Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
1.
Artículo en Inglés | MEDLINE | ID: mdl-38340776

RESUMEN

PURPOSE: The products of lipid peroxidation have been implicated in human diseases and aging. This prompted us to investigate the response to conventional (CONV) versus FLASH irradiation of oxylipins, a family of bioactive lipid metabolites derived from omega-3 or omega-6 polyunsaturated fatty acids through oxygen-dependent non-enzymatic as well as dioxygenase-mediated free radical reactions. METHODS AND MATERIALS: Ultrahigh performance liquid chromatography coupled to tandem mass spectrometry was used to quantify the expression of 37 oxylipins derived from eicosatetraenoic, eicosapentaenoic and docosahexaenoic acid in mouse lung and in normal or cancer cells exposed to either radiation modality under precise monitoring of the temperature and oxygenation. Among the 37 isomers assayed, 14-16 were present in high enough amount to enable quantitative analysis. The endpoints were the expression of oxylipins as a function of the dose of radiation, normoxia versus hypoxia, temperature and post-irradiation time. RESULTS: In normal, normoxic cells at 37°C radiation elicited destruction and neosynthesis of oxylipins acting antagonistically on a background subject to rapid remodeling by oxygenases. Neosynthesis was observed in the CONV mode only, in such a way that the level of oxylipins at 5 minutes after FLASH irradiation was 20-50% lower than in non-irradiated and CONV-irradiated cells. Hypoxia mitigated the differential CONV versus FLASH response in some oxylipins. These patterns were not reproduced in tumor cells. Depression of specific oxylipins following FLASH irradiation was observed in mouse lung at 5 min following irradiation, with near complete recovery in 24 hours and further remodeling at one week and two months post-irradiation. CONCLUSIONS: Down-regulation of oxylipins was a hallmark of FLASH irradiation specific of normal cells. Temperature effects suggest that this process occurs via diffusion-controlled, bimolecular recombination of a primary radical species upstream from peroxyl radical formation and evoke a major role of the membrane composition and fluidity in response to the FLASH modality.

2.
Nat Commun ; 14(1): 2445, 2023 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-37117166

RESUMEN

Radiation Induced Lung Injury (RILI) is one of the main limiting factors of thorax irradiation, which can induce acute pneumonitis as well as pulmonary fibrosis, the latter being a life-threatening condition. The order of cellular and molecular events in the progression towards fibrosis is key to the physiopathogenesis of the disease, yet their coordination in space and time remains largely unexplored. Here, we present an interactive murine single cell atlas of the lung response to irradiation, generated from C57BL6/J female mice. This tool opens the door for exploration of the spatio-temporal dynamics of the mechanisms that lead to radiation-induced pulmonary fibrosis. It depicts with unprecedented detail cell type-specific radiation-induced responses associated with either lung regeneration or the failure thereof. A better understanding of the mechanisms leading to lung fibrosis will help finding new therapeutic options that could improve patients' quality of life.


Asunto(s)
Lesión Pulmonar , Fibrosis Pulmonar , Traumatismos por Radiación , Neumonitis por Radiación , Femenino , Animales , Ratones , Fibrosis Pulmonar/etiología , Fibrosis Pulmonar/patología , Neumonitis por Radiación/etiología , Neumonitis por Radiación/patología , Calidad de Vida , Pulmón/patología , Lesión Pulmonar/etiología , Lesión Pulmonar/patología , Tórax
3.
Int J Radiat Oncol Biol Phys ; 113(5): 985-995, 2022 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-35227789

RESUMEN

PURPOSE: For many years, the effect of dose rate (DR) was considered negligible in external beam radiation therapy (EBRT) until very-high DR (>10 Gy/min) became possible and ultrahigh DR (>40 Gy/s) showed dramatic protection of normal tissues in preclinical experiments. We propose a critical review of preclinical and clinical studies to investigate the biological and clinical effects of DR variation in the range covering brachytherapy to flattening filter free EBRT and FLASH. METHODS AND MATERIALS: Preclinical and clinical studies investigating biological and clinical DR effects were reviewed extensively. We also conducted an in silico study to assess the effect of pulse DR (DRp), taking into account the mean time between 2 tracks during the pulse. RESULTS: Preclinical studies have shown that an increase in DR in the range of 0.01 to 20 Gy/min (not including ultralow or ultrahigh DR) resulted in decreased survival of both normal and tumor cells. This effect was attributed primarily to increasingly unrepaired "sublethal" DNA damage with increasing the DR. However, the models and irradiation conditions have often been very different from one radiobiological study to another. Moreover, the physical parameters on the spatial and temporal microstructure of the beam were not considered systematically. In particular, the DRp was rarely mentioned. The in silico studies showed that for the same average DR, increasing DRp induced an increase of mean track rates. These results could explain the presence of more complex damage when the DRp was increased within the range of DR considered, in relation to the time-dependent probability of accumulating unrepaired, "sublethal" DNA lesions in close proximity. CONCLUSIONS: Knowledge of the beam microstructure is critical to understanding the biological impact and the clinical outcomes of radiation at the DR commonly used in radiation therapy.


Asunto(s)
Braquiterapia , Braquiterapia/métodos , Humanos
4.
Med Phys ; 49(3): 2068-2081, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-34407219

RESUMEN

Current radiotherapy facilities are standardized to deliver dose rates around 0.1-0.4 Gy/s in 2 Gy daily fractions, designed to deliver total accumulated doses to reach the tolerance limit of normal tissues undergoing irradiation. FLASH radiotherapy (FLASH-RT), on the other hand, relies on facilities capable of delivering ultrahigh dose rates in large doses in a single microsecond pulse, or in a few pulses given over a very short time sequence. For example, most studies to date have implemented 4-6 MeV electrons with intra-pulse dose rates in the range 106 -107  Gy/s. The proposed dependence of the FLASH effect on oxygen tension has stimulated several theoretical models based on three different hypotheses: (i) Radiation-induced transient oxygen depletion; (ii) cell-specific differences in the ability to detoxify and/or recover from injury caused by reactive oxygen species; (iii) self-annihilation of radicals by bimolecular recombination. This article focuses on the observations supporting or refuting these models in the frame of the chemical-biological bases of the impact of oxygen on the radiation response of cell free, in vitro and in vivo model systems.


Asunto(s)
Oxígeno , Oncología por Radiación , Electrones , Dosificación Radioterapéutica
5.
Med Phys ; 49(3): 1993-2013, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-34426981

RESUMEN

Radiation exposures at ultrahigh dose rates (UHDRs) at several orders of magnitude greater than in current clinical radiotherapy (RT) have been shown to manifest differential radiobiological responses compared to conventional (CONV) dose rates. This has led to studies investigating the application of UHDR for therapeutic advantage (FLASH-RT) that have gained significant interest since the initial discovery in 2014 that demonstrated reduced lung toxicity with equivalent levels of tumor control compared with conventional dose-rate RT. Many subsequent studies have demonstrated the potential protective role of FLASH-RT in normal tissues, yet the underlying molecular and cellular mechanisms of the FLASH effect remain to be fully elucidated. Here, we summarize the current evidence of the FLASH effect and review FLASH-RT studies performed in preclinical models of normal tissue response. To critically examine the underlying biological mechanisms of responses to UHDR radiation exposures, we evaluate in vitro studies performed with normal and tumor cells. Differential responses to UHDR versus CONV irradiation recurrently involve reduced inflammatory processes and differential expression of pro- and anti-inflammatory genes. In addition, frequently reduced levels of DNA damage or misrepair products are seen after UHDR irradiation. So far, it is not clear what signal elicits these differential responses, but there are indications for involvement of reactive species. Different susceptibility to FLASH effects observed between normal and tumor cells may result from altered metabolic and detoxification pathways and/or repair pathways used by tumor cells. We summarize the current theories that may explain the FLASH effect and highlight important research questions that are key to a better mechanistic understanding and, thus, the future implementation of FLASH-RT in the clinic.


Asunto(s)
Neoplasias , Oncología por Radiación , Protocolos Clínicos , Humanos , Neoplasias/radioterapia , Radiobiología , Dosificación Radioterapéutica
6.
Cancers (Basel) ; 13(19)2021 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-34638424

RESUMEN

The development of innovative approaches that would reduce the sensitivity of healthy tissues to irradiation while maintaining the efficacy of the treatment on the tumor is of crucial importance for the progress of the efficacy of radiotherapy. Recent methodological developments and innovations, such as scanned beams, ultra-high dose rates, and very high-energy electrons, which may be simultaneously available on new accelerators, would allow for possible radiobiological advantages of very short pulses of ultra-high dose rate (FLASH) therapy for radiation therapy to be considered. In particular, very high-energy electron (VHEE) radiotherapy, in the energy range of 100 to 250 MeV, first proposed in the 2000s, would be particularly interesting both from a ballistic and biological point of view for the establishment of this new type of irradiation technique. In this review, we examine and summarize the current knowledge on VHEE radiotherapy and provide a synthesis of the studies that have been published on various experimental and simulation works. We will also consider the potential for VHEE therapy to be translated into clinical contexts.

7.
Radiother Oncol ; 153: 303-310, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32534957

RESUMEN

BACKGROUND AND PURPOSE: FLASH radiotherapy, a technique based on delivering large doses in a single fraction at the micro/millisecond timescale, spares normal tissues from late radiation-induced toxicity, in an oxygen-dependent process, whilst keeping full anti-tumor efficiency. We present a theoretical model taking into account the kinetics of formation and decay of reactive oxygen species, in particular of organic peroxyl radicals ROO. formed by addition of O2 to primary carbon-centred radicals R. and known to play a major role at the origin radio-induced complications. MATERIALS AND METHODS: The model focuses on the time-dependent evolution of radiolytic products in living matter exposed to continuous irradiation at dose-rates in the range 10-3-107Gy·s-1. The 9 differential rate equations resulting from the radiolytic and enzymatic reactions network were solved using the published values of these reactions rate constants in a cellular environment. RESULTS: The model suggests a correlation between the area-under-the-curve of time-evolving [ROO.] and the probability of normal tissue complications. The model does not lend weight to the hypothesis of transient oxygen depletion as a main determinant of FLASH but rather suggests a major role of radical-radical recombination. CONCLUSION: The model gives support to the reduction of ROO. lifetime as the main root of FLASH and compares favorably with published experimental results. We conclude that any process - in this case radical recombination - that shortens the lifetime or limits the radiolytic yield of ROO. is likely to protect normoxic tissues against the deleterious effects of radiation.


Asunto(s)
Oxígeno , Peróxidos , Humanos , Cinética , Recombinación Genética
8.
Phys Med Biol ; 65(21): 21RM02, 2020 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-32380492

RESUMEN

This roadmap outlines the potential roles of metallic nanoparticles (MNPs) in the field of radiation therapy. MNPs made up of a wide range of materials (from Titanium, Z = 22, to Bismuth, Z = 83) and a similarly wide spectrum of potential clinical applications, including diagnostic, therapeutic (radiation dose enhancers, hyperthermia inducers, drug delivery vehicles, vaccine adjuvants, photosensitizers, enhancers of immunotherapy) and theranostic (combining both diagnostic and therapeutic), are being fabricated and evaluated. This roadmap covers contributions from experts in these topics summarizing their view of the current status and challenges, as well as expected advancements in technology to address these challenges.


Asunto(s)
Nanopartículas del Metal/uso terapéutico , Nanomedicina Teranóstica/métodos , Humanos , Hipertermia Inducida
9.
Br J Radiol ; 93(1107): 20190807, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32003574

RESUMEN

After years of lethargy, studies on two non-conventional microstructures in time and space of the beams used in radiation therapy are enjoying a huge revival. The first effect called "FLASH" is based on very high dose-rate irradiation (pulse amplitude ≥106 Gy/s), short beam-on times (≤100 ms) and large single doses (≥10 Gy) as experimental parameters established so far to give biological and potential clinical effects. The second effect relies on the use of arrays of minibeams (e.g., 0.5-1 mm, spaced 1-3.5 mm). Both approaches have been shown to protect healthy tissues as an endpoint that must be clearly specified and could be combined with each other (e.g., minibeams under FLASH conditions). FLASH depends on the presence of oxygen and could proceed from the chemistry of peroxyradicals and a reduced incidence on DNA and membrane damage. Minibeams action could be based on abscopal effects, cell signalling and/or migration of cells between "valleys and hills" present in the non-uniform irradiation field as well as faster repair of vascular damage. Both effects are expected to maintain intact the tumour control probability and might even preserve antitumoural immunological reactions. FLASH in vivo experiments involving Zebrafish, mice, pig and cats have been done with electron beams, while minibeams are an intermediate approach between X-GRID and synchrotron X-ray microbeams radiation. Both have an excellent rationale to converge and be applied with proton beams, combining focusing properties and high dose rates in the beam path of pencil beams, and the inherent advantage of a controlled limited range. A first treatment with electron FLASH (cutaneous lymphoma) has recently been achieved, but clinical trials have neither been presented for FLASH with protons, nor under the minibeam conditions. Better understanding of physical, chemical and biological mechanisms of both effects is essential to optimize the technical developments and devise clinical trials.


Asunto(s)
Tratamientos Conservadores del Órgano/métodos , Terapia de Protones/métodos , Animales , Gatos , Proliferación Celular , Daño del ADN , Reparación del ADN , Fraccionamiento de la Dosis de Radiación , Linfoma Cutáneo de Células T/radioterapia , Ratones , Órganos en Riesgo/efectos de la radiación , Oxígeno , Consumo de Oxígeno , Traumatismos por Radiación/prevención & control , Tolerancia a Radiación , Radiometría/métodos , Neoplasias Cutáneas/radioterapia , Análisis Espacio-Temporal , Porcinos , Pez Cebra
10.
Clin Cancer Res ; 26(6): 1497-1506, 2020 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-31796518

RESUMEN

PURPOSE: One of the main limitations to anticancer radiotherapy lies in irreversible damage to healthy tissues located within the radiation field. "FLASH" irradiation at very high dose-rate is a new treatment modality that has been reported to specifically spare normal tissue from late radiation-induced toxicity in animal models and therefore could be a promising strategy to reduce treatment toxicity. EXPERIMENTAL DESIGN: Lung responses to FLASH irradiation were investigated by qPCR, single-cell RNA sequencing (sc-RNA-Seq), and histologic methods during the acute wound healing phase as well as at late stages using C57BL/6J wild-type and Terc-/- mice exposed to bilateral thorax irradiation as well as human lung cells grown in vitro. RESULTS: In vitro studies gave evidence of a reduced level of DNA damage and induced lethality at the advantage of FLASH. In mouse lung, sc-RNA-seq and the monitoring of proliferating cells revealed that FLASH minimized the induction of proinflammatory genes and reduced the proliferation of progenitor cells after injury. At late stages, FLASH-irradiated lungs presented less persistent DNA damage and senescent cells than after CONV exposure, suggesting a higher potential for lung regeneration with FLASH. Consistent with this hypothesis, the beneficial effect of FLASH was lost in Terc-/- mice harboring critically short telomeres and lack of telomerase activity. CONCLUSIONS: The results suggest that, compared with conventional radiotherapy, FLASH minimizes DNA damage in normal cells, spares lung progenitor cells from excessive damage, and reduces the risk of replicative senescence.


Asunto(s)
Senescencia Celular/efectos de la radiación , Pulmón/efectos de la radiación , ARN/fisiología , Análisis de la Célula Individual/métodos , Células Madre/efectos de la radiación , Telomerasa/fisiología , Animales , Línea Celular Tumoral , Relación Dosis-Respuesta en la Radiación , Femenino , Humanos , Pulmón/metabolismo , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , RNA-Seq/métodos , Células Madre/metabolismo
11.
Phys Med ; 60: 50-57, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31000086

RESUMEN

PURPOSE: Measurements and Monte-Carlo simulations were carried out to model the dose distribution of a prototype electron beam linear accelerator (Kinetron LINAC) to determine the dose to organs in small animal irradiations experiments. Dose distributions were simulated using the GATE8.0/Geant4.10.3 Monte-Carlo platform, and measured in air and solid water phantoms using a commercial scintillating screen detector and new EBT-XD Gafchromic films. METHODS: The LINAC is able to produce 4.5 MeV electron beams at dose-rates ranging from Gy/min to thousands of Gy/s, and is used to study the radiobiological effects of very-high dose-rates that have been shown to protect normal tissues from radiation toxicity. Numerical simulations and experimental dosimetric characterisation of this electron accelerator were performed with the Monte-Carlo toolkit and various detectors. Absolute dose distributions in solid water were measured and compared with simulations. Realistic electron irradiation conditions were simulated in voxelised mice CT images. 3D dose distributions and dose-volume histograms in lungs of mice were simulated and analyzed. RESULTS: Measured and calculated depth-dose profiles for several beam configurations (energy and dose-rate) were compared. Beam emittance was validated by comparing measured and calculated beam sizes along the central axis in air: the deviation for all conditions was less than 1 mm. A good agreement was obtained between experimental dose distributions and the results obtained with simulations (<2% dose differences for lateral and depth-dose profiles). CONCLUSIONS: The method presented here, relying on few free parameters, can be adapted to very-high dose-rate electron irradiation to support the analysis of preclinical research experiments.


Asunto(s)
Aceleradores de Partículas , Animales , Simulación por Computador , Electrones , Diseño de Equipo , Pulmón/diagnóstico por imagen , Pulmón/efectos de la radiación , Ratones , Método de Montecarlo , Fantasmas de Imagen , Radiometría/instrumentación , Tomografía Computarizada por Rayos X , Agua
12.
Clin Cancer Res ; 25(1): 35-42, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29875213

RESUMEN

PURPOSE: Previous studies using FLASH radiotherapy (RT) in mice showed a marked increase of the differential effect between normal tissue and tumors. To stimulate clinical transfer, we evaluated whether this effect could also occur in higher mammals. EXPERIMENTAL DESIGN: Pig skin was used to investigate a potential difference in toxicity between irradiation delivered at an ultrahigh dose rate called "FLASH-RT" and irradiation delivered at a conventional dose rate called "Conv-RT." A clinical, phase I, single-dose escalation trial (25-41 Gy) was performed in 6 cat patients with locally advanced T2/T3N0M0 squamous cell carcinoma of the nasal planum to determine the maximal tolerated dose and progression-free survival (PFS) of single-dose FLASH-RT. RESULTS: Using, respectively, depilation and fibronecrosis as acute and late endpoints, a protective effect of FLASH-RT was observed (≥20% dose-equivalent difference vs. Conv-RT). Three cats experienced no acute toxicity, whereas 3 exhibited moderate/mild transient mucositis, and all cats had depilation. With a median follow-up of 13.5 months, the PFS at 16 months was 84%. CONCLUSIONS: Our results confirmed the potential advantage of FLASH-RT and provide a strong rationale for further evaluating FLASH-RT in human patients.See related commentary by Harrington, p. 3.


Asunto(s)
Carcinoma de Células Escamosas/radioterapia , Neoplasias Nasales/radioterapia , Radioterapia/métodos , Animales , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/veterinaria , Gatos , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Neoplasias Nasales/patología , Neoplasias Nasales/veterinaria , Radioterapia/efectos adversos , Dosificación Radioterapéutica , Porcinos , Porcinos Enanos
13.
Int J Radiat Oncol Biol Phys ; 102(3): 619-626, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30017793

RESUMEN

PURPOSE: Recent in vivo investigations have shown that short pulses of electrons at very high dose rates (FLASH) are less harmful to healthy tissues but just as efficient as conventional dose-rate radiation to inhibit tumor growth. In view of the potential clinical value of FLASH and the availability of modern proton therapy infrastructures to achieve this goal, we herein describe a series of technological developments required to investigate the biology of FLASH irradiation using a commercially available clinical proton therapy system. METHODS AND MATERIALS: Numerical simulations and experimental dosimetric characterization of a modified clinical proton beamline, upstream from the isocenter, were performed with a Monte Carlo toolkit and different detectors. A single scattering system was optimized with a ridge filter and a high current monitoring system. In addition, a submillimetric set-up protocol based on image guidance using a digital camera and an animal positioning system was also developed. RESULTS: The dosimetric properties of the resulting beam and monitoring system were characterized; linearity with dose rate and homogeneity for a 12 × 12 mm2 field size were assessed. Dose rates exceeding 40 Gy/s at energies between 138 and 198 MeV were obtained, enabling uniform irradiation for radiobiology investigations of small animals in a modified clinical proton beam line. CONCLUSIONS: This approach will enable us to conduct FLASH proton therapy experiments on small animals, specifically for mouse lung irradiation. Dose rates exceeding 40 Gy/s were achieved, which was not possible with the conventional clinical mode of the existing beamline.


Asunto(s)
Neoplasias/radioterapia , Terapia de Protones/instrumentación , Animales , Calibración , Simulación por Computador , Modelos Animales de Enfermedad , Diseño de Equipo , Pulmón/efectos de la radiación , Ratones , Método de Montecarlo , Protones , Radiobiología , Radiometría , Dosificación Radioterapéutica , Planificación de la Radioterapia Asistida por Computador
15.
Bull Cancer ; 104(4): 380-384, 2017 Apr.
Artículo en Francés | MEDLINE | ID: mdl-28283183

RESUMEN

In this review, we present the synthesis of the newly acquired knowledge concerning high dose-rate irradiations and the hopes that these new radiotherapy modalities give rise to. The results were presented at a recent symposium on the subject.


Asunto(s)
Neoplasias/radioterapia , Dosificación Radioterapéutica , Animales , Electrones/uso terapéutico , Humanos , Ratones , Terapia de Protones , Radioterapia/efectos adversos , Radioterapia/instrumentación , Radioterapia/métodos
16.
Target Oncol ; 11(3): 371-81, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26668065

RESUMEN

BACKGROUND: Pancreatic adenocarcinoma is characterized by a high frequency of KRAS mutations and frequent deregulation of the epidermal growth factor receptor (EGFR) and other EGFR family members such as HER2/ErbB2. The EGFR inhibitor erlotinib is approved for treatment of pancreatic cancer, but has shown modest activity in most patients. OBJECTIVE: Here we investigated the activity of afatinib, a second-generation irreversible pan-EGFR family kinase inhibitor, alone or in combination with ionizing radiation, toward pancreatic cancer cells. METHODS: The influence of afatinib on cell proliferation, cell cycle distribution, clonogenic survival, nuclear fragmentation, ploidy, and centrosome amplification following irradiation was determined. Expression and phosphorylation of HER receptors, Akt, DNA-PKcs, and ERK1/2 was characterized by Western blot analysis. RESULTS: Afatinib was growth-inhibitory for all three cell lines but cytotoxic only toward BxPC3 (KRAS (wt)) and Capan-2 (KRAS (mut)) cells, both of which express high levels of EGFR, HER2, and HER3 receptors. Afatinib increased the radiosensitivity of BxPC3 and Capan-2 cells, prevented the radio-induced phosphorylation of Akt, and induced mitotic catastrophe following irradiation. In comparison, Panc-1 cells (KRAS (mut)) expressing low levels of EGFR family receptors were resistant to afatinib-induced radiosensitization. LIMITATIONS: These results must be confirmed in vivo. CONCLUSIONS: Afatinib showed cytotoxic and radiosensitizing effects toward a subset of pancreatic cancer cells which was closely correlated with expression of EGFR, HER2, and HER3 receptors, but not with KRAS status.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/radioterapia , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/radioterapia , Quinazolinas/uso terapéutico , Fármacos Sensibilizantes a Radiaciones/uso terapéutico , Adenocarcinoma/patología , Afatinib , Línea Celular Tumoral , Proliferación Celular , Receptores ErbB , Humanos , Neoplasias Pancreáticas/patología , Quinazolinas/administración & dosificación , Quinazolinas/farmacología , Fármacos Sensibilizantes a Radiaciones/administración & dosificación , Fármacos Sensibilizantes a Radiaciones/farmacología , Transducción de Señal
18.
Cell Mol Life Sci ; 72(8): 1585-97, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25336152

RESUMEN

Poly(ADP-ribose) polymerase-2 (PARP-2) activity contributes to a cells' poly(ADP-ribosyl)ating potential and like PARP-1, has been implicated in several DNA repair pathways including base excision repair and DNA single strand break repair. Here the consequences of its stable depletion in HeLa, U20S, and AS3WT2 cells were examined. All three PARP-2 depleted models showed increased sensitivity to the cell killing effects on ionizing radiation as reported in PARP-2 depleted mouse embryonic fibroblasts providing further evidence for a role in DNA strand break repair. The PARP-2 depleted HeLa cells also showed both higher constitutive and DNA damage-induced levels of polymers of ADP-ribose (PAR) associated with unchanged PARP-1 protein levels, but higher PARP activity and a concomitant lower PARG protein levels and activity. These changes were accompanied by a reduced maximal recruitment of PARP-1, XRCC1, PCNA, and PARG to DNA damage sites. This PAR-associated phenotype could be reversed in HeLa cells on re-expression of PARP-2 and was not seen in U20S and AS3WT2 cells. These results highlight the complexity of the relationship between different members of the PARP family on PAR metabolism and suggest that cell model dependent phenotypes associated with the absence of PARP-2 exist within a common background of radiation sensitivity.


Asunto(s)
Poli Adenosina Difosfato Ribosa/metabolismo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Animales , Línea Celular , Supervivencia Celular/efectos de la radiación , Daño del ADN/efectos de la radiación , Reparación del ADN , Proteínas de Unión al ADN/metabolismo , Proteínas Activadoras de GTPasa/metabolismo , Células HeLa , Humanos , Ratones , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Poli(ADP-Ribosa) Polimerasas/genética , Antígeno Nuclear de Célula en Proliferación/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Radiación Ionizante , Proteína 1 de Reparación por Escisión del Grupo de Complementación Cruzada de las Lesiones por Rayos X
19.
Carcinogenesis ; 35(12): 2706-15, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25269805

RESUMEN

G-quadruplex (G4) structures in intron 3 of the p53 pre-mRNA modulate intron 2 splicing, altering the balance between the fully spliced p53 transcript (FSp53, encoding full-length p53) and an incompletely spliced transcript retaining intron 2 (p53I2 encoding the N-terminally truncated Δ40p53 isoform). The nucleotides forming G4s overlap the polymorphism rs17878362 (A1 wild-type allele, A2 16-base pair insertion) which is in linkage disequilibrium with rs1642785 in intron 2 (c.74+38 G>C). Biophysical and biochemical analyses show rs17878362 A2 alleles form similar G4 structures as A1 alleles although their position is shifted with respect to the intron 2 splice acceptor site. In addition basal FSp53 and p53I2 levels showed allele specific differences in both p53-null cells transfected with reporter constructs or lymphoblastoid cell lines. The highest FSp53 and p53I2 levels were associated with combined rs1642785-GG/rs17878362-A1A1 alleles, whereas the presence of rs1642785-C with either rs17878362 allele was associated with lower p53 pre-mRNA, total TP53, FSp53 and p53I2 levels, due to the lower stability of transcripts containing rs1642785-C. Treatment of lymphoblastoid cell with the G4 binding ligands 360A or PhenDC3 or with ionizing radiation increased FSp53 levels only in cells with rs17878362 A1 alleles, suggesting that under this G4 configuration full splicing is favoured. These results demonstrate the complex effects of intronic TP53 polymorphisms on G4 formation and identify a new role for rs1642785 on mRNA splicing and stability, and thus on the differential expression of isoform-specific transcripts of the TP53 gene.


Asunto(s)
Adenocarcinoma/genética , Empalme Alternativo/genética , Neoplasias de la Mama/genética , G-Cuádruplex/efectos de la radiación , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Intrones/genética , Polimorfismo Genético/genética , Proteína p53 Supresora de Tumor/genética , Adenocarcinoma/patología , Adenocarcinoma/radioterapia , Neoplasias de la Mama/patología , Neoplasias de la Mama/radioterapia , Exones , Femenino , Genotipo , Humanos , Isoformas de Proteínas , ARN Mensajero/genética , Radiación Ionizante , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas
20.
Int J Radiat Oncol Biol Phys ; 90(1): 36-43, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-25195988

RESUMEN

PURPOSE: Previously we showed that the relative biological efficiency for induced cell killing by the 76-MeV beam used at the Institut Curie Proton Therapy Center in Orsay increased with depth throughout the spread-out Bragg peak (SOBP). To investigate the repair pathways underlying this increase, we used an isogenic human cell model in which individual DNA repair proteins have been depleted, and techniques dedicated to precise measurements of radiation-induced DNA single-strand breaks (SSBs) and double-strand breaks (DSBs). METHODS AND MATERIALS: The 3-Gy surviving fractions of HeLa cells individually depleted of Ogg1, XRCC1, and PARP1 (the base excision repair/SSB repair pathway) or of ATM, DNA-PKcs, XRCC4, and Artemis (nonhomologous end-joining pathway) were determined at the 3 positions previously defined in the SOBP. Quantification of incident SSBs and DSBs by the alkaline elution technique and 3-dimensional (3D) immunofluorescence of γ-H2AX foci, respectively, was performed in SQ20 B cells. RESULTS: We showed that the amount of SSBs and DSBs depends directly on the particle fluence and that the increase in relative biological efficiency observed in the distal part of the SOBP is due to a subset of lesions generated under these conditions, leading to cell death via a pathway in which the Artemis protein plays a central role. CONCLUSIONS: Because therapies like proton or carbon beams are now being used to treat cancer, it is even more important to dissect the mechanisms implicated in the repair of the lesions generated by these particles. Additionally, alteration of the expression or activity of the Artemis protein could be a novel therapeutic tool before high linear energy transfer irradiation treatment.


Asunto(s)
Supervivencia Celular/efectos de la radiación , Roturas del ADN de Doble Cadena , Roturas del ADN de Cadena Simple , Reparación del ADN/fisiología , Proteínas Nucleares/fisiología , Protones , Efectividad Biológica Relativa , Proteínas de la Ataxia Telangiectasia Mutada/fisiología , Instituciones Oncológicas , Supervivencia Celular/fisiología , ADN Glicosilasas/fisiología , Proteína Quinasa Activada por ADN/fisiología , Proteínas de Unión al ADN/fisiología , Endonucleasas , Francia , Células HeLa , Histonas/análisis , Humanos , Método de Montecarlo , Proteínas Nucleares/deficiencia , Poli(ADP-Ribosa) Polimerasa-1 , Poli(ADP-Ribosa) Polimerasas/fisiología , Terapia de Protones , Proteína 1 de Reparación por Escisión del Grupo de Complementación Cruzada de las Lesiones por Rayos X
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...