the unfolded protein response (UPR). Emerging data suggest that GRP78 is critical in cellular adaptation and survival associated with oncogenesis and may serve as a cancer-specific therapeutic target. On the basis of shared homology with HSP family proteins, we sought to determine whether GRP78 could serve as a molecular target of the HDAC inhibitor vorinostat. Vorinostat treatment led to GRP78 acetylation, dissociation, and subsequent activation of its client protein double-stranded RNA-activated protein-like endoplasmic reticulum kinase (PERK). Investigations in a panel of cancer cell lines identified that UPR activation after vorinostat exposure is specific to certain lines. Mass spectrometry performed on immunoprecipitated GRP78 identified lysine-585 as a specific vorinostat-induced acetylation site of GRP78. Downstream activation of the UPR was confirmed, including eukaryotic initiating factor 2alpha phosphorylation and increase in ATF4 and C/EBP homologous protein expression. To determine the biologic relevance of UPR activation after vorinostat, RNA interference of PERK was performed, demonstrating significantly decreased sensitivity to vorinostat-induced cytotoxicity. Collectively, these findings indicate that GRP78 is a biologic target of vorinostat, and activation of the UPR through PERK phosphorylation contributes toward its antitumor activity.
- Published
- 2010
- Full Text
- View/download PDF
37. High throughput evaluation of gamma-H2AX.
- Author
-
Avondoglio D, Scott T, Kil WJ, Sproull M, Tofilon PJ, and Camphausen K
- Subjects
- Animals, Cell Line, Tumor, Dose-Response Relationship, Radiation, Fluorescent Antibody Technique, Humans, Mice, Mice, Nude, Phosphorylation radiation effects, DNA Breaks, Double-Stranded radiation effects, Electrochemical Techniques, Histones radiation effects, Luminescent Measurements, Radiotherapy adverse effects
- Abstract
The DNA double-strand break (DSB) is the primary lethal lesion after therapeutic radiation. Thus, the development of assays to detect and to quantitate these lesions could have broad preclinical and clinical impact. Phosphorylation of histone H2AX to form gamma-H2AX is a known marker for irradiation-induced DNA DSBs. However, the first generation assay involves the use of immunofluorescent staining of gamma-H2AX foci. This assay is time consuming, operator dependent and is not scalable for high throughput assay development. Thus, we sought to develop a new assay using a high throughput electrochemiluminescent platform from Mesoscale Discovery Systems to quantify gamma-H2AX levels. The results show that our assay utilizes significantly less time and labor, has greater intra-assay reproducibility and has a greater dynamic range of gamma-H2AX versus irradiation dose.
- Published
- 2009
- Full Text
- View/download PDF
38. CD133+ glioblastoma stem-like cells are radiosensitive with a defective DNA damage response compared with established cell lines.
- Author
-
McCord AM, Jamal M, Williams ES, Camphausen K, and Tofilon PJ
- Subjects
- AC133 Antigen, Cell Line, Tumor, Cell Survival radiation effects, DNA Breaks, Double-Stranded radiation effects, DNA Damage genetics, DNA Repair genetics, DNA Repair physiology, DNA, Neoplasm radiation effects, Dose-Response Relationship, Radiation, Glioblastoma genetics, Glioblastoma metabolism, Histones analysis, Histones metabolism, Humans, Neoplastic Stem Cells metabolism, Neoplastic Stem Cells pathology, Neoplastic Stem Cells radiation effects, Rad51 Recombinase analysis, Rad51 Recombinase metabolism, Antigens, CD metabolism, DNA Repair radiation effects, Glioblastoma pathology, Glycoproteins metabolism, Neoplastic Stem Cells physiology, Peptides metabolism, Radiation Tolerance genetics
- Abstract
Purpose: CD133+ glioblastoma tumor stem-like cells (TSC) have been defined as radioresistant. However, although previously classified relative to CD133- cells, the radiosensitivity of CD133+ TSCs with respect to the standard glioblastoma model, established glioma cell lines, has not been determined. Therefore, to better understand the radioresponse of this cancer stem cell, we have used established cell lines as a framework for defining their in vitro radioresponse., Experimental Design: The intrinsic radiosensitivity of CD133+ TSC cultures and established glioma cell lines was determined by clonogenic assay. The TSCs and established cell lines were also compared in terms of DNA double-strand break (DSB) repair capacity and cell cycle checkpoint activation., Results: Based on clonogenic analysis, each of the six TSC cultures evaluated was more sensitive to radiation than the established glioma cell lines. Consistent with increased radiosensitivity, the DSB repair capacity as defined by neutral comet assay and gammaH2AX and Rad51 foci was significantly reduced in TSCs compared with the cell lines. Although G2 checkpoint activation was intact, in contrast to the cell lines, DNA synthesis was not inhibited in TSCs after irradiation, indicating the absence of the intra-S-phase checkpoint., Conclusions: These data indicate that the mechanisms through which CD133+ TSCs respond to radiation are significantly different from those of the traditional glioblastoma in vitro model, established glioma cell lines. If TSCs play a critical role in glioblastoma treatment response, then such differences are likely to be of consequence in the development and testing of radiosensitizing agents.
- Published
- 2009
- Full Text
- View/download PDF
39. Urine analysis and protein networking identify met as a marker of metastatic prostate cancer.
- Author
-
Russo AL, Jedlicka K, Wernick M, McNally D, Kirk M, Sproull M, Smith S, Shankavaram U, Kaushal A, Figg WD, Dahut W, Citrin D, Bottaro DP, Albert PS, Tofilon PJ, and Camphausen K
- Subjects
- Biomarkers, Tumor analysis, Biomarkers, Tumor metabolism, Biomarkers, Tumor physiology, Biomarkers, Tumor urine, Enzyme-Linked Immunosorbent Assay, Humans, Male, Neoplasm Metastasis, Neoplasm Staging methods, Prognosis, Prostatic Neoplasms diagnosis, Proteins analysis, Proteins metabolism, Proto-Oncogene Proteins analysis, Proto-Oncogene Proteins physiology, Proto-Oncogene Proteins c-met, ROC Curve, Receptors, Growth Factor analysis, Receptors, Growth Factor physiology, Tumor Cells, Cultured, Metabolic Networks and Pathways physiology, Prostatic Neoplasms pathology, Prostatic Neoplasms urine, Proto-Oncogene Proteins urine, Urinalysis methods
- Abstract
Purpose: Metastatic prostate cancer is a major cause of death of men in the United States. Expression of met, a receptor tyrosine kinase, has been associated with progression of prostate cancer., Experimental Design: To investigate met as a biomarker of disease progression, urinary met was evaluated via ELISA in men with localized (n = 75) and metastatic (n = 81) prostate cancer. Boxplot analysis was used to compare the distribution of met values between each group. We estimated a receiver operating characteristic curve and the associated area under the curve to summarize the diagnostic accuracy of met for distinguishing between localized and metastatic disease. Protein-protein interaction networking via yeast two-hybrid technology supplemented by Ingenuity Pathway Analysis and Human Interactome was used to elucidate proteins and pathways related to met that may contribute to progression of disease., Results: Met distribution was significantly different between the metastatic group and the group with localized prostate cancer and people with no evidence of cancer (P < 0.0001). The area under the curve for localized and metastatic disease was 0.90, with a 95% confidence interval of 0.84 to 0.95. Yeast two-hybrid technology, Ingenuity Pathway Analysis, and Human Interactome identified 89 proteins that interact with met, of which 40 have previously been associated with metastatic prostate cancer., Conclusion: Urinary met may provide a noninvasive biomarker indicative of metastatic prostate cancer and may be a central regulator of multiple pathways involved in prostate cancer progression.
- Published
- 2009
- Full Text
- View/download PDF
40. Physiologic oxygen concentration enhances the stem-like properties of CD133+ human glioblastoma cells in vitro.
- Author
-
McCord AM, Jamal M, Shankavaram UT, Lang FF, Camphausen K, and Tofilon PJ
- Subjects
- AC133 Antigen, Basic Helix-Loop-Helix Transcription Factors genetics, Basic Helix-Loop-Helix Transcription Factors metabolism, Biomarkers, Tumor genetics, Biomarkers, Tumor metabolism, Brain Neoplasms genetics, Brain Neoplasms metabolism, Cell Differentiation, Cell Proliferation, Flow Cytometry, Gene Expression Profiling, Gene Expression Regulation, Neoplastic, Glioblastoma genetics, Glioblastoma metabolism, Humans, Immunoblotting, In Vitro Techniques, Neoplastic Stem Cells metabolism, Oligonucleotide Array Sequence Analysis, RNA, Small Interfering pharmacology, Tumor Cells, Cultured, Tumor Stem Cell Assay, Antigens, CD metabolism, Brain Neoplasms pathology, Glioblastoma pathology, Glycoproteins metabolism, Neoplastic Stem Cells pathology, Oxygen metabolism, Peptides metabolism
- Abstract
In vitro investigations of tumor stem-like cells (TSC) isolated from human glioblastoma (GB) surgical specimens have been done primarily at an atmospheric oxygen level of 20%. To determine whether an oxygen level more consistent with in situ conditions affects their stem cell-like characteristics, we compared GB TSCs grown under conditions of 20% and 7% oxygen. Growing CD133(+) cells sorted from three GB neurosphere cultures at 7% O(2) reduced their doubling time and increased the self-renewal potential as reflected by clonogenicity. Furthermore, at 7% oxygen, the cultures exhibited an enhanced capacity to differentiate along both the glial and neuronal pathways. As compared with 20%, growth at 7% oxygen resulted in an increase in the expression levels of the neural stem cell markers CD133 and nestin as well as the stem cell markers Oct4 and Sox2. In addition, whereas hypoxia inducible factor 1alpha was not affected in CD133(+) TSCs grown at 7% O(2), hypoxia-inducible factor 2alpha was expressed at higher levels as compared with 20% oxygen. Gene expression profiles generated by microarray analysis revealed that reducing oxygen level to 7% resulted in the up-regulation and down-regulation of a significant number of genes, with more than 140 being commonly affected among the three CD133(+) cultures. Furthermore, Gene Ontology categories up-regulated at 7% oxygen included those associated with stem cells or GB TSCs. Thus, the data presented indicate that growth at the more physiologically relevant oxygen level of 7% enhances the stem cell-like phenotype of CD133(+) GB cells.
- Published
- 2009
- Full Text
- View/download PDF
41. In vitro and in vivo radiosensitization of glioblastoma cells by the poly (ADP-ribose) polymerase inhibitor E7016.
- Author
-
Russo AL, Kwon HC, Burgan WE, Carter D, Beam K, Weizheng X, Zhang J, Slusher BS, Chakravarti A, Tofilon PJ, and Camphausen K
- Subjects
- Animals, Apoptosis, Cell Line, Tumor, Comet Assay, DNA Repair, Dacarbazine analogs & derivatives, Dacarbazine pharmacology, Humans, In Vitro Techniques, Mice, Mitosis, Poly(ADP-ribose) Polymerases metabolism, Radiotherapy methods, Temozolomide, Brain Neoplasms drug therapy, Enzyme Inhibitors pharmacology, Glioblastoma drug therapy, Poly(ADP-ribose) Polymerase Inhibitors
- Abstract
Purpose: Poly (ADP-ribose) polymerase (PARP) inhibitors are undergoing clinical evaluation for cancer therapy. Because PARP inhibition has been shown to enhance tumor cell sensitivity to radiation, we investigated the in vitro and in vivo effects of the novel PARP inhibitor E7016., Experimental Design: The effect of E7016 on the in vitro radiosensitivity of tumor cell lines was evaluated using clonogenic survival. DNA damage and repair were measured using gammaH2AX foci and neutral comet assay. Mitotic catastrophe was determined by immunostaining. Tumor growth delay was evaluated in mice for the effect of E7016 on in vivo (U251) tumor radiosensitivity., Results: Cell lines exposed to E7016 preirradiation yielded an increase in radiosensitivity with dose enhancement factors at a surviving fraction of 0.1 from 1.4 to 1.7. To assess DNA double-strand breaks repair, gammaH2AX measured at 24 hours postirradiation had significantly more foci per cell in the E7016/irradiation group versus irradiation alone. Neutral comet assay further suggested unrepaired double-strand breaks with significantly greater DNA damage at 6 hours postirradiation in the combination group versus irradiation alone. Mitotic catastrophe staining revealed a significantly greater number of cells staining positive at 24 hours postirradiation in the combination group. In vivo, mice treated with E7016/irradiation/temozolomide had an additional growth delay of six days compared with the combination of temozolomide and irradiation., Conclusions: These results indicate that E7016 can enhance tumor cell radiosensitivity in vitro and in vivo through the inhibition of DNA repair. Moreover, enhanced growth delay with the addition of E7016 to temozolomide and radiotherapy in a glioma mouse model suggests a potential role for this drug in the treatment of glioblastoma multiforme.
- Published
- 2009
- Full Text
- View/download PDF
42. The prognostic value of nestin expression in newly diagnosed glioblastoma: report from the Radiation Therapy Oncology Group.
- Author
-
Chinnaiyan P, Wang M, Rojiani AM, Tofilon PJ, Chakravarti A, Ang KK, Zhang HZ, Hammond E, Curran W Jr, and Mehta MP
- Subjects
- Antineoplastic Agents, Alkylating therapeutic use, Brain Neoplasms drug therapy, Cell Lineage, Combined Modality Therapy methods, Dacarbazine analogs & derivatives, Dacarbazine therapeutic use, Disease-Free Survival, Glioblastoma drug therapy, Humans, Nestin, Neurons metabolism, Phenotype, Prognosis, Research Design, Temozolomide, Treatment Outcome, Brain Neoplasms metabolism, Brain Neoplasms radiotherapy, Gene Expression Regulation, Neoplastic, Glioblastoma metabolism, Glioblastoma radiotherapy, Intermediate Filament Proteins biosynthesis, Nerve Tissue Proteins biosynthesis
- Abstract
Background: Nestin is an intermediate filament protein that has been implicated in early stages of neuronal lineage commitment. Based on the heterogeneous expression of nestin in GBM and its potential to serve as a marker for a dedifferentiated, and perhaps more aggressive phenotype, the Radiation Therapy Oncology Group (RTOG) sought to determine the prognostic value of nestin expression in newly diagnosed GBM patients treated on prior prospective RTOG clinical trials., Methods: Tissue microarrays were prepared from 156 patients enrolled in these trials. These specimens were stained using a mouse monoclonal antibody specific for nestin and expression was measured by computerized quantitative image analysis using the Ariol SL-50 system. The parameters measured included both staining intensity and the relative area of expression within a specimen. This resulted into 3 categories: low, intermediate, and high nestin expression, which was then correlated with clinical outcome., Results: A total of 153 of the 156 samples were evaluable for this study. There were no statistically significant differences between pretreatment patient characteristics and nestin expression. There was no statistically significant difference in either overall survival or progression-free survival (PFS) demonstrated, although a trend in decreased PFS was observed with high nestin expression (p = 0.06)., Conclusion: Although the correlation of nestin expression and histologic grade in glioma is of considerable interest, the presented data does not support its prognostic value in newly diagnosed GBM. Further studies evaluating nestin expression may be more informative when studied in lower grade glioma, in the context of markers more specific to tumor stem cells, and using more recent specimens from patients treated with temozolomide in conjunction with radiation.
- Published
- 2008
- Full Text
- View/download PDF
43. Postradiation sensitization of the histone deacetylase inhibitor valproic acid.
- Author
-
Chinnaiyan P, Cerna D, Burgan WE, Beam K, Williams ES, Camphausen K, and Tofilon PJ
- Subjects
- Brain Neoplasms, Cell Line, Tumor, Humans, Valproic Acid administration & dosage, Enzyme Inhibitors pharmacology, Glioma, Histone Deacetylase Inhibitors, Radiation-Sensitizing Agents pharmacology, Valproic Acid pharmacology
- Abstract
Purpose: Preclinical studies evaluating histone deacetylase (HDAC) inhibitor-induced radiosensitization have largely focused on the preirradiation setting based on the assumption that enhanced radiosensitivity was mediated by changes in gene expression. Our previous investigations identified maximal radiosensitization when cells were exposed to HDAC inhibitors in both the preradiation and postradiation setting. We now expand on these studies to determine whether postirradiation exposure alone affects radiosensitivity., Experimental Design: The effects of the HDAC inhibitor valproic acid (VA) on postirradiation sensitivity in human glioma cell lines were evaluated using a clonogenic assay, exposing cells to VA up to 24 h after irradiation. DNA damage repair was evaluated using gammaH2AX and 53BP1 foci and cell cycle phase distribution was analyzed by flow cytometry. Western blot of acetylated gammaH2AX was done following histone extraction on AUT gels., Results: VA enhanced radiosensitivity when delivered up to 24 h after irradiation. Cells accumulated in G(2)-M following irradiation, although they returned to baseline at 24 h, mitigating the role of cell cycle redistribution in postirradiation sensitization by VA. At 12 h after irradiation, significant gammaH2AX and 53BP1 foci dispersal was shown in the control, although cells exposed to VA after irradiation maintained foci expression. VA alone had no effect on the acetylation or phosphorylation of H2AX, although it did acetylate radiation-induced gammaH2AX., Conclusions: These results indicate that VA enhances radiosensitivity at times up to 24 h after irradiation, which has direct clinical application.
- Published
- 2008
- Full Text
- View/download PDF
44. Radiation-induced gene translation profiles reveal tumor type and cancer-specific components.
- Author
-
Kumaraswamy S, Chinnaiyan P, Shankavaram UT, Lü X, Camphausen K, and Tofilon PJ
- Subjects
- Cell Line, Tumor, False Positive Reactions, Gene Expression Profiling, Glioma radiotherapy, Humans, Models, Biological, Neoplasms radiotherapy, Oligonucleotide Array Sequence Analysis, Polyribosomes metabolism, RNA metabolism, Gene Expression Regulation, Neoplastic, Glioma genetics, Glioma metabolism, Neoplasms genetics, Neoplasms metabolism, Protein Biosynthesis
- Abstract
The microarray analysis of total cellular RNA is a common method used in the evaluation of radiation-induced gene expression. However, profiling the cellular transcriptome does not take into account posttranscriptional processes that affect gene expression. To better define the genes whose expression is influenced by ionizing radiation, we used polysome-bound RNA to generate gene translation profiles for a series of tumor and normal cell lines. Cell lines were exposed to 2 Gy, polysome-bound RNA isolated 6 hours later, and then subjected to microarray analysis. To identify the genes whose translation was affected by radiation, the polysome-bound RNA profiles were compared with their corresponding controls using significance analysis of microarrays (<1% false discovery rate). From the statistically significant genes identified for each cell line, hierarchical clustering was performed by average linkage measurement and Pearson's correlation metric. Ingenuity Pathway Analysis was used for distributing genes into biological networks and for evaluation of functional significance. Radiation-induced gene translation profiles clustered according to tissue of origin; the cell lines corresponding to each tissue type contained a significant number of commonly affected genes. Network analyses suggested that the biological functions associated with the genes whose translation was affected by radiation were tumor type-specific. There was also a set of genes/networks that were unique to tumor or normal cells. These results indicate that radiation-induced gene translation profiles provide a unique data set for the analysis of cellular radioresponse and suggest a framework for identifying and targeting differences in the regulation of tumor and normal cell radiosensitivity.
- Published
- 2008
- Full Text
- View/download PDF
45. In vitro and in vivo radiosensitization induced by the DNA methylating agent temozolomide.
- Author
-
Kil WJ, Cerna D, Burgan WE, Beam K, Carter D, Steeg PS, Tofilon PJ, and Camphausen K
- Subjects
- Antineoplastic Agents, Alkylating pharmacology, Apoptosis drug effects, Brain Neoplasms, Cell Cycle drug effects, Cell Line, Tumor, DNA Methylation, DNA Replication drug effects, Dacarbazine pharmacology, Glioblastoma, Humans, Mitosis drug effects, Temozolomide, Tumor Stem Cell Assay, Dacarbazine analogs & derivatives, Radiation-Sensitizing Agents pharmacology
- Abstract
Purpose: Temozolomide, a DNA methylating agent, is currently undergoing clinical evaluation for cancer therapy. Because temozolomide has been shown to increase survival rates of patients with malignant gliomas when given combined with radiation, and there is conflicting preclinical data concerning the radiosensitizing effects of temozolomide, we further investigated the possible temozolomide-induced enhancement of radiosensitivity., Experimental Design: The effects of temozolomide on the in vitro radiosensitivity of U251 (a human glioma) and MDA-MB231BR (a brain-seeking variant of a human breast tumor) cell lines was evaluated using clonogenic assay. DNA damage and repair were evaluated using phosphorylated histone H2AX (gammaH2AX), and mitotic catastrophe was measured using nuclear fragmentation. Growth delay was used to evaluate the effects of temozolomide on in vivo (U251) tumor radiosensitivity., Results: Exposure of each cell line to temozolomide for 1 h before irradiation resulted in an increase in radiosensitivity with dose enhancement factors at a surviving fraction of 0.1 ranging from 1.30 to 1.32. Temozolomide had no effect on radiation-induced apoptosis or on the activation of the G(2) cell cycle checkpoint. As a measure of DNA double strand breaks, gammaH2AX foci were determined as a function of time after the temozolomide + irradiation combination. The number of gammaH2AX foci per cell was significantly greater at 24 h after the combined modality compared with the individual treatments. Mitotic catastrophe, measured at 72 h, was also significantly increased in cells receiving the temozolomide + irradiation combination compared with the single treatments. In vivo studies revealed that temozolomide administration to mice bearing U251 tumor xenografts resulted in a greater than additive increase in radiation-induced tumor growth delay with a dose enhancement factor of 2.8., Conclusions: These results indicate that temozolomide can enhance tumor cell radiosensitivity in vitro and in vivo and suggest that this effect involves an inhibition of DNA repair leading to an increase in mitotic catastrophe.
- Published
- 2008
- Full Text
- View/download PDF
46. Inhibition of Hsp90: a multitarget approach to radiosensitization.
- Author
-
Camphausen K and Tofilon PJ
- Subjects
- Animals, Humans, Neoplasms pathology, HSP90 Heat-Shock Proteins antagonists & inhibitors, Neoplasms radiotherapy, Radiation Tolerance, Radiation-Sensitizing Agents pharmacology
- Abstract
Hsp90, the 90 kDa heat shock protein, is a highly expressed molecular chaperone that modulates the stability and/or transport of a diverse set of critical cellular regulatory proteins. Among Hsp90 clients are a number of proteins, which in a cell type-dependent manner, contribute to tumor cell radioresistance. Exposure of a variety of solid tumor cell lines to clinically relevant Hsp90 inhibitors results in the simultaneous loss of these radioresponse-associated proteins, which is accompanied by an increase in radiosensitivity. This radiosensitization has been linked to a compromise in the DNA damage response to radiation including the inhibition of cell cycle checkpoint activation and DNA double-strand break repair. With respect to potential clinical application, the expression of ErbB3 seems to predict tumor cells that are resistant to the effects of Hsp90 inhibition on radiosensitivity. Moreover, whereas an increase in tumor cell radiosensitivity was consistently reported, the radiosensitivity of normal fibroblasts was not affected by Hsp90 inhibition, suggesting the potential for tumor-selective radiosensitization. This review summarizes the preclinical data available on Hsp90 inhibition and cellular radiosensitivity. Results generated to date suggest that Hsp90 inhibition can provide a multitarget approach to tumor radiosensitization.
- Published
- 2007
- Full Text
- View/download PDF
47. Inhibition of hsp90 compromises the DNA damage response to radiation.
- Author
-
Dote H, Burgan WE, Camphausen K, and Tofilon PJ
- Subjects
- Cell Cycle drug effects, Cell Cycle radiation effects, Cell Line, Tumor, DNA Repair drug effects, DNA, Neoplasm radiation effects, DNA-Activated Protein Kinase metabolism, Enzyme Activation radiation effects, Humans, Pancreatic Neoplasms enzymology, Pancreatic Neoplasms pathology, Benzoquinones pharmacology, DNA Damage physiology, HSP90 Heat-Shock Proteins antagonists & inhibitors, Lactams, Macrocyclic pharmacology, Pancreatic Neoplasms genetics, Pancreatic Neoplasms radiotherapy, Radiation-Sensitizing Agents pharmacology
- Abstract
Inhibitors of the molecular chaperone Hsp90 have been shown to enhance tumor cell radiosensitivity. To begin to address the mechanism responsible, we have determined the effect of the Hsp90 inhibitor 17-(dimethylaminoethylamino)-17-demethoxygeldanamycin (17DMAG) on the DNA damage response to radiation. Exposure of MiaPaCa tumor cells to 17DMAG, which results in radiosensitization, inhibited the repair of DNA double-strand breaks according to gammaH2AX foci dispersal and the neutral comet assay. This repair inhibition was associated with reduced DNA-PK catalytic subunit (DNA-PKcs) phosphorylation after irradiation and a disruption of DNA-PKcs/ErbB1 interaction. These data suggest that the previously established 17DMAG-mediated reduction in ErbB1 activity reduces its interaction with DNA-PKcs and thus accounts for the attenuation of radiation-induced DNA-PK activation. 17DMAG was also found to abrogate the activation of the G(2)- and S-phase cell cycle checkpoints. Associated with these events was a reduction in radiation-induced ataxia-telangiectasia mutated (ATM) activation and foci formation in 17DMAG-treated cells. Although no interaction between ATM and Hsp90 was detected, Hsp90 was found to interact with the MRE11/Rad50/NBS1 (MRN) complex. 17DMAG exposure reduced the ability of the MRN components to form nuclear foci after irradiation. Moreover, 17DMAG exposure reduced the interaction between NBS1 and ATM, although no degradation of the MRN complex was detected. These results suggest that the diminished radiation-induced activation of ATM in 17DMAG-treated cells was the result of a compromise in the function of the MRN complex. These data indicate that Hsp90 can contribute to the DNA damage response to radiation affecting both DNA repair and cell cycle checkpoint activation.
- Published
- 2006
- Full Text
- View/download PDF
48. Inhibition of Akt by the alkylphospholipid perifosine does not enhance the radiosensitivity of human glioma cells.
- Author
-
de la Peña L, Burgan WE, Carter DJ, Hollingshead MG, Satyamitra M, Camphausen K, and Tofilon PJ
- Subjects
- Animals, Apoptosis drug effects, Apoptosis radiation effects, Brain Neoplasms drug therapy, Combined Modality Therapy, Female, Glioma drug therapy, Humans, Immunohistochemistry, Mice, Mice, Nude, Phosphorylation, Phosphorylcholine pharmacology, Transplantation, Heterologous, Tumor Cells, Cultured, Tumor Stem Cell Assay, Brain Neoplasms radiotherapy, Glioma radiotherapy, Phosphorylcholine analogs & derivatives, Proto-Oncogene Proteins c-akt antagonists & inhibitors, Radiation Tolerance drug effects
- Abstract
Akt has been implicated as a molecular determinant of cellular radiosensitivity. Because it is often constitutively activated or overexpressed in malignant gliomas, it has been suggested as a target for brain tumor radiosensitization. To evaluate the role of Akt in glioma radioresponse, we have determined the effects of perifosine, a clinically relevant alkylphospholipid that inhibits Akt activation, on the radiosensitivity of three human glioma cell lines (U87, U251, and LN229). Each of the glioma cell lines expressed clearly detectable levels of phosphorylated Akt indicative of constitutive Akt activity. Exposure to a perifosine concentration that reduced survival by approximately 50% significantly reduced the level of phosphorylated Akt as well as Akt activity. Cell survival analysis using a clonogenic assay, however, revealed that this Akt-inhibiting perifosine treatment did not enhance the radiosensitivity of the glioma cell lines. This evaluation was then extended to an in vivo model using U251 xenografts. Perifosine delivered to mice bearing U251 xenografts substantially reduced tumor phosphorylated Akt levels and inhibited tumor growth rate. However, the combination of perifosine and radiation resulted in a less than additive increase in tumor growth delay. Thus, in vitro and in vivo data indicate that the perifosine-mediated decrease in Akt activity does not enhance the radiosensitivity of three genetically disparate glioma cell lines. These results suggest that, although Akt may influence the radiosensitivity of other tumor types, it does not seem to be a target for glioma cell radiosensitization.
- Published
- 2006
- Full Text
- View/download PDF
49. In vitro and in vivo radiosensitization induced by the ribonucleotide reductase inhibitor Triapine (3-aminopyridine-2-carboxaldehyde-thiosemicarbazone).
- Author
-
Barker CA, Burgan WE, Carter DJ, Cerna D, Gius D, Hollingshead MG, Camphausen K, and Tofilon PJ
- Subjects
- Cell Cycle radiation effects, Cell Line, Tumor, Dose-Response Relationship, Radiation, Enzyme Inhibitors pharmacology, Glioma, Humans, Male, Pancreatic Neoplasms, Prostatic Neoplasms, Ribonucleotide Reductases antagonists & inhibitors, Cell Cycle drug effects, Pyridines pharmacology, Radiation-Sensitizing Agents pharmacology, Thiosemicarbazones pharmacology
- Abstract
Purpose: Because ribonucleotide reductase (RR) plays a role in DNA repair, it may serve as a molecular target for radiosensitization. Unlike previously investigated RR inhibitors, Triapine potently inhibits both RR holoenzymes. Therefore, the effects of Triapine on tumor cell radiosensitivity were investigated., Experimental Design: The effects of Triapine on the in vitro radiosensitivity of three human tumor cell lines and one normal cell line were evaluated using a clonogenic assay. Growth delay was used to evaluate the effects of Triapine on in vivo tumor radiosensitivity. The levels of the RR subunits were determined using immunoblot analysis and DNA damage and repair were evaluated using gammaH2AX foci., Results: Exposure of the tumor cell lines to Triapine before or immediately after irradiation resulted in an increase in radiosensitivity. In contrast, Triapine enhanced the radiosensitivity of the normal fibroblast cell line only when the exposure was before irradiation. There were no consistent differences between cell lines with respect to the expression of the RR subunits. Whereas Triapine had no effect on radiation-induced gammaH2AX foci at 1 hour, the number of gammaH2AX foci per cell was significantly greater in the Triapine-treated cells at 24 hours after irradiation, suggesting the presence of unrepaired DNA damage. Triapine administration to mice bearing tumor xenografts immediately after irradiation resulted in a greater than additive increase in radiation-induced tumor growth delay., Conclusions: These results indicate that Triapine can enhance tumor cell radiosensitivity in vitro and in vivo and suggest that this effect involves an inhibition of DNA repair.
- Published
- 2006
- Full Text
- View/download PDF
50. Radiation-induced changes in gene expression involve recruitment of existing messenger RNAs to and away from polysomes.
- Author
-
Lü X, de la Peña L, Barker C, Camphausen K, and Tofilon PJ
- Subjects
- Astrocytes radiation effects, Gene Expression Profiling, Humans, Oligonucleotide Array Sequence Analysis, Polyribosomes, RNA, Radiation, Ionizing, Transcription, Genetic radiation effects, Brain Neoplasms genetics, Gene Expression Regulation, Neoplastic radiation effects, Protein Biosynthesis radiation effects
- Abstract
Although ionizing radiation has been shown to influence gene transcription, little is known about the effects of radiation on gene translational efficiency. To obtain a genome-wide perspective of the effects of radiation on gene translation, microarray analysis was done on polysome-bound RNA isolated from irradiated human brain tumor cells; to allow for a comparison with the effects of radiation on transcription, microarray analysis was also done using total RNA. The number of genes whose translational activity was modified by radiation was approximately 10-fold greater than those whose transcription was affected. The radiation-induced change in a gene's translational activity was shown to involve the recruitment of existing mRNAs to and away from polysomes. Moreover, the change in a gene's translational activity after irradiation correlated with changes in the level of its corresponding protein. These data suggest that radiation modifies gene expression primarily at the level of translation. In contrast to transcriptional changes, there was considerable overlap in the genes affected at the translational level among brain tumor cell lines and normal astrocytes. Thus, the radiation-induced translational control of a subset of mRNAs seems to be a fundamental component of cellular radioresponse.
- Published
- 2006
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.