Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Mol Cancer Ther ; 17(2): 407-418, 2018 02.
Article in English | MEDLINE | ID: mdl-29030460

ABSTRACT

Despite significant advances in combinations of radiotherapy and chemotherapy, altered fractionation schedules and image-guided radiotherapy, many cancer patients fail to benefit from radiation. A prevailing hypothesis is that targeting repair of DNA double strand breaks (DSB) can enhance radiation effects in the tumor and overcome therapeutic resistance without incurring off-target toxicities. Unrepaired DSBs can block cancer cell proliferation, promote cancer cell death, and induce cellular senescence. Given the slow progress to date translating novel DSB repair inhibitors as radiosensitizers, we have explored drug repurposing, a proven route to improving speed, costs, and success rates of drug development. In a prior screen where we tracked resolution of ionizing radiation-induced foci (IRIF) as a proxy for DSB repair, we had identified pitavastatin (Livalo), an HMG-CoA reductase inhibitor commonly used for lipid lowering, as a candidate radiosensitizer. Here, we report that pitavastatin and other lipophilic statins are potent inhibitors of DSB repair in breast and melanoma models both in vitro and in vivo When combined with ionizing radiation, pitavastatin increased persistent DSBs, induced senescence, and enhanced acute effects of radiation on radioresistant melanoma tumors. shRNA knockdown implicated HMG-CoA reductase, farnesyl diphosphate synthase, and protein farnesyl transferase in IRIF resolution, DSB repair, and senescence. These data confirm on-target activity of statins, although via inhibition of protein prenylation rather than cholesterol biosynthesis. In light of prior studies demonstrating enhanced efficacy of radiotherapy in patients taking statins, this work argues for clinical evaluation of lipophilic statins as nontoxic radiosensitizers to enhance the benefits of image-guided radiotherapy. Mol Cancer Ther; 17(2); 407-18. ©2017 AACRSee all articles in this MCT Focus section, "Developmental Therapeutics in Radiation Oncology."


Subject(s)
DNA Repair/drug effects , Acyl Coenzyme A/pharmacology , Animals , Cellular Senescence , Female , Humans , Mice
2.
Oncotarget ; 7(23): 33919-33, 2016 Jun 07.
Article in English | MEDLINE | ID: mdl-27129153

ABSTRACT

Radiation therapy remains a significant therapeutic modality in the treatment of cancer. An attractive strategy would be to enhance the benefits of ionizing radiation (IR)with radiosensitizers. A high-content drug repurposing screen of approved and investigational agents, natural products and other small molecules has identified multiple candidates that blocked repair of IR damage in vitro. Here, we validated a subset of these hits in vitro and then examined effects on tumor growth after IR in a murine tumor model. Based on robust radiosensitization in vivo and other favorable properties of cephalexin, we conducted additional studies with other beta-lactam antibiotics. When combined with IR, each cephalosporin tested increased DNA damage and slowed tumor growth without affecting normal tissue toxicity. Our data implicate reactive oxygen species in the mechanism by which cephalosporins augment the effects of IR. This work provides a rationale for using commonly prescribed beta-lactam antibiotics as non-toxic radiosensitizers to enhance the therapeutic ratio of radiotherapy.


Subject(s)
Anti-Bacterial Agents/pharmacology , Breast Neoplasms/radiotherapy , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Cellular Senescence/drug effects , Cellular Senescence/radiation effects , Cephalosporins/pharmacology , Drug Repositioning , Radiation-Sensitizing Agents/pharmacology , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , DNA Damage/drug effects , DNA Damage/radiation effects , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Female , Humans , MCF-7 Cells , Mice, Inbred C57BL , Oxidative Stress/drug effects , Oxidative Stress/radiation effects , Reactive Oxygen Species/metabolism , Time Factors , Tumor Burden/drug effects , Tumor Burden/radiation effects , Xenograft Model Antitumor Assays
3.
Mol Cancer Res ; 14(2): 173-84, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26538285

ABSTRACT

UNLABELLED: Conventional wisdom ascribes metabolic reprogramming in cancer to meeting increased demands for intermediates to support rapid proliferation. Prior models have proposed benefits toward cell survival, immortality, and stress resistance, although the recent discovery of oncometabolites has shifted attention to chromatin targets affecting gene expression. To explore further effects of cancer metabolism and epigenetic deregulation, DNA repair kinetics were examined in cells treated with metabolic intermediates, oncometabolites, and/or metabolic inhibitors by tracking resolution of double-strand breaks (DSB) in irradiated MCF7 breast cancer cells. Disrupting cancer metabolism revealed roles for both glycolysis and glutaminolysis in promoting DSB repair and preventing accelerated senescence after irradiation. Targeting pathways common to glycolysis and glutaminolysis uncovered opposing effects of the hexosamine biosynthetic pathway (HBP) and tricarboxylic acid (TCA) cycle. Treating cells with the HBP metabolite N-acetylglucosamine (GlcNAc) or augmenting protein O-GlcNAcylation with small molecules or RNAi targeting O-GlcNAcase each enhanced DSB repair, while targeting O-GlcNAc transferase reversed GlcNAc's effects. Opposing the HBP, TCA metabolites including α-ketoglutarate blocked DSB resolution. Strikingly, DNA repair could be restored by the oncometabolite 2-hydroxyglutarate (2-HG). Targeting downstream effectors of histone methylation and demethylation implicated the PRC1/2 polycomb complexes as the ultimate targets for metabolic regulation, reflecting known roles for Polycomb group proteins in nonhomologous end-joining DSB repair. Our findings that epigenetic effects of cancer metabolic reprogramming may promote DNA repair provide a molecular mechanism by which deregulation of metabolism may not only support cell growth but also maintain cell immortality, drive therapeutic resistance, and promote genomic instability. IMPLICATIONS: By defining a pathway from deregulated metabolism to enhanced DNA damage response in cancer, these data provide a rationale for targeting downstream epigenetic effects of metabolic reprogramming to block cancer cell immortality and overcome resistance to genotoxic stress.


Subject(s)
DNA Repair , Epigenesis, Genetic , Glutamine/metabolism , Glycolysis , Neoplasms/metabolism , Acetylglucosamine/pharmacology , Cellular Senescence , DNA/radiation effects , Genomic Instability , Glycolysis/drug effects , Humans , MCF-7 Cells , Neoplasms/genetics , RNA Interference
4.
PLoS One ; 7(10): e46104, 2012.
Article in English | MEDLINE | ID: mdl-23056240

ABSTRACT

BACKGROUND: Vascular endothelial cells contribute to the pathogenesis of numerous human diseases by actively regulating the stromal inflammatory response; however, little is known regarding the role of endothelial inflammation in the growth of human tumors and its influence on the prognosis of human cancers. METHODS: Using an experimental model of tumor necrosis factor-alpha (TNF-α)-mediated inflammation, we characterized inflammatory gene expression in immunopurified tumor-associated endothelial cells. These genes formed the basis of a multivariate molecular predictor of overall survival that was trained and validated in four types of human cancer. RESULTS: We report that expression of experimentally derived tumor endothelial genes distinguished pathologic tissue specimens from normal controls in several human diseases associated with chronic inflammation. We trained these genes in human cancer datasets and defined a six-gene inflammatory signature that predicted significantly reduced overall survival in breast cancer, colon cancer, lung cancer, and glioma. This endothelial-derived signature predicted outcome independently of, but cooperatively with, standard clinical and pathological prognostic factors. Consistent with these findings, conditioned culture media from human endothelial cells stimulated by pro-inflammatory cytokines accelerated the growth of human colon and breast tumors in immunodeficient mice as compared with conditioned media from untreated endothelial cells. CONCLUSIONS: This study provides the first prognostic cancer gene signature derived from an experimental model of tumor-associated endothelial inflammation. These findings support the notion that activation of inflammatory pathways in non-malignant tumor-infiltrating endothelial cells contributes to tumor growth and progression in multiple human cancers. Importantly, these results identify endothelial-derived factors that could serve as potential targets for therapy in diverse human cancers.


Subject(s)
Endothelium, Vascular/metabolism , Inflammation/genetics , Neoplasms/genetics , Neovascularization, Pathologic/genetics , Adult , Aged , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cells, Cultured , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelium, Vascular/pathology , Female , Gene Expression Profiling , Glioma/genetics , Glioma/pathology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Inflammation/pathology , Kaplan-Meier Estimate , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Mice , Middle Aged , Multivariate Analysis , Neoplasms/blood supply , Neoplasms/pathology , Neovascularization, Pathologic/pathology , Oligonucleotide Array Sequence Analysis , Tumor Necrosis Factor-alpha/pharmacology
5.
Int J High Throughput Screen ; 2: 1-13, 2011 Mar.
Article in English | MEDLINE | ID: mdl-26097382

ABSTRACT

Much like replicative senescence, the irreversible cell-cycle arrest induced by eroded telomeres, accelerated senescence occurs when replicative cells suffer irreparable DNA double-strand breaks (DSBs). Along with apoptosis and necrosis, senescence is a desirable outcome in cancer treatment with ionizing radiation (IR) or chemotherapy. In both normal and cancer cells, DSBs promote the assembly of IR-induced foci (IRIF), domains of modified chromatin that serve a key role in DNA damage signaling. IRIF persistence is a critical determinant of accelerated senescence, making drugs that promote persistent IRIF an attractive strategy to sensitize cancer to genotoxic therapy. As an IRIF reporter, we have expressed an inducible green fluorescent protein (GFP) fusion to the IRIF-binding domain (IBD) of 53BP1 (GFP-IBD) in the breast cancer cell line MCF7. Within minutes of exposure to IR, the GFP-IBD relocalizes to form fluorescent nuclear foci, which disperse within several hours. A pair of high-content screening assays for IRIF formation and persistence were established in multiwell plates based on imaging and quantifying GFP-IBD foci per Hoechst-stained MCF7 nucleus at 2 hours and 24 hours. Using the ataxia telangiectasia-mutated inhibitor CGK733 to block IRIF formation and the topoisomerase II inhibitor etoposide to prevent IRIF resolution, we obtained a Z' >0.8 both for IRIF formation at 2 hours and IRIF persistence at 24 hours. Screening the diverse drugs and natural products in the National Cancer Institute Developmental Therapeutics Program Approved Oncology Drugs Set, the National Institutes of Health Clinical Collection, and the MicroSource Spectrum Collection yielded multiple hits that significantly delayed IRIF resolution. Secondary screening suggested some of these otherwise nontoxic drugs also enhance accelerated senescence, indicating strong potential for their repurposing as radiation sensitizers to improve the efficacy of cancer therapy.

6.
Cancer Res ; 70(15): 6277-82, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20610628

ABSTRACT

Persistent DNA double-strand breaks (DSB) may determine the antitumor effects of ionizing radiation (IR) by inducing apoptosis, necrosis, mitotic catastrophe, or permanent growth arrest. IR induces rapid modification of megabase chromatin domains surrounding DSBs via poly-ADP-ribosylation, phosphorylation, acetylation, and protein assembly. The dynamics of these IR-induced foci (IRIF) have been implicated in DNA damage signaling and DNA repair. As an IRIF reporter, we tracked the relocalization of green fluorescent protein fused to a chromatin binding domain of the checkpoint adapter protein 53BP1 after IR of breast cancer cells and tumors. To block DSB repair in breast cancer cells and tumors, we targeted poly(ADP-ribose) polymerase (PARP) with ABT-888 (veliparib), one of several PARP inhibitors currently in clinical trials. PARP inhibition markedly enhanced IRIF persistence and increased breast cancer cell senescence both in vitro and in vivo, arguing for targeting IRIF resolution as a novel therapeutic strategy.


Subject(s)
Benzimidazoles/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/radiotherapy , Enzyme Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors , Animals , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Cell Growth Processes/drug effects , Cell Growth Processes/radiation effects , Cell Line, Tumor , Cellular Senescence/drug effects , Cellular Senescence/radiation effects , Combined Modality Therapy , Dose-Response Relationship, Radiation , Female , Green Fluorescent Proteins/analysis , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Infrared Rays , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Nude , Recombinant Fusion Proteins/analysis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Tumor Suppressor p53-Binding Protein 1 , Xenograft Model Antitumor Assays
7.
Int J Radiat Biol ; 85(5): 421-31, 2009 May.
Article in English | MEDLINE | ID: mdl-19437244

ABSTRACT

PURPOSE: To determine the mechanisms of Signal Transducer and Activator of Transcription 1 (Stat1)-associated radioresistance developed by nu61 tumour selected in vivo by fractionated irradiation of the parental radiosensitive tumour SCC61. MATERIALS AND METHODS: Radioresistence of nu61 and SCC61 in vitro was measured by clonogenic assay. Apoptotic response of nu61 and SCC61 cells to genotoxic stress was examined using caspase-based apoptotic assays. Co-cultivation of carboxyfluorescein diacetate, succinimidyl ester (CFDE-SE)-labeled nu61 with un-labeled SCC61 was performed at 1:1 ratio. Production of interleukin-6, interleukin-8 and soluble receptor of interleukin 6 (IL6, IL8 and sIL6R) was measured using Enzyme-Linked Immunosorbent Assay (ELISA). RESULTS: Radioresistant nu61 was also resistant to interferon-gamma (IFNgamma) and the death ligands of tumour necrosis factor alpha receptor (TNFR) family when compared to SCC61. This combined resistance is due to an impaired apoptotic response in nu61. Relative to SCC61, nu61 produced more IL6, IL8 and sIL6R. Using Stat1 knock-downs we demonstrated that IL6 and IL8 production is Stat1-dependent. Treatment with neutralising antibodies to IL6 and IL8, but not to either cytokine alone sensitised nu61 to genotoxic stress induced apoptosis. CONCLUSION: Nu61, which over-expresses Stat1 pathway, is deficient in apoptotic response to ionising radiation and cytotoxic ligands. This resistance to apoptosis is associated with Stat1-dependent production of IL6 and IL8 and suppression of caspases 8, 9 and 3.


Subject(s)
Apoptosis/drug effects , Apoptosis/radiation effects , Interleukins/metabolism , Neoplasms/pathology , Radiation Tolerance , STAT1 Transcription Factor/metabolism , Signal Transduction , Animals , Caspases/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/radiation effects , Coculture Techniques , Cytokines/toxicity , Cytotoxins/toxicity , DNA Damage , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/radiation effects , Humans , Interferon-gamma/pharmacology , Interleukin-6/metabolism , Interleukin-8/metabolism , Neoplasms/genetics , Radiation, Ionizing , Receptors, Tumor Necrosis Factor/metabolism , Signal Transduction/drug effects , Signal Transduction/radiation effects
8.
Tissue Eng Part C Methods ; 15(3): 309-21, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19115821

ABSTRACT

Interactions with the extracellular matrix (ECM) play an important role in regulating cell function. Cells cultured in, or on, three-dimensional ECM recapitulate similar features to those found in vivo that are not present in traditional two-dimensional culture. In addition, both natural and synthetic materials containing ECM components have shown promise in a number of tissue engineering applications. Current materials available for cell culture and tissue engineering do not adequately reflect the diversity of ECM composition between tissues. In this paper, a method is presented for extracting solutions of proteins and glycoproteins from soft tissues and inducing assembly of these proteins into gels. The extracts contain ECM proteins specific to the tissue source with low levels of intracellular molecules. Gels formed from the tissue-derived extracts have nanostructure similar to ECM in vivo and can be used to culture cells as both a thin substrate coating and a thick gel. This technique could be used to assemble hydrogels with varying composition depending upon the tissue source, hydrogels for three-dimensional culture, as scaffolds for tissue engineering therapies, and to study cell-matrix interactions.


Subject(s)
Biocompatible Materials/chemistry , Extracellular Matrix Proteins/chemistry , Extracellular Matrix Proteins/isolation & purification , Gels/chemistry , Tissue Engineering/methods , Tissue Extracts/chemistry , Tissue Extracts/isolation & purification , Animals , Biocompatible Materials/isolation & purification , Cell Culture Techniques/methods , Humans , Materials Testing , Mice , Mice, Nude
9.
Int J Oncol ; 31(6): 1519-28, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17982679

ABSTRACT

TNFalpha was initially described as inducing necrotic death in tumors in vivo, and more recently as a cytokine that mediates cytoprotection and inflammation. The anti-tumor effects of TNFalpha are poorly characterized because TNFalpha-induced death of human tumor cells has largely been studied in the presence of agents that block transcription or protein synthesis. Also, most reports in model cell systems describe apoptosis within relatively early time points as the principal mode of cell death induced by TNFalpha. We investigated the cytotoxic effects of 10 ng/ml TNFalpha on human tumor cells of different histological types without concomitant exposure to these inhibitors. Eleven of 21 human tumor cell lines underwent TNFalpha-induced cell death which ranged from 41% to complete loss of viability. Only one cell line demonstrated caspase-dependent apoptosis within 24 h. Nine cell lines underwent death between 48 h and 21 days. Seven of these lines underwent caspase-3 independent death consistent with necrosis. One tumor line exhibited characteristics of senescence following TNFalpha exposure. Nine of 9 cell lines activated NF-kappaB following TNFalpha exposure by 24 h. In all cell lines studied, with the exception of the epidermoid carcinoma cell line that underwent early apoptosis, expression of one or more NF-kappaB target genes was demonstrated at 24-96 h. BMS-345541, a specific IKK inhibitor, increased TNFalpha killing in TNFalpha resistant tumor cell lines by increasing apoptosis, suggesting that inhibition of NF-kappaB may be an effective strategy to enhance the tumoricidal effects of TNFalpha.


Subject(s)
Apoptosis/drug effects , NF-kappa B/antagonists & inhibitors , Neoplasms/drug therapy , Tumor Necrosis Factor-alpha/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Cellular Senescence/drug effects , Humans , Imidazoles/pharmacology , NF-kappa B/physiology , Necrosis , Neoplasms/pathology , Quinoxalines/pharmacology
10.
Cancer Res ; 67(19): 9214-20, 2007 Oct 01.
Article in English | MEDLINE | ID: mdl-17909027

ABSTRACT

Elsewhere, we reported that multiple serial in vivo passage of a squamous cell carcinoma cells (SCC61) concurrent with ionizing radiation (IR) treatment resulted in the selection of radioresistant tumor (nu61) that overexpresses the signal transducer and activator of transcription 1 (Stat1)/IFN-dependent pathway. Here, we report that (a) the Stat1 pathway is induced by IR, (b) constitutive overexpression of Stat1 is linked with failure to transmit a cytotoxic signal by radiation or IFNs, (c) selection of parental cell line SCC61 against IFN-alpha and IFN-gamma leads to the same IR- and IFN-resistant phenotype as was obtained by IR selection, and (d) suppression of Stat1 by short hairpin RNA renders the IR-resistant nu61 cells radiosensitive to IR. We propose a model that transient induction of Stat1 by IFN, IR, or other stress signals activates cytotoxic genes and cytotoxic response. Constitutive overexpression of Stat1 on the other hand leads to the suppression of the cytotoxic response and induces prosurvival genes that, at high levels of Stat1, render the cells resistant to IR or other inducers of cell death.


Subject(s)
Carcinoma, Squamous Cell/metabolism , STAT1 Transcription Factor/metabolism , Animals , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/radiotherapy , Drug Resistance, Neoplasm , Female , Gene Expression/drug effects , Gene Expression/radiation effects , Humans , Interferon-alpha/pharmacology , Interferon-gamma/pharmacology , Mice , Mice, Nude , Neoplasm Transplantation , RNA, Small Interfering/genetics , Radiation Tolerance , STAT1 Transcription Factor/antagonists & inhibitors , STAT1 Transcription Factor/biosynthesis , STAT1 Transcription Factor/genetics , Transplantation, Heterologous
11.
Technol Cancer Res Treat ; 6(5): 395-401, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17877427

ABSTRACT

Recently, nanoparticles have been considered as a method of providing radiation dose enhancement in tumors. In order to quantify this affect, a dose enhancement factor (DEF) is defined that represents the ratio of the dose deposited in tumor with nanoparticles, divided by the dose deposited in the tumor without nanoparticles. Materials with atomic numbers (Z) ranging from 25 to 90 are considered in this analysis. In addition, the energy spectrum for a number of external beam x-ray sources and common radionuclides are evaluated. For a nanoparticle concentration of 5 mg/ml, the DEF is < 1.05 for Co-60, Ir-192, Au-198, Cs-137, 6, 18, and 25 MV x-rays for all materials considered. However, relatively large increases in the DEF are observed for 50, 80, 100, and 140 KVp x-rays as well as Pd-103 and I-125. The DEF increases for all sources as Z varies from 25-35. From Z = 40-60, the DEF plateaus or slightly decreases. For higher Z materials (Z>70), the DEF increases and is a maximum for the highest Z materials. High atomic number nanoparticles coupled with low energy external beam x-rays or brachytherapy sources offer the potential of significantly enhancing the delivered dose.


Subject(s)
Nanoparticles/therapeutic use , Neoplasms/radiotherapy , Radiation Dosage , Radioisotopes/administration & dosage , Animals , Brachytherapy/methods , Dose-Response Relationship, Radiation , Humans , Models, Theoretical , Radiotherapy Dosage
12.
FEBS Lett ; 565(1-3): 167-70, 2004 May 07.
Article in English | MEDLINE | ID: mdl-15135073

ABSTRACT

We demonstrate that human umbilical vein endothelial cells (HUVEC) grown in co-culture (CC) with U87 glioblastoma cells transfected with green fluorescent protein (GFP-U87) exhibit resistance to radiation-mediated apoptosis. cDNA macroarray analysis reveals increases in the accumulation of RNAs for HUVEC genes encoding cell adhesion molecules, growth factor-related proteins, and cell cycle regulatory/DNA repair proteins. An increase in protein expression of integrin alphav, integrin beta1, MAPK(p42), Rad51, DNA-PK(CS), and ataxia telangiectasia gene (ATM) was detected in HUVEC grown in CC with GFP-U87 cells compared with HUVEC grown in mono-culture. Treatment with anti-VEGF antibody decreases the expression of integrin alphav, integrin beta1, DNA-PK(CS) and ATM with a corresponding increase in ionizing radiation (IR)-induced apoptosis. These data support the concept that endothelial cells growing in the tumor microenvironment may develop resistance to cytotoxic therapies due to the up-regulation by tumor cells of endothelial cells genes associated with survival.


Subject(s)
Apoptosis , Endothelial Cells/pathology , Glioblastoma/pathology , Ataxia Telangiectasia Mutated Proteins , Blotting, Western , Cell Adhesion , Cell Cycle , Cell Cycle Proteins , Cell Division , Cell Line, Tumor , Cells, Cultured , Coculture Techniques , DNA Repair , DNA, Complementary/metabolism , DNA-Activated Protein Kinase , DNA-Binding Proteins/metabolism , Endothelial Cells/radiation effects , Endothelium, Vascular/cytology , Glioblastoma/radiotherapy , Green Fluorescent Proteins , Humans , Infrared Rays , Integrin alphaV/biosynthesis , Integrin alphaVbeta3/metabolism , Integrin beta1/biosynthesis , Luminescent Proteins/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Nuclear Proteins , Oligonucleotide Array Sequence Analysis , Protein Serine-Threonine Kinases/metabolism , Rad51 Recombinase , Radiotherapy , Recombination, Genetic , Transcription, Genetic , Transfection , Tumor Suppressor Proteins , Umbilical Veins/cytology , Up-Regulation , Vascular Endothelial Growth Factor A/metabolism
13.
Proc Natl Acad Sci U S A ; 101(6): 1714-9, 2004 Feb 10.
Article in English | MEDLINE | ID: mdl-14755057

ABSTRACT

Nu61, a radiation-resistant human tumor xenograft, was selected from a parental radiosensitive tumor SCC-61 by eight serial cycles of passage in athymic nude mice and in vivo irradiation. Replicate DNA array experiments identified 52 genes differentially expressed in nu61 tumors compared with SCC-61 tumors. Of these, 19 genes were in the IFN-signaling pathway and moreover, 25 of the 52 genes were inducible by IFN in the nu61 cell line. Among the genes involved in IFN signaling, STAT1alpha and STAT1beta were the most highly overexpressed in nu61 compared to SCC-61. STAT1alpha and STAT1beta cDNAs were cloned and stably transfected into SCC-61 tumor cells. Clones of SCC-61 tumor cells transfected with vectors expressing STAT1alpha and STAT1beta demonstrated radioprotection after exposure to 3 Gy (P < 0.038). The results indicate that radioresistance acquired during radiotherapy treatment may account for some treatment failures and demonstrate an association of acquired tumor radioresistance with up-regulation of components of the IFN-related signaling pathway.


Subject(s)
DNA-Binding Proteins/metabolism , Radiation Tolerance/genetics , Selection, Genetic , Trans-Activators/metabolism , Transduction, Genetic , Animals , Humans , Mice , Mice, Nude , STAT1 Transcription Factor , Tumor Cells, Cultured
14.
Int J Cancer ; 109(2): 214-9, 2004 Mar 20.
Article in English | MEDLINE | ID: mdl-14750172

ABSTRACT

We performed expressional profiling of isogenic glioblastoma cell lines U87-Lux8 and U87-175.4. These cell lines differ in that U87-Lux8 expresses wild-type p53 and U87-175.4 expresses a dominant-negative p53 (175(His) mutation). DNA array analysis and real-time PCR measurements demonstrated that basal expression and response to irradiation were different in these isogenic glioblastoma cell lines. These differences included genes involved in growth regulation and genes associated with cell-to-cell and cell/ECM communications. Co-cultivation of U87-175.4 and U87-Lux8 with HUVE cells demonstrated that U87-175.4 cells suppress the angiogenic phenotype of HUVEC and increase their sensitivity to radiation-induced apoptosis compared to co-culture of U87-Lux8/HUVEC. These data suggest that blockade of p53 function may alter the communication between tumor cells and endothelial cells such that endothelial cells exhibit an increase in radiosensitivity. These findings may have important implications for the treatment of glioblastoma tumors and other human cancers.


Subject(s)
Apoptosis/radiation effects , Brain Neoplasms/pathology , Endothelium, Vascular/pathology , Genes, Dominant , Glioblastoma/pathology , Tumor Suppressor Protein p53/genetics , Brain Neoplasms/genetics , Cell Movement/radiation effects , Coculture Techniques , Enzyme-Linked Immunosorbent Assay , Gene Expression Profiling , Glioblastoma/genetics , Humans , Phenotype , Radiation Tolerance , Transfection , Tumor Cells, Cultured
15.
J Cell Sci ; 116(Pt 6): 1013-22, 2003 Mar 15.
Article in English | MEDLINE | ID: mdl-12584245

ABSTRACT

Tumour angiogenesis is a complex process based upon a sequence of interactions between tumour cells and endothelial cells. To model tumour/endothelial-cell interactions, we co-cultured U87 human glioma cells with human umbilical vein endothelial cells (HUVECs). U87 cells induced an 'activated' phenotype in HUVECs, including an increase in proliferation, migration and net-like formation. Activation was observed in co-cultures where cells were in direct contact and physically separated, suggesting an important role for soluble factor(s) in the phenotypic and genotypic changes observed. Expressional profiling of tumour-activated endothelial cells was evaluated using cDNA arrays and confirmed by quantitative PCR. Matching pairs of receptors/ligands were found to be coordinately expressed, including TGFbetaRII with TGFbeta3, FGFRII and cysteine-rich fibroblast growth factor receptor (CRF-1) with FGF7 and FGF12, CCR1, CCR3, CCR5 with RANTES and calcitronin receptor-like gene (CALCRL) with adrenomedullin. Consistent with cDNA array data, immunohistochemical staining of expressed proteins revealed the upregulation of Tie-2 receptor in vitro and in vivo. Our data suggest that tumour-induced activation of quiescent endothelial cells involves the expression of angiogenesis-related receptors and the induction of autocrine growth loops. We suggest that tumour cells release growth factors that induce endothelial cells to express specific ligands and their cognate receptors coordinately.


Subject(s)
Cell Communication/physiology , Endothelium, Vascular/cytology , Glioma , Neovascularization, Pathologic/pathology , Angiogenesis Inhibitors/pharmacology , Angiostatins , Autocrine Communication/physiology , Cell Movement/physiology , Coculture Techniques , Gene Expression/physiology , Humans , Neovascularization, Pathologic/metabolism , Peptide Fragments/pharmacology , Phenotype , Plasminogen/pharmacology , RNA, Messenger/analysis , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, TIE-2 , Receptors, Cytokine/genetics , Receptors, Fibroblast Growth Factor/genetics , Receptors, Transforming Growth Factor beta/genetics , Transcription, Genetic/physiology , Tumor Cells, Cultured/cytology , Umbilical Veins/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...