Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nucleic Acids Res ; 52(4): e22, 2024 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-38261985

RESUMEN

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.


Asunto(s)
Ensayo Cometa , Daño del ADN , Ensayo Cometa/métodos , ADN/genética , ADN Polimerasa Dirigida por ADN , Peróxido de Hidrógeno/toxicidad
2.
J Appl Toxicol ; 39(4): 582-589, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30426514

RESUMEN

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.


Asunto(s)
Conducta Animal , Encéfalo/crecimiento & desarrollo , Rayos gamma/efectos adversos , Síndromes de Neurotoxicidad/etiología , Paraquat/toxicidad , Animales , Animales Recién Nacidos , Conducta Animal/efectos de los fármacos , Conducta Animal/efectos de la radiación , Encéfalo/efectos de los fármacos , Encéfalo/efectos de la radiación , Relación Dosis-Respuesta en la Radiación , Femenino , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/efectos de la radiación , Ratones , Ratones Endogámicos C57BL , Actividad Motora/efectos de los fármacos , Actividad Motora/efectos de la radiación , Síndromes de Neurotoxicidad/genética , Síndromes de Neurotoxicidad/psicología , Transcripción Genética/efectos de los fármacos , Transcripción Genética/efectos de la radiación
3.
Int J Mol Sci ; 20(23)2019 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-31817026

RESUMEN

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.


Asunto(s)
Región CA1 Hipocampal/patología , Ketamina/toxicidad , Neuronas/patología , Neuronas/efectos de la radiación , Radiación Ionizante , Animales , Animales Recién Nacidos , Forma de la Célula/efectos de los fármacos , Forma de la Célula/efectos de la radiación , Citoesqueleto/efectos de los fármacos , Citoesqueleto/metabolismo , Citoesqueleto/efectos de la radiación , Masculino , Ratones , Plasticidad Neuronal/efectos de los fármacos , Plasticidad Neuronal/efectos de la radiación , Neuronas/efectos de los fármacos , Proteoma/metabolismo
4.
Eur J Nucl Med Mol Imaging ; 43(5): 974-982, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26627081

RESUMEN

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.


Asunto(s)
Benzamidas/farmacocinética , Biomarcadores de Tumor/metabolismo , Carcinoma de Células Escamosas/diagnóstico por imagen , Receptores ErbB/metabolismo , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Receptores de Hialuranos/metabolismo , Isoindoles/farmacocinética , Animales , Benzamidas/efectos adversos , Benzamidas/uso terapéutico , Carcinoma de Células Escamosas/radioterapia , Línea Celular , Femenino , Fluorodesoxiglucosa F18 , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Radioisótopos de Yodo , Isoindoles/efectos adversos , Isoindoles/uso terapéutico , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Tomografía de Emisión de Positrones , Radioinmunoterapia , Radiofármacos
5.
Tumour Biol ; 35(4): 3525-34, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24338765

RESUMEN

In response to ionizing radiation, several signaling cascades in the cell are activated to repair the DNA breaks, prevent apoptosis, and keep the cells proliferating. AKT is important for survival and proliferation and may also be an activating factor for DNA-PKcs and MRE11, which are essential proteins in the DNA repair process. AKT (PKB) is hyperactivated in several cancers and is associated with resistance to radiotherapy and chemotherapy. There are three AKT isoforms (AKT1, AKT2, and AKT3) with different expression patterns and functions in several cancer tumors. The role of AKT isoforms has been investigated in relation to radiation response and their effects on DNA repair proteins (DNA-PKcs and MRE11) in colon cancer cell lines. The knockout of AKT1 and/or AKT2 affected the radiation sensitivity, and a deficiency of both isoforms impaired the rejoining of radiation-induced DNA double strand breaks. Importantly, the active/phosphorylated forms of AKT and DNA-PKcs associate and exposure to ionizing radiation causes an increase in this interaction. Moreover, an increased expression of both DNA-PKcs and MRE11 was observed when AKT expression was ablated, yet only DNA-PKcs expression influenced AKT phosphorylation. Taken together, these results demonstrate a role for both AKT1 and AKT2 in radiotherapy response in colon cancer cells involving DNA repair capacity through the nonhomologous end joining pathway, thus suggesting that AKT in combination with DNA-PKcs inhibition may be used for radiotherapy sensitizing strategies in colon cancer.


Asunto(s)
Neoplasias del Colon/radioterapia , Reparación del ADN , Proteínas Proto-Oncogénicas c-akt/fisiología , Tolerancia a Radiación , Línea Celular Tumoral , Neoplasias del Colon/genética , Reparación del ADN/efectos de la radiación , Proteína Quinasa Activada por ADN/genética , Proteína Quinasa Activada por ADN/fisiología , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Receptores ErbB/fisiología , Humanos , Proteína Homóloga de MRE11 , Fosforilación , Isoformas de Proteínas/fisiología
6.
Cell Microbiol ; 15(1): 98-113, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22998585

RESUMEN

Epidemiological evidence links chronic bacterial infections to the increased incidence of certain types of cancer but the molecular mechanisms by which bacteria contribute to tumour initiation and progression are still poorly characterized. Here we show that chronic exposure to the genotoxin cytolethal distending toxin (CDT) of Gram-negative bacteria promotes genomic instability and acquisition of phenotypic properties of malignancy in fibroblasts and colon epithelial cells. Cells grown for more than 30 weeks in the presence of sublethal doses of CDT showed increased mutation frequency, and accumulation of chromatin and chromosomal aberrations in the absence of significant alterations of cell cycle distribution, decreased viability or senescence. Cell survival was dependent on sustained activity of the p38 MAP kinase. The ongoing genomic instability was associated with impaired activation of the DNA damage response and failure to efficiently activate cell cycle checkpoints upon exposure to genotoxic stress. Independently selected sublines showed enhanced anchorage-independent growth as assessed by the formation of colonies in semisolid agarose. These findings support the notion that chronic infection by CDT-producing bacteria may promote malignant transformation, and point to the impairment of cellular control mechanisms associated with the detection and repair of DNA damage as critical events in the process.


Asunto(s)
Toxinas Bacterianas/metabolismo , Daño del ADN/efectos de los fármacos , Inestabilidad Genómica/efectos de los fármacos , Bacterias Gramnegativas/patogenicidad , Mutágenos/metabolismo , Animales , Línea Celular , Supervivencia Celular/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Humanos , Ratas
7.
Artículo en Inglés | MEDLINE | ID: mdl-38898535

RESUMEN

BACKGROUND: The local anaesthetic lidocaine is widely used in the neonatal intensive unit to treat seizures in premature babies. However, other antiepileptics administered during early development in various animal models have shown negative long-term behavioural effects. Since no long-term behavioural data so far exist regarding lidocaine exposure at an early age, we decided to perform this extended follow-up study using a sensitive behavioural test. METHODS: Neonatal mice received a subcutaneous administration of saline or one dose of lidocaine (0.5, 4, or 12 mg kg-1) on postnatal day 10 (P10; peak of the Brain Growth Spurt). A well-established test to detect long-term behavioural alterations was conducted at 2 and 6 months of age, corresponding to early and late adulthood in humans. RESULTS: All animal survived to later testing. No signs of acute toxicity were observed. Lidocaine exposure did not result in any negative behavioural effects during habituation to a new home environment at any of the two studied time points, compared to saline placebo. CONCLUSIONS: Lidocaine does not by itself produce any negative long-term behavioural effects in mice exposed in early life (P10) despite long-term follow-up. This is reassuring regarding the current practice of treating seizures in premature babies with intravenous lidocaine.

8.
Transl Oncol ; 26: 101543, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36126563

RESUMEN

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.

9.
Cell Death Discov ; 8(1): 284, 2022 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-35690610

RESUMEN

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.

10.
Mol Cell Proteomics ; 8(5): 1117-29, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19168796

RESUMEN

During the past decade, we have witnessed an explosive increase in generation of large proteomics data sets, not least in cancer research. There is a growing need to extract and correctly interpret information from such data sets to generate biologically relevant hypotheses. A pathway search engine (PSE) has recently been developed as a novel tool intended to meet these requirements. Ionizing radiation (IR) is an anticancer treatment modality that triggers multiple signal transduction networks. In this work, we show that high linear energy transfer (LET) IR induces apoptosis in a non-small cell lung cancer cell line, U-1810, whereas low LET IR does not. PSE was applied to study changes in pathway status between high and low LET IR to find pathway candidates of importance for high LET-induced apoptosis. Such pathways are potential clinical targets, and they were further validated in vitro. We used an unsupervised shotgun proteomics approach where high resolution mass spectrometry coupled to nanoflow liquid chromatography determined the identity and relative abundance of expressed proteins. Based on the proteomics data, PSE suggested the JNK pathway (p = 6.10(-6)) as a key event in response to high LET IR. In addition, the Fas pathway was found to be activated (p = 3.10(-5)) and the p38 pathway was found to be deactivated (p = 0.001) compared with untreated cells. Antibody-based analyses confirmed that high LET IR caused an increase in phosphorylation of JNK. Moreover pharmacological inhibition of JNK blocked high LET-induced apoptotic signaling. In contrast, neither an activation of p38 nor a role for p38 in high LET IR-induced apoptotic signaling was found. We conclude that, in contrast to conventional low LET IR, high LET IR can trigger activation of the JNK pathway, which in turn is critical for induction of apoptosis in these cells. Thus PSE predictions were largely confirmed, and PSE was proven to be a useful hypothesis-generating tool.


Asunto(s)
Apoptosis/efectos de la radiación , Carcinoma de Pulmón de Células no Pequeñas/enzimología , Carcinoma de Pulmón de Células no Pequeñas/patología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Transferencia Lineal de Energía , Sistema de Señalización de MAP Quinasas/efectos de la radiación , Proteómica , Apoptosis/efectos de los fármacos , Caspasa 3/metabolismo , Línea Celular Tumoral , Biología Computacional , Activación Enzimática/efectos de los fármacos , Activación Enzimática/efectos de la radiación , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/patología , Inhibidores de Proteínas Quinasas/farmacología , Reproducibilidad de los Resultados , Factores de Tiempo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
11.
Sci Rep ; 10(1): 5923, 2020 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-32246062

RESUMEN

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.


Asunto(s)
Benzamidas/farmacología , Quimioradioterapia/métodos , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Isoindoles/farmacología , Neoplasias/terapia , Animales , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Benzamidas/uso terapéutico , Movimiento Celular/efectos de los fármacos , Movimiento Celular/efectos de la radiación , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Roturas del ADN de Doble Cadena/efectos de los fármacos , Roturas del ADN de Doble Cadena/efectos de la radiación , Reparación del ADN/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Células HCT116 , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Isoindoles/uso terapéutico , Ratones , Neoplasias/patología , Tolerancia a Radiación/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Int J Oncol ; 55(6): 1287-1295, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31638190

RESUMEN

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.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Benzamidas/farmacología , Quimioradioterapia/métodos , Complejos de Coordinación/farmacología , Isoindoles/farmacología , Tumores Neuroendocrinos/terapia , Octreótido/análogos & derivados , Fármacos Sensibilizantes a Radiaciones/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Benzamidas/uso terapéutico , Línea Celular Tumoral , Complejos de Coordinación/uso terapéutico , Sinergismo Farmacológico , Receptores ErbB/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Humanos , Isoindoles/uso terapéutico , Tumores Neuroendocrinos/patología , Octreótido/farmacología , Octreótido/uso terapéutico , Fármacos Sensibilizantes a Radiaciones/uso terapéutico , Receptores de Somatostatina/agonistas , Esferoides Celulares
13.
Int J Oncol ; 33(2): 341-9, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18636155

RESUMEN

Effects of radiation on growth of two human tumour cell lines that survived a previous high dose, low dose-rate radionuclide exposure simulating intensive radionuclide therapy, were analyzed. The purpose was to investigate whether the survivors gained therapy induced changes in growth and radiation response. The U118MG, ParRes (parental resistant), and U373MG, ParSen (parental sensitive), glioma cells were used because they are known to be low dose-rate radiation resistant and sensitive, respectively. These cells were initially exposed to high dose, low dose-rate radiation for 24 h and surviving U118MG and U373MG cells formed new cultures called SurRes (surviving resistant) and SurSen (surviving sensitive), respectively. All four cell types were then exposed to graded acute radiation doses, 0-8 Gy, and analyzed for radiation induced growth disturbances. They were also analyzed regarding DNA-content and cell cycle distributions. The SurRes cells regained in most cases the same growth rate, had the same growth delays and showed generally a similar response as the original ParRes cells to the 0-8 Gy exposures. In contrast, the SurSen cells had in all cases slower growth rate and longer growth delays than the original ParSen cells after the 0-8 Gy exposures. There were no signs of radiation-induced radioresistance. The slow growing SurSen cells contained about 80% more DNA and had more cells in G1 and fewer in G2 than the ParSen cells. The conclusion is that tumour cells surviving high dose, low dose-rate, radionuclide therapy, afterwards can react differently to a new radiation exposure.


Asunto(s)
Proliferación Celular/efectos de la radiación , Neoplasias/radioterapia , Tolerancia a Radiación/fisiología , Apoptosis/efectos de la radiación , Línea Celular Tumoral , Relación Dosis-Respuesta en la Radiación , Humanos
14.
Radiat Res ; 169(5): 506-12, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18439038

RESUMEN

Ionizing radiation induces a variety of different DNA lesions; in addition to the most critical DNA damage, the DSB, numerous base alterations, SSBs and other modifications of the DNA double-helix are formed. When several non-DSB lesions are clustered within a short distance along DNA, or close to a DSB, they may interfere with the repair of DSBs and affect the measurement of DSB induction and repair. We have shown previously that a substantial fraction of DSBs measured by pulsed-field gel electrophoresis (PFGE) are in fact due to heat-labile sites within clustered lesions, thus reflecting an artifact of preparation of genomic DNA at elevated temperature. To further characterize the influence of heat-labile sites on DSB induction and repair, cells of four human cell lines (GM5758, GM7166, M059K, U-1810) with apparently normal DSB rejoining were tested for biphasic rejoining after gamma irradiation. When heat-released DSBs were excluded from the measurements, the fraction of fast rejoining decreased to less than 50% of the total. However, the half-times of the fast (t(1/2) = 7-8 min) and slow (t(1/2) = 2.5 h) DSB rejoining were not changed significantly. At t = 0, the heat-released DSBs accounted for almost 40% of the DSBs, corresponding to 10 extra DSBs per cell per Gy in the initial DSB yield. These heat-released DSBs were repaired within 60-90 min in all cells tested, including M059K cells treated with wortmannin and DNA-PKcs-defective M059J cells. Furthermore, cells lacking XRCC1 or poly(ADP-ribose) polymerase 1 (PARP1) rejoined both total DSBs and heat-released DSBs similarly to normal cells. In summary, the presence of heat-labile sites has a substantial impact on DSB induction and DSB rejoining rates measured by pulsed-field gel electrophoresis, and heat-labile sites repair is independent of DNA-PKcs, XRCC1 and PARP.


Asunto(s)
Reparación del ADN/efectos de la radiación , Proteínas de Unión al ADN/metabolismo , ADN/metabolismo , Calor , Poli(ADP-Ribosa) Polimerasas/metabolismo , Línea Celular , Humanos , Factores de Tiempo , Proteína 1 de Reparación por Escisión del Grupo de Complementación Cruzada de las Lesiones por Rayos X
15.
Mol Cancer Ther ; 6(8): 2303-9, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17699725

RESUMEN

Trifluoperazine (TFP), a member of the phenothiazine class of antipsychotic drugs, has been shown to augment the cytotoxicity of the DNA-damaging agent bleomycin. In the present study, we investigated the effect of trifluoperazine on (a) survival of bleomycin-treated human non-small cell lung carcinoma U1810 cells, (b) induction and repair of bleomycin-induced DNA strand breaks, and (c) nonhomologous end-joining (NHEJ), the major DNA double-strand break (DSB) repair pathway in mammalian cells. By using a clonogenic survival assay, we show here that concomitant administration of trifluoperazine at a subtoxic concentration enhances the cytotoxicity of bleomycin. Moreover, trifluoperazine also increases the longevity of bleomycin-induced DNA strand breaks in U1810 cells, as shown by both comet assay and fraction of activity released (FAR)-assay. This action seems to be related to suppression of cellular DNA DSB repair activities because NHEJ-mediated rejoining of DSBs occurs with significantly lower efficiency in the presence of trifluoperazine. We propose that TFP might be capable of inhibiting one or more elements of the DNA DSB repair machinery, thereby increasing the cytotoxicity of bleomycin in lung cancer cells.


Asunto(s)
Antipsicóticos/farmacología , Carcinoma de Pulmón de Células no Pequeñas/patología , Roturas del ADN de Doble Cadena/efectos de los fármacos , Reparación del ADN/efectos de los fármacos , Neoplasias Pulmonares/patología , Trifluoperazina/farmacología , Bleomicina/farmacología , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Sinergismo Farmacológico , Humanos , Recombinación Genética/efectos de los fármacos , Ensayo de Tumor de Célula Madre
16.
PLoS One ; 13(12): e0209594, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30592737

RESUMEN

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.


Asunto(s)
Roturas del ADN de Doble Cadena , Daño del ADN , Calor , Línea Celular Tumoral , Reparación del ADN , Electroforesis en Gel de Campo Pulsado , Humanos , Microscopía de Fuerza Atómica
17.
BMC Mol Biol ; 8: 97, 2007 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-17963495

RESUMEN

BACKGROUND: Efficient and correct repair of DNA damage, especially DNA double-strand breaks, is critical for cellular survival. Defects in the DNA repair may lead to cell death or genomic instability and development of cancer. Non-homologous end-joining (NHEJ) is the major repair pathway for DNA double-strand breaks in mammalian cells. The ability of other repair pathways, such as homologous recombination, to compensate for loss of NHEJ and the ways in which contributions of different pathways are regulated are far from fully understood. RESULTS: In this report we demonstrate that long single-stranded DNA (ssDNA) ends are formed at radiation-induced DNA double-strand breaks in NHEJ deficient cells. At repair times > or = 1 h, processing of unrejoined DNA double-strand breaks generated extensive ssDNA at the DNA ends in cells lacking the NHEJ protein complexes DNA-dependent protein kinase (DNA-PK) or DNA Ligase IV/XRCC4. The ssDNA formation was cell cycle dependent, since no ssDNA ends were observed in G1-synchronized NHEJ deficient cells. Furthermore, in wild type cells irradiated in the presence of DNA-PKcs (catalytic subunit of DNA-PK) inhibitors, or in DNA-PKcs deficient cells complemented with DNA-PKcs mutated in six autophosphorylation sites (ABCDE), no ssDNA was formed. The ssDNA generation also greatly influences DNA double-strand break quantification by pulsed-field gel electrophoresis, resulting in overestimation of the DNA double-strand break repair capability in NHEJ deficient cells when standard protocols for preparing naked DNA (i. e., lysis at 50 degrees C) are used. CONCLUSION: We provide evidence that DNA Ligase IV/XRCC4 recruitment by DNA-PK to DNA double-strand breaks prevents the formation of long ssDNA ends at double-strand breaks during the S phase, indicating that NHEJ components may downregulate an alternative repair process where ssDNA ends are required.


Asunto(s)
Roturas del ADN de Doble Cadena , Trastornos por Deficiencias en la Reparación del ADN/patología , ADN de Cadena Simple/biosíntesis , Fase S/genética , Animales , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , ADN/aislamiento & purificación , Roturas del ADN de Doble Cadena/efectos de la radiación , ADN Ligasa (ATP) , ADN Ligasas/metabolismo , Trastornos por Deficiencias en la Reparación del ADN/genética , ADN de Cadena Simple/metabolismo , ADN de Cadena Simple/efectos de la radiación , Proteína Quinasa Activada por ADN/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Humanos , Modelos Biológicos , Unión Proteica , Fase S/efectos de la radiación , Análisis de Secuencia de ADN , Temperatura
18.
Radiat Res ; 168(2): 175-82, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17638403

RESUMEN

In this study the induction of double-strand breaks (DSBs) was investigated in Chinese hamster V79-379A cells irradiated with the Auger-electron emitter (125)I incorporated into DNA. The role of chromatin organization was studied by pulse-labeling synchronized cells with (125)IdU before decay accumulation in early or late S phase. Pulsed-field gel electrophoresis and fragment-size analysis were used to quantify the distribution of DNA fragments in irradiated intact cells and naked DNA as well as in DNA from asynchronously labeled cultures in a different scavenging environment. The results show that in intact cells, after accumulation of decays at -70 degrees C in the presence of 10% DMSO, almost four times more DSBs were induced in late S phase compared with early S phase and the fragment distribution was clearly non-random with an excess of fragments <0.2 Mbp. The DSB yield was 0.6 DSB/cell and decay for cells irradiated in early S phase and 2.3 DSBs/cell and decay for cells irradiated in late S phase. When similar experiments were performed on naked genomic DNA or intact cells irradiated with gamma rays, the difference in yield was not as prominent. These data imply a role of chromatin organization in the induction of DSBs by DNA-incorporated (125)I. In summary, the results presented here suggest that the yield of DSBs as well as the fragment distribution induced by (125)IdU decay may vary significantly depending on the chromatin organization during S phase and the labeling procedure used.


Asunto(s)
Cromatina/química , Roturas del ADN de Doble Cadena/efectos de la radiación , Radioisótopos de Yodo , Animales , Línea Celular , Cricetinae , Cricetulus , Relación Dosis-Respuesta en la Radiación , Rayos gamma , Fase S
19.
Radiat Res ; 167(3): 312-8, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17316073

RESUMEN

The purpose of this study was to quantify and to determine the distribution of DNA double-strand breaks (DSBs) in human cells irradiated in vitro and to evaluate the relative biological effectiveness (RBE) of the alpha-particle emitter (211)At for DSB induction. The influence of the irradiation temperature on the induction of DSBs was also investigated. Human fibroblasts were irradiated as intact cells with alpha particles from (211)At, (60)Co gamma rays and X rays. The numbers and distributions of DSBs were determined by pulsed-field gel electrophoresis with fragment analysis for separation of DNA fragments in sizes 10 kbp-5.7 Mbp. A non-random distribution was found for DSB induction after irradiation with alpha particles from (211)At, while irradiation with low-LET radiation led to more random distributions. The RBEs for DSB induction were 2.1 and 3.1 for (60)Co gamma rays and X rays as the reference radiation, respectively. In the experiments studying temperature effects, nuclear monolayers were irradiated with (211)At alpha particles or (60)Co gamma rays at 2 degrees C or 37 degrees C and intact cells were irradiated with (211)At alpha particles at the same temperatures. The dose-modifying factor (DMF(temp)) for irradiation of nuclear monolayers at 37 degrees C compared with 2 degrees C was 1.7 for (211)At alpha particles and 1.6 for (60)Co gamma rays. No temperature effect was observed for intact cells irradiated with (211)At. In conclusion, irradiation with alpha particles from (211)At induced two to three times more DSB than gamma rays and X rays.


Asunto(s)
Partículas alfa , Astato/toxicidad , Roturas del ADN de Doble Cadena/efectos de la radiación , Daño del ADN/efectos de la radiación , Daño del ADN/genética , Fibroblastos/metabolismo , Fibroblastos/efectos de la radiación , Humanos , Temperatura
20.
Radiat Res ; 188(6): 597-604, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28952912

RESUMEN

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.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN por Unión de Extremidades , Proteína Quinasa Activada por ADN/fisiología , Citometría de Flujo , Proteínas Nucleares/fisiología , Procesamiento Proteico-Postraduccional , Línea Celular , Cromonas/farmacología , ADN/efectos de la radiación , Proteína Quinasa Activada por ADN/antagonistas & inhibidores , Relación Dosis-Respuesta en la Radiación , Electroforesis en Gel de Campo Pulsado , Rayos gamma , Histonas/fisiología , Humanos , Morfolinas/farmacología , Proteínas Nucleares/antagonistas & inhibidores , Fosforilación/efectos de los fármacos , Fosfotreonina/metabolismo , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA