Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
J Exp Orthop ; 9(1): 39, 2022 May 02.
Article in English | MEDLINE | ID: mdl-35499653

ABSTRACT

PURPOSE: Molecular processes in primary osteoblasts were analyzed in response to magnetic and electric field exposure to examine its potential impact on bone healing. METHODS: Primary osteoblasts were exposed to a combination of a magnetic field and an additional electric field (EFMF) (20 Hz, 700 mV, 5 mT, continuous sinusoids) in vitro. mRNA- and protein-expressions were assessed during a time interval of 21 days and compared with expression data obtained from control osteoblasts. RESULTS: We observed an autonomous osteoblast differentiation process in vitro under the chosen cultivation conditions. The initial proliferative phase was characterized by a constitutively high mRNA expression of extracellular matrix proteins. Concurrent EFMF exposure resulted in significanly increased cell proliferation (fold change: 1.25) and reduced mRNA-expressions of matrix components (0.5-0.75). The following reorganization of the extracellular matrix is prerequisite for matrix mineralization and is characterised by increased Ca2+ deposition (1.44). On molecular level EFMF exposure led to a significant decreased thrombospondin 1 (THBS1) mRNA- (0.81) and protein- (0.54) expression, which in turn reduced the TGFß1-dependent mRNA- (0.68) and protein- (0.5) expression of transforming growth factor beta induced (ßIG-H3) significantly, an inhibitor of endochondral ossification. Consequently, EFMF exposure stimulated the expression of genes characteristic for endochondral ossification, such as collagen type 10, A1 (1.50), osteopontin (1.50) and acellular communication network factor 3 (NOV) (1.45). CONCLUSIONS: In vitro exposure of osteoblasts to EFMF supports cell differentiation and induces gene- and protein-expression patterns characteristic for endochondral ossification during bone fracture healing in vivo.

2.
Theranostics ; 11(16): 7700-7714, 2021.
Article in English | MEDLINE | ID: mdl-34335959

ABSTRACT

CD4+ T helper cells are capable of mediating long-term antitumoral immune responses. We developed a combined immunotherapy (COMBO) using tumor antigen-specific T helper 1 cells (Tag-Th1), dual PD-L1/LAG-3 immune checkpoint blockade, and a low-dose total body irradiation (TBI) of 2 Gy, that was highly efficient in controlling the tumor burden of non-immunogenic RIP1-Tag2 mice with late-stage endogenous pancreatic islet carcinomas. In this study, we aimed to explore the impact of 2 Gy TBI on the treatment efficacy and the underlying mechanisms to boost CD4+ T cell-based immunotherapies. Methods: Heavily progressed RIP1-Tag2 mice underwent COMBO treatment and their survival was compared to a cohort without 2 Gy TBI. Positron emission tomography/computed tomography (PET/CT) with radiolabeled anti-CD3 monoclonal antibodies and flow cytometry were applied to investigate 2 Gy TBI-induced alterations in the biodistribution of endogenous T cells of healthy C3H mice. Migration and homing properties of Cy5-labeled adoptive Tag-Th1 cells were monitored by optical imaging and flow cytometric analyses in C3H and tumor-bearing RIP1-Tag2 mice. Splenectomy or sham-surgery of late-stage RIP1-Tag2 mice was performed before onset of COMBO treatment to elucidate the impact of the spleen on the therapy response. Results: First, we determined a significant longer survival of RIP1-Tag2 mice and an increased CD4+ T cell tumor infiltrate when 2 Gy TBI was applied in addition to Tag-Th1 cell PD-L1/LAG-3 treatment. In non-tumor-bearing C3H mice, TBI induced a moderate host lymphodepletion and a tumor antigen-independent accumulation of Tag-Th1 cells in lymphoid and non-lymphoid organs. In RIP1-Tag2, we found increased numbers of effector memory-like Tag-Th1 and endogenous CD4+ T cells in the pancreatic tumor tissue after TBI, accompanied by a tumor-specific Th1-driven immune response. Furthermore, the spleen negatively regulated T cell effector function by upregulation PD-1/LAG-3/TIM-3 immune checkpoints, providing a further rationale for this combined treatment approach. Conclusion: Low-dose TBI represents a powerful tool to foster CD4+ T cell-based cancer immunotherapies by favoring Th1-driven antitumoral immunity. As TBI is a clinically approved and well-established technique it might be an ideal addition for adoptive cell therapy with CD4+ T cells in the clinical setting.


Subject(s)
Immunotherapy/methods , Th1 Cells/metabolism , Whole-Body Irradiation/methods , Animals , Antibodies, Monoclonal/pharmacology , Antigens, CD/immunology , Antigens, CD/metabolism , Antigens, Neoplasm , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , B7-H1 Antigen/metabolism , Female , Immunity/drug effects , Immunotherapy, Adoptive/methods , Mice , Mice, Inbred C3H , Optical Imaging , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , Positron Emission Tomography Computed Tomography , Th1 Cells/immunology , Tissue Distribution , Lymphocyte Activation Gene 3 Protein
3.
Int J Mol Sci ; 20(24)2019 Dec 14.
Article in English | MEDLINE | ID: mdl-31847370

ABSTRACT

Homologous recombination repair (HRR), non-homologous end-joining (NHEJ) and alternative NHEJ are major pathways that are utilized by cells for processing DNA double strand breaks (DNA-DSBs); their function plays an important role in the radiation resistance of tumor cells. Conflicting data exist regarding the role of Akt in homologous recombination (HR), i.e., the regulation of Rad51 as a major protein of this pathway. This study was designed to investigate the specific involvement of Akt isoforms in HRR. HCT116 colon cancer cells with stable AKT-knock-out and siRNA-mediated AKT-knockdown phenotypes were used to investigate the role of Akt1 and Akt2 isoforms in HR. The results clearly demonstrated that HCT116 AKT1-KO and AKT2-KO cells have a significantly reduced Rad51 foci formation 6 h post irradiation versus parental cells. Depletion of Akt1 and Akt2 protein levels as well as inhibition of Akt kinase activity resulted in an increased number of residual-γH2AX in CENP-F positive cells mainly representing the S and G2 phase cells. Furthermore, inhibition of NHEJ and HR using DNA-PK and Rad51 antagonists resulted in stronger radiosensitivity of AKT1 and AKT2 knockout cells versus wild type cells. These data collectively show that both Akt1 and Akt2 are involved in DSBs repair through HRR.


Subject(s)
DNA Repair/genetics , DNA/genetics , Homologous Recombination/genetics , Proto-Oncogene Proteins c-akt/genetics , A549 Cells , Cell Line, Tumor , DNA Breaks, Double-Stranded , DNA-Binding Proteins/genetics , G2 Phase/genetics , HCT116 Cells , Humans , Rad51 Recombinase/genetics , Radiation Tolerance/genetics , Recombinational DNA Repair/genetics , S Phase/genetics
4.
Exp Cell Res ; 383(1): 111498, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31302031

ABSTRACT

Radiation-induced bystander effects (RIBE) are discussed as relevant processes during radiotherapy. Irradiated cells are suggested to release growth-inhibitory/DNA-damaging factors transported to non-irradiated cells. However, the molecular nature of this phenomenon has not yet been resolved. We aimed at identifying the growth-inhibitory factor(s) transmitted to non-irradiated cells. RIBE-competent PC3 cells were used to produce conditioned medium (CM) after exposure to ionizing radiation. Indicator cells were incubated with CM and clonogenic survival as well as cell proliferation were determined as endpoints. A549 indicator cells exhibited a bystander effect upon incubation with CM from irradiated PC3 cells. This bystander effect was not due to DNA-damaging factors, but a radiation-triggered reduction of mitogenic/clonogenic activity present in CM. Several tumor cells, but not normal fibroblasts secrete this factor, whose release is reduced by irradiation. We identified L-Plastin to be responsible for the mitogenic/clonogenic activity. Removal of L-Plastin from CM by immunoprecipitation or siRNA-mediated knockdown of L-Plastin expression resulted in loss or reduction of mitogenic/clonogenic activity transmitted via CM, respectively. Exosome-transported L-Plastin was constitutively Ser5-phosphorylated, indicative of its bioactive conformation. In summary, we observed production and exosomal secretion of L-Plastin by cancer cells. Via exosome-transmitted L-Plastin, tumors induce clonogenic and mitogenic activity in cancer and normal cells of the tumor microenvironment. Irradiation inhibits L-Plastin production targeting both cancer cells and the tumor niche and may explain the high impact of radiotherapy in tumor control.


Subject(s)
Bystander Effect/radiation effects , Cell Proliferation/radiation effects , Exosomes/metabolism , Lung Neoplasms/pathology , Microfilament Proteins/metabolism , Prostatic Neoplasms/pathology , Radiation, Ionizing , Bystander Effect/drug effects , Cell Proliferation/drug effects , Cell Survival , Cells, Cultured , Culture Media, Conditioned/pharmacology , Culture Media, Conditioned/radiation effects , Exosomes/radiation effects , Fibroblasts/radiation effects , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/radiotherapy , Male , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/radiotherapy
5.
Radiother Oncol ; 126(3): 465-470, 2018 03.
Article in English | MEDLINE | ID: mdl-29100699

ABSTRACT

BACKGROUND AND PURPOSE: Tumour cells are characterized by aerobic glycolysis and thus have high glucose consumption. Because repairing radiation-induced DNA damage is an energy-demanding process, we hypothesized that glucose starvation combined with radiotherapy could be an effective strategy to selectively target tumour cells. MATERIAL AND METHODS: We glucose-starved tumour cells (A549, FaDu) in vitro and analysed their radiation-induced cell responses compared to normal fibroblasts (HSF7). RESULTS: Irradiation depleted intracellular ATP levels preferentially in cancer cells. Consequently, glucose starvation impaired DNA double-strand break (DSB) repair and radiosensitized confluent tumour cells but not normal fibroblasts. In proliferating tumour cells glucose starvation resulted in a reduction of proliferation, but failed to radiosensitize cells. Glucose supply was indispensable during the late DSB repair in confluent tumour cells starting approximately 13 h after irradiation, and glucose starvation inhibited radiation-induced histone acetylation, which is essential for chromatin relaxation. Sirtinol - an inhibitor of histone deacetylases - reverted the effects of glucose depletion on histone acetylation and DNA DSB repair in tumour cells. Furthermore, a glucose concentration of 2.8 mmol/L was sufficient to impair DSB repair in tumour cells and reduced their clonogenic survival under a fractionated irradiation regimen. CONCLUSIONS: In resting tumour cells, glucose starvation combined with irradiation resulted in the impairment of late DSB repair and the reduction of clonogenic survival, which was associated with disrupted radiation-induced histone acetylation. However, in normal cells, DNA repair and radiosensitivity were not affected by glucose depletion.


Subject(s)
Carcinoma, Non-Small-Cell Lung/therapy , Carcinoma, Squamous Cell/therapy , DNA Repair , Glucose/deficiency , Head and Neck Neoplasms/therapy , Histones/metabolism , Lung Neoplasms/therapy , A549 Cells , Acetylation , Adenosine Triphosphate/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/radiotherapy , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/radiotherapy , DNA Breaks, Double-Stranded , DNA, Neoplasm/radiation effects , Fibroblasts/radiation effects , Glucose/administration & dosage , Glucose/metabolism , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/radiotherapy , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/radiotherapy , Radiation Tolerance , Squamous Cell Carcinoma of Head and Neck
6.
PLoS One ; 12(12): e0189087, 2017.
Article in English | MEDLINE | ID: mdl-29253018

ABSTRACT

Cell membrane-associated epidermal growth factor receptor (EGFR) translocates into a perinuclear/nuclear location upon stimulation, where it complexes with mRNAs. Treatment with radiation and cisplatin decreases the amounts of mRNAs present within this complex. Gene array analyses of mRNAs in complex with immunoprecipitated nEGFR revealed significant enrichment of different mRNA species compared to the control immunoprecipitation. Functional annotation with help of DAVID Gene Ontology Analysis identified under other terms the HIF-1A/VEGF signaling pathway as one of the top scoring KEGG pathways. RT-PCR and western blots revealed the radiation-induced expression of mRNAs and proteins involved in HIF-1A/VEGF signaling. Simultaneously, the levels of the corresponding validated miRNAs within the complex containing nEGFR and mRNAs were decreased. This finding argues that an mRNA/miRNA/nEGFR complex regulates protein expression. Indeed, we detected the GW182, AGO2, PABPC1 and cNOT1 proteins, which belong to the deadenylase complex, in a complex with nuclear EGFR. Erlotinib-mediated inhibition of EGFR kinase reduced the radiation-induced increase in mRNA expression. In this context, erlotinib reduced AGO2 phosphorylation by the EGFR kinase at residue Y393, which was associated with increased cNOT1 deadenylase activity and reduced mRNA stability. To prove the roles of miRNAs in this context, we transfected cells with an inhibitor of Hsa-mir-1180p5, which targets the NFATC4 mRNA, an mRNA associated with VEGF signaling, or pretreated cells with erlotinib. Indeed, Hsa-mir-1180p5 knockdown increased and the erlotinib treatment decreased the expression of the NFATC4 protein. The expression of the NFATC4 protein controlled the cloning efficiency and radiosensitivity of A549 and FaDu tumor cells. Thus, this study is the first to show that a membrane-located tyrosine kinase receptor, such as EGFR, is internalized to a nuclear/perinuclear location upon exposure to stress and modulates the stability and translation of miRNA-selected mRNAs. This mechanism enables cells to directly express proteins in response to EGFR activation and may contribute to treatment resistance in EGFR-overexpressing tumors.


Subject(s)
Cell Nucleus/metabolism , ErbB Receptors/metabolism , Protein Biosynthesis , RNA Stability , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism , A549 Cells , Adenosine Triphosphate/metabolism , Cell Survival , Clone Cells , DNA, Complementary/genetics , Humans , NFATC Transcription Factors/metabolism , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Induced Silencing Complex/metabolism , Transcription Factors/metabolism
7.
Radiother Oncol ; 124(3): 462-467, 2017 09.
Article in English | MEDLINE | ID: mdl-28711224

ABSTRACT

BACKGROUND: Radiotherapy (RT) is used to treat retinoblastoma (Rb), the most frequent ocular tumour in children. Besides eradicating the tumour, RT can cause severe side effects including secondary malignancies. This study aimed to define whether the radioprotector ortho-phospho-L-tyrosine (pTyr) prevents RT-induced side effects and affects local tumour control in a xenograft and a genetic orthotopic Rb mouse model. METHODS: B6;129-Rb1tm3Tyj/J (Rb+/-) and Y79-Rb cell-xenografted nude mice were fractionated external beam irradiated (15 fractions of 5Gy 6MV photons during 3weeks) with or without pTyr pre-treatment (100mg/kg BW, 16h prior to each irradiation). One, three, six and nine months after RT, tumour control and RT toxicity were evaluated using in vivo imaging and histology. We also analysed pTyr dependant post irradiation cell survival and p53 activity in vitro. RESULTS: In vitro pTyr pre-treatment showed no radioprotection on Y79 cells, but led to p53 stabilisation in unirradiated Y79 cells and to a facilitation of radiation-induced p21 up-regulation, confirming a modulation of p53 activity by pTyr. In both mouse models, secondary tumours were undetectable. In Rb+/- mice, pTyr significantly lowered RT-induced greying of the fur, retinal thickness reduction and photoreceptor loss. However, in the xenografted Rb model, pTyr considerably decreased RT-mediated tumour control, which was observed in 16 out of 22 control eyes but in none of the 24 pTyr treated eyes. CONCLUSIONS: In Rb+/- mice pTyr significantly prevents RT-induced greying of the fur as well as retinal degeneration. However, since non-irradiated control mice were not used in our study, a formal possibility exists that the effect shown in the retina of Rb+/- mice may be due to ageing of the animals and/or actions of pTyr alone. Unfortunately, as tested in a xenograft model, pTyr treatment reduced the control of Rb tumours.


Subject(s)
Dose Fractionation, Radiation , Phosphotyrosine/pharmacology , Radiation-Protective Agents/pharmacology , Retinal Neoplasms/radiotherapy , Retinoblastoma/radiotherapy , Animals , Cell Survival , Disease Models, Animal , Mice , Retinoblastoma/pathology , Tumor Suppressor Protein p53/physiology
8.
Radiother Oncol ; 116(3): 431-7, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26320552

ABSTRACT

BACKGROUND AND PURPOSE: EGFR is translocated into the cell nucleus in response to irradiation, where it is involved in regulation of radio-sensitivity. The aim of this study is to elucidate the functional role of nuclear EGFR. MATERIAL AND METHODS: To identify EGFR-bound nuclear proteins and mRNAs, Maldi-TOF analysis and mRNA gene arrays were used. Complex formation of proteins was shown by confocal microscopy, immunoprecipitation and Western blotting. The effect of EGFR binding to mRNAs was exhibited by quantitative RT-PCR. Cellular endpoints were shown by Western blotting, mitochondrial mass quantification, lactate quantification and clonogenic survival assays. RESULTS: Maldi-TOF analysis of proteins bound to nuclear EGFR in response to irradiation showed colocalization with Lamin A and heterogeneous nuclear ribonucleoproteins. Confocal microscopy and Western blotting confirmed this colocalization. Both Lamin A and heterogeneous nuclear ribonucleoproteins are involved in mRNA processing. To support a role of nEGFR in this context after irradiation, we isolated EGFR-bound mRNA and observed an EGFR kinase-dependent mRNA stabilizing effect. With the help of DNA microarrays, we identified mRNAs associated with the Warburg effect that were bound to nuclear EGFR. In this context, we observed radiation-induced HIF1α expression, which triggers inhibition of pyruvate dehydrogenase and blocks the tricarboxylic acid cycle. Consequently, we detected mitophagy and increased lactate production, which is associated with increased treatment resistance. Reduction of nEGFR decreased radiation-induced expression of Hif1α and lactate production. CONCLUSIONS: We showed that nuclear EGFR selectively binds and stabilizes mRNA involved in the Warburg effect in response to irradiation. As a consequence, cells switch from aerobic to anaerobic glucose metabolism, which can be prevented by HIF1α inhibitor BAY87-2243, Dasatinib, Erlotinib or EGFR siRNA.


Subject(s)
ErbB Receptors/physiology , Lactic Acid/biosynthesis , RNA, Messenger/metabolism , Blotting, Western , Bronchial Neoplasms/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Dasatinib/pharmacology , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/radiation effects , Erlotinib Hydrochloride/pharmacology , Gene Expression , Head and Neck Neoplasms/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Nuclear Proteins/metabolism , Oxadiazoles/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Radiation Tolerance/drug effects
9.
Mol Cancer Res ; 13(9): 1283-95, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26041939

ABSTRACT

UNLABELLED: Ca(2+)-activated K(+) channels, such as BK and IK channels, have been proposed to fulfill pivotal functions in neoplastic transformation, malignant progression, and brain infiltration of glioblastoma cells. Here, the ionizing radiation (IR) effect of IK K(+) channel targeting was tested in human glioblastoma cells. IK channels were inhibited pharmacologically by TRAM-34 or genetically by knockdown, cells were irradiated with 6 MV photons and IK channel activity, Ca(2+) signaling, cell cycling, residual double-strand breaks, and clonogenic survival were determined. In addition, the radiosensitizing effect of TRAM-34 was analyzed in vivo in ectopic tumors. Moreover, The Cancer Genome Atlas (TCGA) was queried to expose the dependence of IK mRNA abundance on overall survival (OS) of patients with glioma. Results indicate that radiation increased the activity of IK channels, modified Ca(2+) signaling, and induced a G2-M cell-cycle arrest. TRAM-34 decreased the IR-induced accumulation in G2-M arrest and increased the number of γH2AX foci post-IR, suggesting that TRAM-34 mediated an increase of residual DNA double-strand breaks. Mechanistically, IK knockdown abolished the TRAM-34 effects indicating the IK specificity of TRAM-34. Finally, TRAM-34 radiosensitized ectopic glioblastoma in vivo and high IK mRNA abundance associated with shorter patient OS in low-grade glioma and glioblastoma. IMPLICATIONS: Together, these data support a cell-cycle regulatory function for IK K(+) channels, and combined therapy using IK channel targeting and radiation is a new strategy for anti-glioblastoma therapy.


Subject(s)
Calcium/metabolism , Dacarbazine/analogs & derivatives , Glioblastoma/drug therapy , Glioblastoma/radiotherapy , Intermediate-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Potassium Channel Blockers/pharmacology , Radiation-Sensitizing Agents/pharmacology , Animals , Cell Line, Tumor , Combined Modality Therapy , Dacarbazine/pharmacology , Disease-Free Survival , Female , G2 Phase Cell Cycle Checkpoints/drug effects , Gene Knockdown Techniques , Glioblastoma/metabolism , Glioblastoma/pathology , Glioma/metabolism , Histones/metabolism , Humans , Intermediate-Conductance Calcium-Activated Potassium Channels/genetics , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Mice , Pyrazoles/pharmacology , RNA, Messenger/metabolism , Signal Transduction/drug effects , Temozolomide
10.
Radiother Oncol ; 107(2): 247-51, 2013 May.
Article in English | MEDLINE | ID: mdl-23602371

ABSTRACT

BACKGROUND AND PURPOSE: EGFR and the sodium-dependent glucose transporter, SGLT1, are found in complex after radiation treatment. The aim of this study was to elucidate the role of EGFR in glucose uptake and chromatin remodeling. MATERIAL AND METHODS: Glucose accumulation was quantified with help of (3)H-glucose. Involvement of SGLT was detected by a specific inhibitor. Role of EGFR was proved by EGFR overexpression and siRNA driven knockdown. Functional endpoints were intracellular ATP levels, protein expression, residual DNA-damage and colony formation. RESULTS: EGFR/SGLT1 interactions in response to ionizing radiation were associated with increased glucose uptake. Nevertheless, tumor cells exhibit ATP depletion following irradiation. Recovery from radiation-induced ATP crisis was EGFR/SGLT-dependent and associated with increased cell survival and improved DNA-repair. The blockage of either EGFR or SGLT inhibited ATP level recovery and histone H3 modifications crucial for both chromatin remodeling and DNA repair in response to irradiation. Inhibition of the acetyltransferase TIP60, which is essential for histone H3-K9 acetylation and ATM activation, prevented energy crisis and chromatin remodeling. CONCLUSIONS: Radiation-associated interactions between SGLT1 and EGFR resulted in increased glucose uptake, which counteracts the ATP crisis in tumor cells due to chromatin remodeling. The blockage of recovery from ATP crisis led to radio-sensitization in tumor cells.


Subject(s)
Chromatin Assembly and Disassembly/radiation effects , ErbB Receptors/physiology , Sodium-Glucose Transporter 1/physiology , Adenosine Triphosphate/analysis , Animals , CHO Cells , Cell Line, Tumor , Cricetinae , Cricetulus , Glucose/metabolism , Humans , Phlorhizin/pharmacology
11.
Radiother Oncol ; 103(3): 373-9, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22516777

ABSTRACT

BACKGROUND AND PURPOSE: Solid tumor cells may adapt to an ischemic microenvironment by upregulation of sodium/glucose cotransport (SGLT) in the plasma membrane which supplies the tumor cell with glucose even at very low extracellular glucose concentration. Since SGLT activity has been shown to depend on the epithelial growth factor receptor (EGFR) and EGFR reportedly is activated by ionizing radiation, we tested for irradiation-induced SGLT activity. MATERIALS AND METHODS: A549 lung adenocarcinoma and FaDu head and neck squamous cancer cells were irradiated with 0 and 4 Gy X-ray and electrogenic SGLT transport activity was recorded by patch clamp current clamp in the presence and absence of extracellular glucose (5mM), the SGLT inhibitor phlorizin (500 µM), and the inhibitor of the EGFR tyrosine kinase activity erlotinib (1 µM). In addition, the effect of phlorizin and erlotinib on glucose uptake and clonogenic survival was tested in irradiated and control cells by tracer flux and colony formation assays, respectively. RESULTS: Irradiated A549 cells exhibited a significantly lower membrane potential 3h after irradiation than the control cells. Phlorizin, erlotinib or removal of extracellular glucose, hyperpolarized the irradiated A549 cells to a significantly higher extent than the control cells. Similarly, but less pronounced, glucose removal hyperpolarized irradiated FaDu cells. In addition, irradiated A549 cells exhibited a highly increased (3)H-glucose uptake which was sensitive to phlorizin. Finally, phlorizin radiosensitized the A549 and FaDu cells as evident from the colony formation assays. CONCLUSIONS: Taken together, these data suggest an irradiation-stimulated and EGFR-mediated increase in SGLT-generated glucose uptake which is required for the survival of the genotoxically stressed tumor cells.


Subject(s)
Adenocarcinoma/physiopathology , ErbB Receptors/pharmacology , Lung Neoplasms/physiopathology , Sodium-Glucose Transport Proteins/metabolism , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adenocarcinoma/radiotherapy , Adenocarcinoma of Lung , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/physiopathology , Cell Line, Tumor/radiation effects , Cell Survival/drug effects , Cell Survival/radiation effects , Erlotinib Hydrochloride , Glucose/metabolism , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Head and Neck Neoplasms/physiopathology , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/radiotherapy , Membrane Potentials/drug effects , Patch-Clamp Techniques , Phlorhizin/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Quinazolines/pharmacology , Radiation Dosage , Sodium-Glucose Transport Proteins/antagonists & inhibitors , Sodium-Glucose Transport Proteins/drug effects , Sodium-Glucose Transport Proteins/radiation effects , Tumor Cells, Cultured/radiation effects
12.
Radiother Oncol ; 101(1): 122-6, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21704404

ABSTRACT

BACKGROUND AND PURPOSE: Glioblastoma cells express high levels of Ca(2+)-activated BK K(+) channels which have been proposed to be indispensable for glioblastoma proliferation and migration. Since migration of glioblastoma cells is reportedly stimulated by ionizing radiation (IR), we tested for an IR-induced increase in BK channel activity and its effect on cell migration. MATERIALS AND METHODS: T98G and U87MG cells were X-ray-irradiated with 0-2 Gy, BK channel activity was assessed by patch-clamp recording, migration by trans-well migration assay, and activation of the Ca(2+)/calmodulin-dependent kinase II (CaMKII) by immunoblotting. RESULTS: IR dose-dependently stimulated migration of glioblastoma cells which was sensitive to the BK channel inhibitor paxilline. Ca(2+)-permeabilization of T98G cells activated up to 350 BK channels per cells. Importantly, IR stimulated an increase in BK channel open probability but did not modify the total number of channels. Moreover, IR activated CaMKII in a paxilline-sensitive manner. Finally, inhibition of CaMKII by KN-93 abolished the IR-stimulated migration. CONCLUSIONS: We conclude that IR stimulates BK channel activity which results in activation of CaMKII leading to enhanced glioblastoma cell migration.


Subject(s)
Brain Neoplasms/radiotherapy , Cell Movement/radiation effects , Glioblastoma/radiotherapy , Large-Conductance Calcium-Activated Potassium Channels/radiation effects , Potassium Channels, Calcium-Activated/radiation effects , Benzylamines/pharmacology , Brain Neoplasms/pathology , Cell Movement/drug effects , Glioblastoma/pathology , Humans , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Patch-Clamp Techniques , Potassium Channels, Calcium-Activated/metabolism , Radiation, Ionizing , Signal Transduction , Sulfonamides/pharmacology , Tumor Cells, Cultured
13.
Radiother Oncol ; 99(3): 317-22, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21704408

ABSTRACT

PURPOSE: Nuclear EGFR is involved in cellular stress management and regulation of cellular radio-sensitivity. The aim of this study was to elucidate the molecular mode of nuclear EGFR action. METHODS: Radiation induced nuclear EGFR-shuttling and EGFR-foci formation was analyzed with immunohistochemistry and confocal microscopy. Composition of γH(2)AX-protein complexes was analyzed by western-blotting after immuno-precipitation. Functional relevance of nuclear EGFR was analyzed after siRNA mediated depletion of EGFR with respect to activation of ATM, histone H3 acetylation, residual DNA-damage and cell survival after irradiation. RESULTS: Following radiation nuclear EGFR was localized in foci similar to γH(2)AX. EGFR co-localized in a sub-fraction of γH(2)AX-foci. Analysis of composition of γH(2)AX-complexes revealed presence of EGFR, ATM, promyelocytic leukemia protein (PML), histone H3 and hetero-chromatin binding protein (HP1) in response to radiation. Depletion of EGFR protein inhibited ATM activation due to inhibition of acetylase TIP60 activity following irradiation. Consequently, histone H3 acetylation and phosphorylation was blocked and chromatin could not be opened for repair. Thus, residual DNA-damage was increased 24 h after irradiation and cells were radio-sensitized. Comparable results were obtained when cells were treated with EGFR-NLS-peptide, which blocks EGFR nuclear shuttling specifically. CONCLUSIONS: Nuclear EGFR is part of DNA-damage repair complex and is involved in regulation of TIP60-acetylase activity. TIP60 is essential for ATM activation and chromatin relaxation which is a prerequisite for DNA-repair in heterochromatic DNA. Thus interventional EGFR strategies during tumor treatment may also interact with DNA-repair by blocking access to damaged DNA.


Subject(s)
Bronchial Neoplasms/metabolism , Chromatin/metabolism , ErbB Receptors/pharmacology , Blotting, Western , Cell Line, Tumor , Cell Nucleus/radiation effects , Cell Survival/radiation effects , DNA Damage/radiation effects , Histones/metabolism , Humans , Immunohistochemistry , Immunoprecipitation , Microscopy, Confocal , Radiation Tolerance , Up-Regulation/radiation effects
14.
FEBS Lett ; 584(18): 3878-84, 2010 Sep 24.
Article in English | MEDLINE | ID: mdl-20692258

ABSTRACT

Nuclear localisation of EGFR is associated with treatment resistance of tumor cells. The aim of this study was to identify molecular targets to block nuclear shuttling of EGFR. Mutation of Thr654, located within the putative EGFR NLS demonstrated that phosphorylation of this residue is essential for nuclear EGFR shuttling following irradiation. Deletion of Thr654 blocked nuclear transport of EGFR, whereas mutation to Glu increased shuttling. Treatment with a peptide, corresponding to the phosphorylated NLS, abolished nuclear EGFR transport and reduced radiation-induced activation of DNA-PK, essential for DNA-repair. In accordance with that, lack of nuclear EGFR increased residual DNA damage in tumor cells and reduced cellular survival following irradiation. Blockage of nuclear EGFR shuttling may be a new strategy to fight treatment resistance.


Subject(s)
Cell Nucleus/enzymology , ErbB Receptors/metabolism , Active Transport, Cell Nucleus/genetics , Active Transport, Cell Nucleus/radiation effects , Cell Line, Tumor , Cytosol/enzymology , ErbB Receptors/genetics , Humans , Nuclear Localization Signals/genetics , Nuclear Localization Signals/metabolism , Phosphorylation , Radiation, Ionizing , Threonine/metabolism
15.
Int J Radiat Oncol Biol Phys ; 77(1): 226-34, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20394854

ABSTRACT

PURPOSE: Increased expression of survivin has been identified as a negative prognostic marker in a variety of human cancers. We have previously shown that survivin is a radiation-resistance factor and that the therapeutic effect of survivin knock-down might result from an impaired DNA repair capacity. In this study, we aimed to elucidate an interrelationship between survivin's cellular localization and DNA double-strand break repair. METHODS AND MATERIALS: Survivin's cellular distribution and nuclear complex formation were assayed by Western blotting of subcellular fractions, by immunofluorescence staining, and co-immunoprecipitation in SW480 colorectal cancer cells. DNA repair capacity was analyzed by kinetics of gamma-H2AX foci formation, and by DNA-dependent protein kinase (DNA-PKcs) assays in the presence of survivin-specific or nonspecific control siRNA. RESULTS: Following irradiation, we observed a rapid nuclear accumulation of survivin and subsequent phosphorylation of the protein in the nucleus. Co-immunoprecipitation analyses from nuclear extracts revealed an interaction among survivin, Ku70, gamma-H2AX, MDC1, and DNA-PKcs that was confirmed by immunofluorescence co-localization in nuclear foci. Survivin knock down by siRNA resulted in an impaired DNA double strand break repair, as demonstrated by an increased detection of gamma-H2AX foci/nucleus at 60 min and a higher amount of residual gamma-H2AX foci at 24 hr postirradiation. Furthermore, we detected in survivin-depleted cells a hampered S2056 autophosphorylation of DNA-PKcs and a significantly decreased DNA-PKcs kinase activity. CONCLUSION: These data indicate that nuclear survivin is linked to DNA double-strand break repair by interaction with members of the DNA double-strand breaks repair machinery, thus regulating DNA-PKcs activity.


Subject(s)
Cell Nucleus/metabolism , DNA Repair/physiology , Microtubule-Associated Proteins/metabolism , Adaptor Proteins, Signal Transducing , Cell Cycle Proteins , Cytoplasm/metabolism , DNA Breaks, Double-Stranded , DNA Damage , DNA Repair/radiation effects , DNA-Activated Protein Kinase/metabolism , Histones/metabolism , Humans , Inhibitor of Apoptosis Proteins , Microtubule-Associated Proteins/genetics , Nuclear Proteins/metabolism , Phosphorylation , RNA, Small Interfering , Survivin , Trans-Activators/metabolism , Tumor Cells, Cultured/radiation effects
16.
Strahlenther Onkol ; 186(1): 1-6, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20082181

ABSTRACT

Emerging evidence suggests the existence of a new mode of epidermal growth factor receptor (EGFR) signaling in which activated EGFR undergoes nuclear translocation following treatment with ionizing radiation. The authors provide evidence that the nuclear EGFR transport is a stress-specific cellular reaction, which is linked to src-dependent EGFR internalization into caveolae. These flask-shaped pits can fuse with endoplasmic reticulum and the EGFR is sorted into a perinuclear localization. This compartment may serve as a reservoir for nuclear EGFR transport which is regulated by PKCepsilon (protein kinase Cepsilon). Nuclear EGFR is able to induce transcription of genes essential for cell proliferation and cell-cycle regulation. Moreover, nuclear EGFR has physical contact with compounds of the DNA repair machinery and is involved in removal of DNA damage. Anti-EGFR strategies target radiation-associated EGFR nuclear translocation in different manners. EGFR-inhibitory antibodies, i.e., cetuximab (Erbitux((R))), can block nuclear translocation by EGFR immobilization within the cytosol in responder cell lines, whereas tyrosine kinase inhibitors rather target nuclear kinase activity of EGFR linked with cytosolic or nuclear functions. However, both strategies can inhibit DNA repair following irradiation.


Subject(s)
Cell Nucleus/radiation effects , Cell Survival/radiation effects , ErbB Receptors/radiation effects , Signal Transduction/radiation effects , Translocation, Genetic/radiation effects , Tumor Cells, Cultured/radiation effects , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/pharmacology , Caveolae/radiation effects , Cell Cycle/genetics , Cell Cycle/radiation effects , Cell Division/genetics , Cell Division/radiation effects , Cell Line , Cell Nucleus/drug effects , Cell Nucleus/genetics , Cell Survival/drug effects , Cell Survival/genetics , Cetuximab , DNA Damage/genetics , DNA Damage/radiation effects , DNA Repair/drug effects , DNA Repair/genetics , DNA Repair/radiation effects , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/radiation effects , Genes, src/radiation effects , Humans , Protein Kinase C-epsilon/physiology , Protein-Tyrosine Kinases/antagonists & inhibitors , Signal Transduction/drug effects , Signal Transduction/genetics , Transcription, Genetic/genetics , Transcription, Genetic/radiation effects , Translocation, Genetic/drug effects , Tumor Cells, Cultured/drug effects
17.
Radiother Oncol ; 92(3): 379-82, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19560222

ABSTRACT

PURPOSE: Elucidation of the molecular mechanism of radiation-induced activation of src kinase, which initiates EGFR internalization and nuclear transport. MATERIAL AND METHODS: Radiation-induced src activation was investigated in the bronchial carcinoma cell line A549. Proteins were Western blotted and quantified by the help of specific antibodies. Residual DNA-damage was quantified with gammaH(2)AX-foci analysis. Radiation-induced lipid peroxidation was prevented by acetyl-cysteine. RESULTS: The radiation-induced src activation and EGFR stabilization could be mimicked by addition of hydroxy-nonenal (HNE), one of the major lipid peroxidation products. Radiation-generated HNE is bound to EGFR and src and correlated with complex formation between both following radiation. Treatment with HNE activated src and stimulated radiation-associated EGFR and caveolin 1 phosphorylations resulting in increased nuclear transport of EGFR. Consequently, radiation-induced phosphorylation and activation of DNA-PK were increased. This phosphorylation was associated with improved removal of residual damage 24h after irradiation. Inhibition of radiation-induced HNE generation by acetyl-cysteine blocked radiation-induced src activation and EGFR phosphorylation. CONCLUSIONS: HNE generated in response to radiation exposure activates src kinase and is involved in regulation of radiation-stimulated DNA-repair processes.


Subject(s)
Caveolin 1/metabolism , DNA Repair/physiology , ErbB Receptors/metabolism , Lipid Peroxidation/radiation effects , src-Family Kinases/metabolism , Active Transport, Cell Nucleus , Blotting, Western , Carcinoma, Bronchogenic/pathology , Carcinoma, Bronchogenic/radiotherapy , Cell Line, Tumor/metabolism , Cell Line, Tumor/radiation effects , DNA Breaks, Double-Stranded/radiation effects , ErbB Receptors/radiation effects , Humans , Phosphorylation/radiation effects , Radiation, Ionizing , Sensitivity and Specificity , Tumor Cells, Cultured/metabolism , Tumor Cells, Cultured/radiation effects , src-Family Kinases/radiation effects
18.
Mol Cancer ; 7: 69, 2008 Sep 12.
Article in English | MEDLINE | ID: mdl-18789131

ABSTRACT

BACKGROUND: To elucidate the role of src kinase in caveolin-1 driven internalization and nuclear transport of EGFR linked to regulation of DNA-repair in irradiated cells. RESULTS: Ionizing radiation resulted in src kinase stabilization, activation and subsequent src mediated caveolin-1 Y14- and EGFR Y845-phosphorylations. Both phosphorylations were radiation specific and could not be observed after treatment with EGF. Inhibition of EGFR by the antibody Erbitux resulted in a strong accumulation of caveolin/EGFR complexes within the cytoplasm, which could not be further increased by irradiation. Radiation-induced caveolin-1- and EGFR-phosphorylations were associated with nuclear EGFR transport and activation of DNA-PK, as detected by phosphorylation at T2609. Blockage of src activity by the specific inhibitor PP2, decreased nuclear transport of EGFR and inhibited caveolin-1- and DNA-PK-phosphorylation. Knockdown of src by specific siRNA blocked EGFR phosphorylation at Y845, phosphorylation of caveolin-1 at Y14 and abolished EGFR transport into the nucleus and phosphorylation of DNA-PK. Consequently, both knockdown of src by specific siRNA and also inhibition of src activity by PP2 resulted in an enhanced residual DNA-damage as quantified 24 h after irradiation and increased radiosensitivity. CONCLUSION: Src kinase activation following irradiation triggered caveolin-1 dependent EGFR internalization into caveolae. Subsequently EGFR shuttled into the nucleus. As a consequence, inhibition of internalization and nuclear transport of EGFR blocked radiation-induced phosphorylation of DNA-PK and hampered repair of radiation-induced double strand breaks.


Subject(s)
Caveolin 1/metabolism , Cell Nucleus/metabolism , DNA-Activated Protein Kinase/metabolism , ErbB Receptors/metabolism , Active Transport, Cell Nucleus/radiation effects , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Cell Line, Tumor , Cetuximab , DNA Breaks, Double-Stranded , DNA Repair , Humans , Phosphorylation/radiation effects , RNA, Small Interfering/metabolism , Transfection , src-Family Kinases/metabolism
19.
DNA Repair (Amst) ; 7(10): 1746-56, 2008 Oct 01.
Article in English | MEDLINE | ID: mdl-18678286

ABSTRACT

As demonstrated recently, ionizing radiation (IR) can mediate phosphorylation of DNA-PKcs in human tumor cells through stimulation of the PI3K/Akt pathway. It is also known that DNA-PKcs directly interacts the X-ray repair cross-complementing group 1 protein (XRCC1) involved in base excision repair (BER). Therefore, in the present study we investigated the role of PI3K/Akt activity and DNA-PKcs on XRCC1 expression/stabilization. In contrast to the DNA-PKcs-deficient glioblastoma cell line MO59J, the DNA-PKcs-proficient counterpart MO59K as well as human lung adenocarcinoma A549 cells presented a high basal level of XRCC1 expression. Radiation doses of 3-12Gy did not stimulate a further enhanced expression of XRCC1 in DNA-PKcs-proficient cells (MO59K and A549) within 180min post-irradiation. However, a marked induction of XRCC1 expression was apparent in DNA-PKcs-deficient MO59J cells. Targeting of DNA-PKcs as well as PI3K/Akt pathway by specific kinase inhibitors and/or siRNA reduced basal XRCC1 expression in un-irradiated DNA-PKcs-proficient cells to the level observed in DNA-PKcs-deficient cells. Reduction of basal expression of XRCC1 by XRCC1-siRNA, AKT-siRNA as well as DNA-PKcs inhibitor facilitated IR-induced XRCC1 expression. XRCC1 expression induced by irradiation, however, was independent of PI3K/Akt signaling, but dependent of MAPK-ERK1/2. By immuno-precipitation experiments and confocal microscopy a complex formation of XRCC1 and DNA-PKcs was shown. Applying gamma-H2AX foci analysis it was shown that basal expression of XRCC1 is important for the repair of IR-induced DNA-double strand breaks (DNA-DSBs). These data indicate that IR-induced XRCC1 expression is dependent on the expression level of DNA-PKcs and basal activity status of PI3K/Akt signaling. Likewise, potential of IR-induced XRCC1 expression depends on its basal expression level.


Subject(s)
DNA-Binding Proteins/metabolism , MAP Kinase Signaling System , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Cell Line, Tumor , DNA Breaks, Double-Stranded/drug effects , DNA Breaks, Double-Stranded/radiation effects , DNA Repair/drug effects , DNA Repair/radiation effects , DNA-Activated Protein Kinase/deficiency , DNA-Activated Protein Kinase/metabolism , ErbB Receptors/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/radiation effects , Mitogen-Activated Protein Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Protein Binding/drug effects , Protein Binding/radiation effects , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Radiation, Ionizing , Recombinant Fusion Proteins/metabolism , Recombination, Genetic/drug effects , Recombination, Genetic/radiation effects , X-ray Repair Cross Complementing Protein 1
20.
Radiother Oncol ; 86(3): 375-82, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18237807

ABSTRACT

BACKGROUND AND PURPOSE: The purpose of the study was to elucidate the underlying molecular mechanism of the radioprotector, Bowman-Birk proteinase inhibitor (BBI), and its interaction with EGFR nuclear transport. MATERIALS AND METHODS: Molecular effects of BBI at the level of EGFR responses were investigated in vitro with wt. TP53 bronchial carcinoma cell line A549 and the transformed fibroblast cell line HH4dd characterized by a mt. TP53. EGFR and associated protein expression were quantified by Western blotting and confocal microscopy in the cytoplasmic and nuclear cell fraction. Residual DNA double strand breaks were quantified by means of a gammaH(2)AX focus assay. RESULTS: Both irradiation and BBI-treatment stimulated EGFR internalization into the cytoplasm. This process involved src kinase activation, EGFR phosphorylation at Y845, and caveolin 1 phosphorylation at Y14. EGFR internalization correlated with nuclear EGFR transport and was associated with phosphorylation of EGFR at T654. Nuclear EGFR was linked with DNA-PK complex formation and activation. Furthermore, nuclear EGFR was found in complex with TP53, phosphorylated at S15, and with MDC1, following irradiation and BBI treatment. It is noteworthy that MDC1 was strongly decreased in the nuclear EGFR complex in cells with mt. TP53 and failed to be increased by either BBI treatment or irradiation. Interestingly, in cells with mt. TP53 the BBI mediated stimulation of double strand break repair was hampered significantly. CONCLUSION: These data indicate that BBI stimulates complex formation between EGFR, TP53 and MDC1 protein in wt. TP53 cells only. Since MDC1 is essential for recruitment of DNA repair foci, this observation may explain how BBI selectively stimulated repair of DNA double strand breaks in wt. TP53 cells.


Subject(s)
DNA Repair/drug effects , ErbB Receptors/metabolism , Serine Proteinase Inhibitors/pharmacology , Trypsin Inhibitor, Bowman-Birk Soybean/pharmacology , Active Transport, Cell Nucleus/drug effects , Adaptor Proteins, Signal Transducing , Blotting, Western , Cell Cycle Proteins , Cell Line, Tumor , Cell Nucleus/metabolism , DNA Breaks, Double-Stranded , Gene Expression , Humans , Microscopy, Confocal , Nuclear Proteins/metabolism , Phosphorylation/drug effects , Trans-Activators/metabolism , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...