Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
Add more filters










Publication year range
1.
Nucleic Acids Res ; 52(4): e22, 2024 Feb 28.
Article in English | MEDLINE | ID: mdl-38261985

ABSTRACT

In the comet assay, tails are formed after single-cell gel electrophoresis if the cells have been exposed to genotoxic agents. These tails include a mixture of both DNA single-strand breaks (SSBs) and double-strand breaks (DSBs). However, these two types of strand breaks cannot be distinguished using comet assay protocols with conventional DNA stains. Since DSBs are more problematic for the cells, it would be useful if the SSBs and DSBs could be differentially identified in the same comet. In order to be able to distinguish between SSBs and DSBs, we designed a protocol for polymerase-assisted DNA damage analysis (PADDA) to be used in combination with the Flash comet protocol, or on fixed cells. By using DNA polymerase I to label SSBs and terminal deoxynucleotidyl transferase to label DSBs with fluorophore-labelled nucleotides. Herein, TK6-cells or HaCat cells were exposed to either hydrogen peroxide (H2O2), ionising radiation (X-rays) or DNA cutting enzymes, and then subjected to a comet protocol followed by PADDA. PADDA offers a wider detection range, unveiling previously undetected DNA strand breaks.


Subject(s)
Comet Assay , DNA Damage , Comet Assay/methods , DNA/genetics , DNA-Directed DNA Polymerase , Hydrogen Peroxide/toxicity
2.
Transl Oncol ; 26: 101543, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36126563

ABSTRACT

The bone-seeking radiopharmaceutical Xofigo (Radium-223 dichloride) has demonstrated both extended survival and palliative effects in treatment of bone metastases in prostate cancer. The alpha-particle emitter Ra-223, targets regions undergoing active bone remodeling and strongly binds to bone hydroxyapatite (HAp). However, the toxicity mechanism and properties of Ra-223 binding to hydroxyapatite are not fully understood. By exposing 2D and 3D (spheroid) prostate cancer cell models to free and HAp-bound Ra-223 we here studied cell toxicity, apoptosis and formation and repair of DNA double-strand breaks (DSBs). The rapid binding with a high affinity of Ra-223 to bone-like HAp structures was evident (KD= 19.2 × 10-18 M) and almost no dissociation was detected within 24 h. Importantly, there was no significant uptake of Ra-223 in cells. The Ra-223 alpha-particle decay produced track-like distributions of the DNA damage response proteins 53BP1 and É£H2AX induced high amounts of clustered DSBs in prostate cancer cells and activated DSB repair through non-homologous end-joining (NHEJ). Ra-223 inhibited growth of prostate cancer cells, independent of cell type, and induced high levels of apoptosis. In summary, we suggest the high cell killing efficacy of the Ra-223 was attributed to the clustered DNA damaged sites induced by α-particles.

3.
Cell Death Discov ; 8(1): 284, 2022 Jun 11.
Article in English | MEDLINE | ID: mdl-35690610

ABSTRACT

The antibody conjugate gemtuzumab ozogamicin (GO; Mylotarg®) provides targeted therapy of acute myeloid leukemia (AML), with recent approvals for patients with CD33-positive disease at diagnosis or relapse, as monotherapy or combined with chemotherapeutics. While its clinical efficacy is well documented, the molecular routes by which GO induces AML cell death warrant further analyses. We have earlier reported that this process is initiated via mitochondria-mediated caspase activation. Here we provide additional data, focusing on the involvement of caspase-2 in this mechanism. We show that this enzyme plays an important role in triggering apoptotic death of human AML cells after exposure to GO or its active moiety calicheamicin. Accordingly, the caspase-2 inhibitor z-VDVAD-fmk reduced GO-induced caspase-3 activation. This finding was validated with shRNA and siRNA targeting caspase-2, resulting in reduced caspase-3 activation and cleavage of poly [ADP-ribose] polymerase 1 (PARP-1). We previously demonstrated that GO-induced apoptosis included a conformational change of Bax into a pro-apoptotic state. Present data reveal that GO-treatment also induced Bid cleavage, which was partially reduced by caspase-2 specific inhibition while the effect on GO-induced Bax conformational change remained unaltered. In mononuclear cells isolated from AML patients that responded to GO treatment in vitro, processing of caspase-2 was evident, whereas in cells from an AML patient refractory to treatment no such processing was seen. When assessing diagnostic samples from 22 AML patients, who all entered complete remission (CR) following anthracycline-based induction therapy, and comparing patients with long versus those with short CR duration no significant differences in baseline caspase-2 or caspase-3 full-length protein expression levels were found. In summary, we demonstrate that GO triggers caspase-2 cleavage in human AML cells and that the subsequent apoptosis of these cells in part relies on caspase-2. These findings may have future clinical implications.

4.
Sci Rep ; 10(1): 5923, 2020 04 03.
Article in English | MEDLINE | ID: mdl-32246062

ABSTRACT

Oncogenic client-proteins of the chaperone Heat shock protein 90 (HSP90) insure unlimited tumor growth and are involved in resistance to chemo- and radiotherapy. The HSP90 inhibitor Onalespib initiates the degradation of oncoproteins, and might also act as a radiosensitizer. The aim of this study was therefore to evaluate the efficacy of Onalespib in combination with external beam radiotherapy in an in vitro and in vivo approach. Onalespib downregulated client proteins, lead to increased apoptosis and caused DNA-double-strands. Monotherapy and combination with radiotherapy reduced colony formation, proliferation and migration assessed in radiosensitive HCT116 and radioresistant A431 cells. In vivo, a minimal treatment regimen for 3 consecutive days of Onalespib (3 × 10 mg/kg) doubled survival, whereas Onalespib with radiotherapy (3 × 2 Gy) caused a substantial delay in tumor growth and prolonged the survival by a factor of 3 compared to the HCT116 xenografted control group. Our results demonstrate that Onalespib exerts synergistic anti-cancer effects when combined with radiotherapy, most prominent in the radiosensitive cell models. We speculate that the depletion and downregulation of client proteins involved in signalling, migration and DNA repair mechanisms is the cause. Thus, individually, or in combination with radiotherapy Onalespib inhibits tumor growth and has the potential to improve radiotherapy outcomes, prolonging the overall survival of cancer patients.


Subject(s)
Benzamides/pharmacology , Chemoradiotherapy/methods , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Isoindoles/pharmacology , Neoplasms/therapy , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Benzamides/therapeutic use , Cell Movement/drug effects , Cell Movement/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 , Down-Regulation/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/radiation effects , HCT116 Cells , HSP90 Heat-Shock Proteins/metabolism , Humans , Isoindoles/therapeutic use , Mice , Neoplasms/pathology , Radiation Tolerance/drug effects , Xenograft Model Antitumor Assays
5.
Int J Mol Sci ; 20(23)2019 Dec 03.
Article in English | MEDLINE | ID: mdl-31817026

ABSTRACT

In children, ketamine sedation is often used during radiological procedures. Combined exposure of ketamine and radiation at doses that alone did not affect learning and memory induced permanent cognitive impairment in mice. The aim of this study was to elucidate the mechanism behind this adverse outcome. Neonatal male NMRI mice were administered ketamine (7.5 mg kg-1) and irradiated (whole-body, 100 mGy or 200 mGy, 137Cs) one hour after ketamine exposure on postnatal day 10. The control mice were injected with saline and sham-irradiated. The hippocampi were analyzed using label-free proteomics, immunoblotting, and Golgi staining of CA1 neurons six months after treatment. Mice co-exposed to ketamine and low-dose radiation showed alterations in hippocampal proteins related to neuronal shaping and synaptic plasticity. The expression of brain-derived neurotrophic factor, activity-regulated cytoskeleton-associated protein, and postsynaptic density protein 95 were significantly altered only after the combined treatment (100 mGy or 200 mGy combined with ketamine, respectively). Increased numbers of basal dendrites and branching were observed only after the co-exposure, thereby constituting a possible reason for the displayed alterations in behavior. These data suggest that the risk of radiation-induced neurotoxic effects in the pediatric population may be underestimated if based only on the radiation dose.


Subject(s)
CA1 Region, Hippocampal/pathology , Ketamine/toxicity , Neurons/pathology , Neurons/radiation effects , Radiation, Ionizing , Animals , Animals, Newborn , Cell Shape/drug effects , Cell Shape/radiation effects , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Cytoskeleton/radiation effects , Male , Mice , Neuronal Plasticity/drug effects , Neuronal Plasticity/radiation effects , Neurons/drug effects , Proteome/metabolism
6.
Int J Oncol ; 55(6): 1287-1295, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31638190

ABSTRACT

177Lu­DOTATATE was recently approved for the treatment of somatostatin receptor (SSTR)­positive neuroendocrine tumors (NETs). However, despite impressive response rates, complete responses are rare. Heat shock protein 90 (HSP90) inhibitors have been suggested as suitable therapeutic agents for NETs, as well as a potential radiosensitizers. Consequently, the aim of this study was to investigate whether the HSP90­inhibitor onalespib could reduce NET cell growth and act as a radiosensitizer when used in combination with 177Lu­DOTATATE. The NET cell lines BON, NCI­H727 and NCI­H460, were first characterized with regards to 177Lu­DOTATATE uptake and sensitivity to onalespib treatment in monolayer cell assays. The growth inhibitory effects of the monotherapies and combination treatments were then examined in three­dimensional multicellular tumor spheroids. Lastly, the molecular effects of the treatments were assessed. 177Lu­DOTATATE uptake was observed in the BON and NCI­H727 cells, while the NCI­H460 cells exhibited no detectable uptake. Accordingly, 177Lu­DOTATATE reduced the growth of BON and NCI­H727 spheroids, while no effect was observed in the NCI­H460 spheroids. Onalespib reduced cell viability and spheroid growth in all three cell lines. Furthermore, the combination of onalespib and 177Lu­DOTATATE exerted synergistic therapeutic effects on the BON and NCI­H727 spheroids. Western blot analysis of BON spheroids revealed the downregulation of epidermal growth factor receptor (EGFR) and the upregulation of γ H2A histone family member X (γH2AX) following combined treatment with onalespib and 177Lu­DOTATATE. Moreover, flow cytometric analyses revealed a two­fold increase in caspase 3/7 activity in the combination group. In conclusion, the findings of this study demonstrate that onalespib exerts antitumorigenic effects on NET cells and may thus be a feasible treatment option for NETs. Furthermore, onalespib was able to synergistically potentiate 177Lu­DOTATATE treatment in a SSTR­specific manner. The radiosensitizing mechanisms of onalespib involved the downregulation of EGFR expression and the induction of apoptosis. Consequently, the combination of onalespib and 177Lu­DOTATATE may prove to be a promising strategy with which to improve therapeutic responses in patients with NETs. Further studies investigating this strategy in vivo regarding the therapeutic effects and potential toxicities are warranted to expand these promising findings.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Benzamides/pharmacology , Chemoradiotherapy/methods , Coordination Complexes/pharmacology , Isoindoles/pharmacology , Neuroendocrine Tumors/therapy , Octreotide/analogs & derivatives , Radiation-Sensitizing Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Benzamides/therapeutic use , Cell Line, Tumor , Coordination Complexes/therapeutic use , Drug Synergism , ErbB Receptors/metabolism , Gene Expression Regulation, Neoplastic/drug effects , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Humans , Isoindoles/therapeutic use , Neuroendocrine Tumors/pathology , Octreotide/pharmacology , Octreotide/therapeutic use , Radiation-Sensitizing Agents/therapeutic use , Receptors, Somatostatin/agonists , Spheroids, Cellular
7.
J Appl Toxicol ; 39(4): 582-589, 2019 04.
Article in English | MEDLINE | ID: mdl-30426514

ABSTRACT

Radiological methods for screening, diagnostics and therapy are often used in healthcare; however, it has recently been reported that developmental exposure to low-dose ionizing radiation (IR) causes neurotoxicity. Environmental chemicals also have the potential to affect the developing brain and the concomitant effects caused by IR and chemicals are of high interest today. We therefore aim to investigate if low-dose IR can interact with the known neurotoxicant paraquat to induce neurotoxicity in the neonatal mouse model. Using the same model, we also aim to investigate if fractionated low-dose IR can be as neurotoxic as higher acute doses. Male mice were exposed to a single dose of paraquat (0.2 or 0.02 mg/kg) on postnatal day 10 and 11. Two hours following paraquat exposure, mice were whole body irradiated with 100 or 300 mGy gamma radiation (137 Cs). Behavioural observations were performed at 2 and 3 months of age. Following behavioural testing, we evaluated striatal dopaminergic gene transcription. Animals co-exposed to IR and paraquat generally displayed altered spontaneous behaviour compared to controls and single agent exposed mice. Stronger effects by combined exposure were also observed on adult memory and learning. However, dopaminergic gene transcript levels remained unchanged by treatment. Co-exposure to low-dose IR and paraquat can interact to exacerbate neurotoxic effects and to impair cognitive function. Furthermore, fractionation of the radiation dose was observed to be as potent as higher acute exposure for induction of developmental neurotoxicity.


Subject(s)
Behavior, Animal , Brain/growth & development , Gamma Rays/adverse effects , Neurotoxicity Syndromes/etiology , Paraquat/toxicity , Animals , Animals, Newborn , Behavior, Animal/drug effects , Behavior, Animal/radiation effects , Brain/drug effects , Brain/radiation effects , Dose-Response Relationship, Radiation , Female , Male , Maze Learning/drug effects , Maze Learning/radiation effects , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Motor Activity/radiation effects , Neurotoxicity Syndromes/genetics , Neurotoxicity Syndromes/psychology , Transcription, Genetic/drug effects , Transcription, Genetic/radiation effects
8.
PLoS One ; 13(12): e0209594, 2018.
Article in English | MEDLINE | ID: mdl-30592737

ABSTRACT

DNA double-strand breaks (DSBs) are the most deleterious lesions that can arise in cells after ionizing radiation or radiometric drug treatment. In addition to prompt DSBs, DSBs may also be produced during repair, evolving from a clustered DNA damaged site, which is composed of two or more distinct lesions that are located within two helical turns. A specific type of cluster damage is the heat-sensitive clustered site (HSCS), which transforms into DSBs upon treatment at elevated temperatures. The actual lesions or mechanisms that mediate the HSCS transformation into DSBs are unknown. However, there are two possibilities; either these lesions are transformed into DSBs due to DNA lesion instability, e.g., transfer of HSCS into single-strand breaks (SSBs), or they are formed due to local DNA structure instability, e.g., DNA melting, where two SSBs on opposite strands meet and transform into a DSB. The importance of these processes in living cells is not understood, but they significantly affect estimates of DSB repair capacity. In this study, we show that HSCS removal in human cells is not affected by defects in DSB repair or inhibition of DSB repair. Under conditions where rejoining of prompt DSBs was almost completely inhibited, heat-sensitive DSBs were successfully rejoined, without resulting in increased DSB levels, indicating that HSCS do not transfer into DSB in cells under physiological conditions. Furthermore, analysis by atomic force microscopy suggests that prolonged heating of chromosomal DNA can induce structural changes that facilitate transformation of HSCS into DSB. In conclusion, the HSCS do not generate additional DSBs at physiological temperatures in human cells, and the repair of HSCS is independent of DSB repair.


Subject(s)
DNA Breaks, Double-Stranded , DNA Damage , Hot Temperature , Cell Line, Tumor , DNA Repair , Electrophoresis, Gel, Pulsed-Field , Humans , Microscopy, Atomic Force
9.
Radiat Res ; 188(6): 597-604, 2017 12.
Article in English | MEDLINE | ID: mdl-28952912

ABSTRACT

Uncontrolled generation of DNA double-strand breaks (DSBs) in cells is regarded as a highly toxic event that threatens cell survival. Radiation-induced DNA DSBs are commonly measured by pulsed-field gel electrophoresis, microscopic evaluation of accumulating DNA damage response proteins (e.g., 53BP1 or γ-H2AX) or flow cytometric analysis of γ-H2AX. The advantage of flow cytometric analysis is that DSB formation and repair can be studied in relationship to cell cycle phase or expression of other proteins. However, γ-H2AX is not able to monitor repair kinetics within the first 60 min postirradiation, a period when most DSBs undergo repair. A key protein in non-homologous end joining repair is the catalytic subunit of DNA-dependent protein kinase. Among several phosphorylation sites of DNA-dependent protein kinase, the threonine at position 2609 (T2609), which is phosphorylated by ataxia telangiectasia mutated (ATM) or DNA-dependent protein kinase catalytic subunit itself, activates the end processing of DSB. Using flow cytometry, we show here that phosphorylation at T2609 is faster in response to DSBs than γ-H2AX. Furthermore, flow cytometric analysis of T2609 resulted in a better representation of fast repair kinetics than analysis of γ-H2AX. In cells with reduced ligase IV activity, and wild-type cells where DNA-dependent protein kinase activity was inhibited, the reduced DSB repair capacity was observed by T2609 evaluation using flow cytometry. In conclusion, flow cytometric evaluation of DNA-dependent protein kinase T2609 can be used as a marker for early DSB repair and gives a better representation of early repair events than analysis of γ-H2AX.


Subject(s)
DNA Breaks, Double-Stranded , DNA End-Joining Repair , DNA-Activated Protein Kinase/physiology , Flow Cytometry , Nuclear Proteins/physiology , Protein Processing, Post-Translational , Cell Line , Chromones/pharmacology , DNA/radiation effects , DNA-Activated Protein Kinase/antagonists & inhibitors , Dose-Response Relationship, Radiation , Electrophoresis, Gel, Pulsed-Field , Gamma Rays , Histones/physiology , Humans , Morpholines/pharmacology , Nuclear Proteins/antagonists & inhibitors , Phosphorylation/drug effects , Phosphothreonine/metabolism , Protein Processing, Post-Translational/drug effects , Time Factors
10.
Int J Oncol ; 50(1): 5-14, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27878243

ABSTRACT

AKT is a central protein in many cellular pathways such as cell survival, proliferation, glucose uptake, metabolism, angiogenesis, as well as radiation and drug response. The three isoforms of AKT (AKT1, AKT2 and AKT3) are proposed to have different physiological functions, properties and expression patterns in a cell type-dependent manner. As of yet, not much is known about the influence of the different AKT isoforms in the genome and their effects in the metabolism of colorectal cancer cells. In the present study, DLD-1 isogenic AKT1, AKT2 and AKT1/2 knockout colon cancer cell lines were used as a model system in conjunction with the parental cell line in order to further elucidate the differences between the AKT isoforms and how they are involved in various cellular pathways. This was done using genome wide expression analyses, metabolic profiling and cell migration assays. In conclusion, downregulation of genes in the cell adhesion, extracellular matrix and Notch-pathways and upregulation of apoptosis and metastasis inhibitory genes in the p53-pathway, confirm that the knockout of both AKT1 and AKT2 will attenuate metastasis and tumor cell growth. This was verified with a reduction in migration rate in the AKT1 KO and AKT2 KO and most explicitly in the AKT1/2 KO. Furthermore, the knockout of AKT1, AKT2 or both, resulted in a reduction in lactate and alanine, suggesting that the metabolism of carbohydrates and glutathione was impaired. This was further verified in gene expression analyses, showing downregulation of genes involved in glucose metabolism. Additionally, both AKT1 KO and AKT2 KO demonstrated an impaired fatty acid metabolism. However, genes were upregulated in the Wnt and cell proliferation pathways, which could oppose this effect. AKT inhibition should therefore be combined with other effectors to attain the best effect.


Subject(s)
Cell Movement/genetics , Colonic Neoplasms/genetics , Proto-Oncogene Proteins c-akt/metabolism , Alanine/genetics , Alanine/metabolism , Apoptosis/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Fatty Acids/metabolism , Gene Expression Regulation, Neoplastic , Gene Knockout Techniques , Glucose/metabolism , Humans , Lactic Acid/metabolism , Metabolic Networks and Pathways/genetics , Neoplasm Metastasis , Proto-Oncogene Proteins c-akt/biosynthesis , Proto-Oncogene Proteins c-akt/genetics
11.
Int J Radiat Biol ; 92(7): 371-9, 2016 07.
Article in English | MEDLINE | ID: mdl-27043364

ABSTRACT

PURPOSE: To investigate whether neonatal exposure to fractionated external gamma radiation and co-exposure to radiation and nicotine can affect/exacerbate developmental neurotoxic effects, including altered behavior/cognitive function and the susceptibility of the cholinergic system in adult male mice. MATERIALS AND METHODS: Neonatal male Naval Medical Research Institute (NMRI) mice were irradiated with one 200 mGy fraction/day and/or exposed to nicotine (66 µg/kg b.w.) twice daily on postnatal day (PND) 10, 10-11, 10-12 or 10-13 (nicotine only). At 2 months of age the animals were tested for spontaneous behavior in a novel home environment, habituation capacity and nicotine-induced behavior. RESULTS: Fractionated irradiation and co-exposure to radiation and nicotine on three consecutive days disrupted behavior and habituation and altered susceptibility of the cholinergic system. All observed effects were significantly more pronounced in mice co-exposed to both radiation and nicotine. CONCLUSIONS: The fractionated irradiation regime affects behavior/cognitive function in a similar manner as has previously been observed for single-dose exposures. Neonatal co-exposure to radiation and nicotine, during a critical period of brain development in general and cholinergic system development in particular, enhance these behavioral defects suggesting that the cholinergic system can be a target system for this type of developmental neurotoxic effects.


Subject(s)
Behavioral Symptoms/etiology , Brain/radiation effects , Cholinergic Neurons/drug effects , Cognitive Dysfunction/etiology , Nicotine/toxicity , Radiation Tolerance/drug effects , Animals , Animals, Newborn , Behavioral Symptoms/physiopathology , Brain/drug effects , Cholinergic Neurons/radiation effects , Cognitive Dysfunction/physiopathology , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Gamma Rays/adverse effects , Male , Mice
12.
Behav Brain Res ; 304: 11-9, 2016 May 01.
Article in English | MEDLINE | ID: mdl-26876140

ABSTRACT

Development of the brain includes periods which can be critical for its normal maturation. The present study investigates specifically vulnerable peri-/postnatal periods in mice which are essential for understanding the etiology behind radiation induced neurotoxicity and functional defects, including evaluation of neurotoxicity between sexes or commonly used laboratory mouse strains following low/moderate doses of ionizing radiation (IR). Male Naval Medical Research Institute (NMRI) mice, whole body irradiated to a single 500 mGy IR dose, on postnatal day (PND) 3 or PND 10 showed an altered adult spontaneous behaviour and impaired habituation capacity, whereas irradiation on PND 19 did not have any impact on the studied variables. Both NMRI and C57bl/6 male and female mice showed an altered adult spontaneous behaviour and impaired habituation following a single whole body irradiation of 500 or 1000 mGy, but not after 20 or 100 mGy, on PND 10. The present study shows that exposure to low/moderate doses of IR during critical life stages might be involved in the induction of neurological/neurodegenerative disorder/disease. A specifically vulnerable period for radiation induced neurotoxicity seems to be around PND 3-10 in mice. Further studies are needed to investigate mechanisms involved in induction of developmental neurotoxicity following low-dose irradiation.


Subject(s)
Behavioral Symptoms/etiology , Neurodevelopmental Disorders/etiology , Sex Characteristics , Whole-Body Irradiation/adverse effects , Age Factors , Animals , Animals, Newborn , Dose-Response Relationship, Radiation , Exploratory Behavior/radiation effects , Female , Locomotion/radiation effects , Male , Mice , Pregnancy , Radiation, Ionizing , Species Specificity , Statistics, Nonparametric , Time Factors
13.
Eur J Nucl Med Mol Imaging ; 43(5): 974-982, 2016 May.
Article in English | MEDLINE | ID: mdl-26627081

ABSTRACT

PURPOSE: Heat shock protein 90 (HSP90) is essential for the activation and stabilization of numerous oncogenic client proteins. AT13387 is a novel HSP90 inhibitor promoting degradation of oncogenic proteins upon binding, and may also act as a radiosensitizer. For optimal treatment there is, however, the need for identification of biomarkers for patient stratification and therapeutic response monitoring, and to find suitable targets for combination treatments. The aim of this study was to assess the response of surface antigens commonly expressed in squamous cell carcinoma to AT13387 treatment, and to find suitable biomarkers for molecular imaging and radioimmunotherapy in combination with HSP90 inhibition. METHODS: Cancer cell proliferation and radioimmunoassays were used to evaluate the effect of AT13387 on target antigen expression in vitro. Inhibitor effects were then assessed in vivo in mice-xenografts. Animals were treated with AT13387 (5 × 50 mg/kg), and were imaged with PET using either (18)F-FDG or (124)I-labelled tracers for EGFR and CD44v6, and this was followed by ex-vivo biodistribution analysis and immunohistochemical staining. RESULTS: AT13387 exposure resulted in high cytotoxicity and possible radiosensitization with IC50 values below 4 nM. Both in vitro and in vivo AT13387 effectively downregulated HSP90 client proteins. PET imaging with (124)I-cetuximab showed a significant decrease of EGFR in AT13387-treated animals compared with untreated animals. In contrast, the squamous cell carcinoma-associated biomarker CD44v6, visualized with (124)I-AbD19384 as well as (18)F-FDG uptake, were not significantly altered by AT13387 treatment. CONCLUSION: We conclude that AT13387 downregulates HSP90 client proteins, and that molecular imaging of these proteins may be a suitable approach for assessing treatment response. Furthermore, radioimmunotherapy targeting CD44v6 in combination with AT13387 may potentiate the radioimmunotherapy outcome due to radiosensitizing effects of the drug, and could potentially lead to a lower dose to normal tissues.


Subject(s)
Benzamides/pharmacokinetics , Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/diagnostic imaging , ErbB Receptors/metabolism , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Hyaluronan Receptors/metabolism , Isoindoles/pharmacokinetics , Animals , Benzamides/adverse effects , Benzamides/therapeutic use , Carcinoma, Squamous Cell/radiotherapy , Cell Line , Female , Fluorodeoxyglucose F18 , HSP90 Heat-Shock Proteins/metabolism , Humans , Iodine Radioisotopes , Isoindoles/adverse effects , Isoindoles/therapeutic use , Mice , Mice, Inbred BALB C , Mice, Nude , Positron-Emission Tomography , Radioimmunotherapy , Radiopharmaceuticals
14.
Oncotarget ; 6(34): 35652-66, 2015 Nov 03.
Article in English | MEDLINE | ID: mdl-26452257

ABSTRACT

Overexpression of heat shock protein 90 (HSP90) is associated with increased tumor cell survival and radioresistance. In this study we explored the efficacy of the novel HSP90 inhibitor AT13387 and examined its radiosensitizing effects in combination with gamma-radiation in 2D and 3D structures as well as mice-xenografts. AT13387 induced effective cytotoxic activity and radiosensitized cancer cells in monolayer and tumor spheroid models, where low drug doses triggered significant synergistic effects on cell survival together with radiation. Furthermore, AT13387 treatment resulted in G2/M-phase arrest and significantly reduced the migration capacity. The expression of selected client proteins involved in DNA repair, cell-signaling and cell growth was downregulated in vitro, though the expression of most investigated proteins recurred after 8-24 h. These results were confirmed in vivo where AT13387 treated tumors displayed effective downregulation of HSP90 and its oncogenic client proteins.In conclusion, our results demonstrate that AT13387 is a potent new cancer drug and effective radiosensitizer in vitro with an excellent in vivo efficacy. AT13387 treatment has the potential to improve external beam therapy and radionuclide therapy outcomes and restore treatment efficacy in cancers that are resistant to initial therapeutic regimes.


Subject(s)
Adenocarcinoma/radiotherapy , Benzamides/administration & dosage , Carcinoma, Squamous Cell/radiotherapy , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Isoindoles/administration & dosage , Radiation-Sensitizing Agents/administration & dosage , Animals , Benzamides/adverse effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Movement/drug effects , DNA Repair/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Isoindoles/adverse effects , Mice , Mice, Nude , Radiation-Sensitizing Agents/adverse effects , Xenograft Model Antitumor Assays
15.
Int J Radiat Biol ; 91(10): 820-6, 2015.
Article in English | MEDLINE | ID: mdl-26136085

ABSTRACT

PURPOSE: Radiation with high linear energy transfer (LET) produces clustering of DNA double-strand breaks (DSB) as well as non-DSB lesions. Heat-labile sites (HLS) are non-DSB lesions in irradiated cells that may convert into DSB at elevated temperature during preparation of naked DNA for electrophoretic assays and here we studied the initial formation and repair of these clustered damaged sites after irradiation with high LET ions. MATERIALS AND METHODS: Induction and repair of DSB were studied in normal human skin fibroblast (GM5758) after irradiation with accelerated carbon and nitrogen ions at an LET of 125 eV/nm. DNA fragmentation was analyzed by pulsed-field gel electrophoresis (PFGE) and by varying the lysis condition we could differentiate between prompt DSB and heat-released DSB. RESULTS: Before repair (t = 0 h), the 125 eV/nm ions produced a significant fraction of heat-released DSB, which appeared clustered on DNA fragments with sizes of 1 Mbp or less. These heat-released DSB increased the total number of DSB by 30-40%. This increase is similar to what has been found in low-LET irradiated cells, suggesting that the relative biological effectiveness (RBE) for DSB induction will not be largely affected by the lysis temperature. After 1-2 hours repair, a large fraction of DSB was still unrejoined but there was essentially no heat-released DSB present. CONCLUSIONS: These results suggest that high LET radiation, as low LET gamma radiation, induces a significant fraction of heat-labile sites which can be converted into DSB, and these heat-released DSB may affect both induction yields and estimates of repair.


Subject(s)
DNA Damage , DNA Repair/radiation effects , Linear Energy Transfer , Carbon/adverse effects , DNA Breaks, Double-Stranded/radiation effects , DNA Fragmentation/radiation effects , Fibroblasts/metabolism , Fibroblasts/radiation effects , Humans , Nitrogen/adverse effects , Skin/cytology
16.
Mol Neurodegener ; 9: 57, 2014 Dec 16.
Article in English | MEDLINE | ID: mdl-25515237

ABSTRACT

BACKGROUND/PURPOSE OF THE STUDY: Epidemiological evidence suggests that low doses of ionising radiation (≤1.0 Gy) produce persistent alterations in cognition if the exposure occurs at a young age. The mechanisms underlying such alterations are unknown. We investigated the long-term effects of low doses of total body gamma radiation on neonatally exposed NMRI mice on the molecular and cellular level to elucidate neurodegeneration. RESULTS: Significant alterations in spontaneous behaviour were observed at 2 and 4 months following a single 0.5 or 1.0 Gy exposure. Alterations in the brain proteome, transcriptome, and several miRNAs were analysed 6-7 months post-irradiation in the hippocampus, dentate gyrus (DG) and cortex. Signalling pathways related to synaptic actin remodelling such as the Rac1-Cofilin pathway were altered in the cortex and hippocampus. Further, synaptic proteins MAP-2 and PSD-95 were increased in the DG and hippocampus (1.0 Gy). The expression of synaptic plasticity genes Arc, c-Fos and CREB was persistently reduced at 1.0 Gy in the hippocampus and cortex. These changes were coupled to epigenetic modulation via increased levels of microRNAs (miR-132/miR-212, miR-134). Astrogliosis, activation of insulin-growth factor/insulin signalling and increased level of microglial cytokine TNFα indicated radiation-induced neuroinflammation. In addition, adult neurogenesis within the DG was persistently negatively affected after irradiation, particularly at 1.0 Gy. CONCLUSION: These data suggest that neurocognitive disorders may be induced in adults when exposed at a young age to low and moderate cranial doses of radiation. This raises concerns about radiation safety standards and regulatory practices.


Subject(s)
Brain/radiation effects , Cognition/radiation effects , Neurogenesis/radiation effects , Neuronal Plasticity/radiation effects , Radiation Injuries, Experimental , Animals , Animals, Newborn , Behavior, Animal/radiation effects , Fluorescent Antibody Technique , Immunoblotting , Immunohistochemistry , Male , Mice , Signal Transduction/radiation effects
17.
PLoS One ; 9(10): e110464, 2014.
Article in English | MEDLINE | ID: mdl-25329592

ABSTRACT

Patients suffering from brain malignancies are treated with high-dose ionising radiation. However, this may lead to severe learning and memory impairment. Preventive treatments to minimise these side effects have not been possible due to the lack of knowledge of the involved signalling pathways and molecular targets. Mouse hippocampal neuronal HT22 cells were irradiated with acute gamma doses of 0.5 Gy, 1.0 Gy and 4.0 Gy. Changes in the cellular proteome were investigated by isotope-coded protein label technology and tandem mass spectrometry after 4 and 24 hours. To compare the findings with the in vivo response, male NMRI mice were irradiated on postnatal day 10 with a gamma dose of 1.0 Gy, followed by evaluation of the cellular proteome of hippocampus and cortex 24 hours post-irradiation. Analysis of the in vitro proteome showed that signalling pathways related to synaptic actin-remodelling were significantly affected at 1.0 Gy and 4.0 Gy but not at 0.5 Gy after 4 and 24 hours. We observed radiation-induced reduction of the miR-132 and Rac1 levels; miR-132 is known to regulate Rac1 activity by blocking the GTPase-activating protein p250GAP. In the irradiated hippocampus and cortex we observed alterations in the signalling pathways similar to those in vitro. The decreased expression of miR-132 and Rac1 was associated with an increase in hippocampal cofilin and phospho-cofilin. The Rac1-Cofilin pathway is involved in the modulation of synaptic actin filament formation that is necessary for correct spine and synapse morphology to enable processes of learning and memory. We suggest that acute radiation exposure leads to rapid dendritic spine and synapse morphology alterations via aberrant cytoskeletal signalling and processing and that this is associated with the immediate neurocognitive side effects observed in patients treated with ionising radiation.


Subject(s)
Actin Cytoskeleton/metabolism , Gamma Rays/adverse effects , Hippocampus/metabolism , Neuronal Plasticity/radiation effects , Signal Transduction/radiation effects , Animals , Cell Line, Transformed , GTPase-Activating Proteins/metabolism , Hippocampus/pathology , Learning/radiation effects , Male , Memory/radiation effects , Mice , MicroRNAs/metabolism , Neuropeptides/metabolism , Proteome/metabolism , rac1 GTP-Binding Protein/metabolism
18.
Neurotoxicology ; 45: 48-55, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25265567

ABSTRACT

Medical use of ionizing radiation (IR) has great benefits for treatment and diagnostic imaging, but procedures as computerized tomography (CT) may deliver a significant radiation dose to the patient. Recently, awareness has been raised about possible non-cancer consequences from low dose exposure to IR during critical phases of perinatal and/or neonatal brain development. In the present study neonatal NMRI mice were whole body irradiated with a single dose of gamma radiation (0; 350 and 500 mGy) on postnatal day 10 (PND 10). At 2 and 4 months of age, mice of both sexes were observed for spontaneous behaviour in a novel home environment. The neuroproteins CaMKII, GAP-43, synaptophysin and total tau in male mouse cerebral cortex and hippocampus were analysed 24h post-irradiation and in adults at 6 months of age exposed to 0 or 500 mGy on PND 10. A significantly dose-response related deranged spontaneous behaviour in 2- and 4-month-old mice was observed, where both males and females displayed a modified habituation, indicating reduced cognitive function. The dose of 350 mGy seems to be a tentative threshold. Six-month-old male mice showed a significantly increased level of total tau in cerebral cortex after irradiation to 500 mGy compared to controls. This demonstrates that a single moderate dose of IR, given during a defined critical period of brain development, is sufficient to cause persistently reduced cognitive function. Moreover, an elevation of tau protein was observed in male mice displaying reduced cognitive function.


Subject(s)
Behavior, Animal/radiation effects , Cerebral Cortex/radiation effects , Gamma Rays/adverse effects , Animals , Animals, Newborn , Calcium-Calmodulin-Dependent Protein Kinase Type 2/analysis , Cerebral Cortex/metabolism , Dose-Response Relationship, Radiation , Female , GAP-43 Protein/analysis , Magnetic Resonance Spectroscopy , Male , Mice , Motor Activity/radiation effects , Synaptophysin/analysis , tau Proteins/analysis
19.
PLoS One ; 9(4): e94621, 2014.
Article in English | MEDLINE | ID: mdl-24760019

ABSTRACT

The cell surface proteins CD133, CD24 and CD44 are putative markers for cancer stem cell populations in colon cancer, associated with aggressive cancer types and poor prognosis. It is important to understand how these markers may predict treatment outcomes, determined by factors such as radioresistance. The scope of this study was to assess the connection between EGFR, CD133, CD24, and CD44 (including isoforms) expression levels and radiation sensitivity, and furthermore analyze the influence of AKT isoforms on the expression patterns of these markers, to better understand the underlying molecular mechanisms in the cell. Three colon cancer cell-lines were used, HT-29, DLD-1, and HCT116, together with DLD-1 isogenic AKT knock-out cell-lines. All three cell-lines (HT-29, HCT116 and DLD-1) expressed varying amounts of CD133, CD24 and CD44 and the top ten percent of CD133 and CD44 expressing cells (CD133high/CD44high) were more resistant to gamma radiation than the ten percent with lowest expression (CD133low/CD44low). The AKT expression was lower in the fraction of cells with low CD133/CD44. Depletion of AKT1 or AKT2 using knock out cells showed for the first time that CD133 expression was associated with AKT1 but not AKT2, whereas the CD44 expression was influenced by the presence of either AKT1 or AKT2. There were several genes in the cell adhesion pathway which had significantly higher expression in the AKT2 KO cell-line compared to the AKT1 KO cell-line; however important genes in the epithelial to mesenchymal transition pathway (CDH1, VIM, TWIST1, SNAI1, SNAI2, ZEB1, ZEB2, FN1, FOXC2 and CDH2) did not differ. Our results demonstrate that CD133high/CD44high expressing colon cancer cells are associated with AKT and increased radiation resistance, and that different AKT isoforms have varying effects on the expression of cancer stem cell markers, which is an important consideration when targeting AKT in a clinical setting.


Subject(s)
Antigens, CD/metabolism , CD24 Antigen/metabolism , Colonic Neoplasms/metabolism , Glycoproteins/metabolism , Hyaluronan Receptors/metabolism , Neoplastic Stem Cells/metabolism , Peptides/metabolism , Protein Isoforms/metabolism , AC133 Antigen , Cell Line, Tumor , HCT116 Cells , HT29 Cells , Humans , Oncogene Protein v-akt/metabolism
20.
Mutat Res ; 769: 1-10, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25771720

ABSTRACT

Efficient and correct repair of DNA double-strand break (DSB) is critical for cell survival. Defects in the DNA repair may lead to cell death, genomic instability and development of cancer. The catalytic subunit of DNA-dependent protein kinase (DNA-PKcs) is an essential component of the non-homologous end joining (NHEJ) which is the major DSB repair pathway in mammalian cells. In the present study, by using siRNA against DNA-PKcs in four human cell lines, we examined how low levels of DNA-PKcs affected cellular response to ionizing radiation. Decrease of DNA-PKcs levels by 80-95%, induced by siRNA treatment, lead to extreme radiosensitivity, similar to that seen in cells completely lacking DNA-PKcs and low levels of DNA-PKcs promoted cell accumulation in G2/M phase after irradiation and blocked progression of mitosis. Surprisingly, low levels of DNA-PKcs did not affect the repair capacity and the removal of 53BP1 or γ-H2AX foci and rejoining of DSB appeared normal. This was in strong contrast to cells completely lacking DNA-PKcs and cells treated with the DNA-PKcs inhibitor NU7441, in which DSB repair were severely compromised. This suggests that there are different mechanisms by which loss of DNA-PKcs functions can sensitize cells to ionizing radiation. Further, foci of phosphorylated DNA-PKcs (T2609 and S2056) co-localized with DSB and this was independent of the amount of DNA-PKcs but foci of DNA-PKcs was only seen in siRNA-treated cells. Our study emphasizes on the critical role of DNA-PKcs for maintaining survival after radiation exposure which is uncoupled from its essential function in DSB repair. This could have implications for the development of therapeutic strategies aiming to radiosensitize tumors by affecting the DNA-PKcs function.


Subject(s)
Chromones/pharmacology , DNA Breaks, Double-Stranded/drug effects , DNA Repair/drug effects , DNA-Activated Protein Kinase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Morpholines/pharmacology , Radiation Tolerance/drug effects , Cell Survival/drug effects , Cell Survival/radiation effects , Cells, Cultured , DNA Repair/genetics , DNA-Activated Protein Kinase/genetics , DNA-Activated Protein Kinase/metabolism , Gene Expression Regulation, Enzymologic/drug effects , HCT116 Cells , Humans , Mitosis/drug effects , Mitosis/genetics , Mitosis/radiation effects , RNA, Small Interfering/pharmacology , Radiation Tolerance/genetics , Radiation-Sensitizing Agents/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...