Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 157
Filtrar
1.
Radiother Oncol ; 196: 110326, 2024 07.
Artículo en Inglés | MEDLINE | ID: mdl-38735536

RESUMEN

PURPOSE: The oxygen depletion hypothesis has been proposed as a rationale to explain the observed phenomenon of FLASH-radiotherapy (FLASH-RT) sparing normal tissues while simultaneously maintaining tumor control. In this study we examined the distribution of DNA Damage Response (DDR) markers in irradiated 3D multicellular spheroids to explore the relationship between FLASH-RT protection and radiolytic-oxygen-consumption (ROC) in tissues. METHODS: Studies were performed using a Varian Truebeam linear accelerator delivering 10 MeV electrons with an average dose rate above 50 Gy/s. Irradiations were carried out on 3D spheroids maintained under a range of O2 and temperature conditions to control O2 consumption and create gradients representative of in vivo tissues. RESULTS: Staining for pDNA-PK (Ser2056) produced a linear radiation dose response whereas γH2AX (Ser139) showed saturation with increasing dose. Using the pDNA-PK staining, radiation response was then characterised for FLASH compared to standard-dose-rates as a function of depth into the spheroids. At 4 °C, chosen to minimize the development of metabolic oxygen gradients within the tissues, FLASH protection could be observed at all distances under oxygen conditions of 0.3-1 % O2. Whereas at 37 °C a FLASH-protective effect was limited to the outer cell layers of tissues, an effect only observed at 3 % O2. Modelling of changes in the pDNA-PK-based oxygen enhancement ratio (OER) yielded a tissue ROC g0-value estimate of 0.73 ± 0.25 µM/Gy with a km of 5.4 µM at FLASH dose rates. CONCLUSIONS: DNA damage response markers are sensitive to the effects of transient oxygen depletion during FLASH radiotherapy. Findings support the rationale that well-oxygenated tissues would benefit more from FLASH-dose-rate protection relative to poorly-oxygenated tissues.


Asunto(s)
Daño del ADN , Esferoides Celulares , Daño del ADN/efectos de la radiación , Humanos , Esferoides Celulares/efectos de la radiación , Histonas/metabolismo , Histonas/análisis , Consumo de Oxígeno/efectos de la radiación , Relación Dosis-Respuesta en la Radiación , Tratamientos Conservadores del Órgano/métodos
2.
J Biophotonics ; 17(6): e202300552, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38494760

RESUMEN

The multifaceted nature of photodynamic therapy (PDT) requires a throughout evaluation of a multitude of parameters when devising preclinical protocols. In this study, we constructed MCF-7 human breast tumor spheroid assays to infer PDT irradiation doses at four gradient levels for violet light at 408 nm and red light at 625 nm under normal and hypoxic oxygen conditions. The compacted three-dimensional (3D) tumor models conferred PDT resistance as compared to monolayer cultures due to heterogenous distribution of photosensitizers along with the presence of internal hypoxic region. Cell viability results indicated that the violet light was more efficient to kill cells in the spheroids under normal oxygen conditions, while cells exposed to the hypoxic microenvironment exhibited minimal PDT-induced death. The combination of 3D tumor spheroid assays and the multiparametric screening platform presented a solid framework for assessing PDT efficacy across a wide range of different physiological conditions and therapeutic regimes.


Asunto(s)
Luz , Fotoquimioterapia , Esferoides Celulares , Humanos , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/patología , Esferoides Celulares/efectos de la radiación , Células MCF-7 , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Fármacos Fotosensibilizantes/farmacología , Gases/farmacología , Gases/química , Radiometría , Hipoxia de la Célula/efectos de los fármacos
3.
Radiat Oncol ; 16(1): 159, 2021 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-34412654

RESUMEN

BACKGROUND: Invasiveness is a major factor contributing to metastasis of tumour cells. Given the broad variety and plasticity of invasion mechanisms, assessing potential metastasis-promoting effects of irradiation for specific mechanisms is important for further understanding of potential adverse effects of radiotherapy. In fibroblast-led invasion mechanisms, fibroblasts produce tracks in the extracellular matrix in which cancer cells with epithelial traits can follow. So far, the influence of irradiation on this type of invasion mechanisms has not been assessed. METHODS: By matrix-embedding coculture spheroids consisting of breast cancer cells (MCF-7, BT474) and normal fibroblasts, we established a model for fibroblast-led invasion. To demonstrate applicability of this model, spheroid growth and invasion behaviour after irradiation with 5 Gy were investigated by microscopy and image analysis. RESULTS: When not embedded, irradiation caused a significant growth delay in the spheroids. When irradiating the spheroids with 5 Gy before embedding, we find comparable maximum migration distance in fibroblast monoculture and in coculture samples as seen in unirradiated samples. Depending on the fibroblast strain, the number of invading cells remained constant or was reduced. CONCLUSION: In this spheroid model and with the cell lines and fibroblast strains used, irradiation does not have a major invasion-promoting effect. 3D analysis of invasiveness allows to uncouple effects on invading cell number and maximum invasion distance when assessing radiation effects.


Asunto(s)
Neoplasias de la Mama/radioterapia , Fibroblastos/fisiología , Esferoides Celulares/efectos de la radiación , Neoplasias de la Mama/patología , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Femenino , Humanos , Invasividad Neoplásica , Esferoides Celulares/patología
4.
Sci Rep ; 11(1): 14192, 2021 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-34262055

RESUMEN

X-ray irradiation of high Z elements causes photoelectric effects that include the release of Auger electrons that can induce localized DNA breaks. We have previously established a tumor spheroid-based assay that used gadolinium containing mesoporous silica nanoparticles and synchrotron-generated monochromatic X-rays. In this work, we focused on iodine and synthesized iodine-containing porous organosilica (IPO) nanoparticles. IPO were loaded onto tumor spheroids and the spheroids were irradiated with 33.2 keV monochromatic X-ray. After incubation in CO2 incubator, destruction of tumor spheroids was observed which was accompanied by apoptosis induction, as determined by the TUNEL assay. By employing the γH2AX assay, we detected double strand DNA cleavages immediately after the irradiation. These results suggest that IPO first generate double strand DNA breaks upon X-ray irradiation followed by apoptosis induction of cancer cells. Use of three different monochromatic X-rays having energy levels of 33.0, 33.2 and 33.4 keV as well as X-rays with 0.1 keV energy intervals showed that the optimum effect of all three events (spheroid destruction, apoptosis induction and generation of double strand DNA breaks) occurred with a 33.2 keV monochromatic X-ray. These results uncover the preferential effect of K-edge energy X-ray for tumor spheroid destruction mediated by iodine containing nanoparticles.


Asunto(s)
Roturas del ADN/efectos de la radiación , Yodo/química , Nanopartículas/química , Neoplasias/patología , Compuestos Orgánicos/química , Dióxido de Silicio/química , Esferoides Celulares/efectos de la radiación , Apoptosis/efectos de la radiación , Línea Celular Tumoral , Roturas del ADN de Doble Cadena/efectos de la radiación , Humanos , Nanopartículas/ultraestructura , Compuestos Orgánicos/síntesis química , Porosidad , Dióxido de Silicio/síntesis química , Rayos X
5.
Int J Mol Sci ; 22(9)2021 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-34062920

RESUMEN

Radioligand therapy targeting the prostate-specific membrane antigen (PSMA) is rapidly evolving as a promising treatment for metastatic castration-resistant prostate cancer. The PSMA-targeting ligand p-SCN-Bn-TCMC-PSMA (NG001) labelled with 212Pb efficiently targets PSMA-positive cells in vitro and in vivo. The aim of this preclinical study was to evaluate the therapeutic potential of 212Pb-NG001 in multicellular tumour spheroid and mouse models of prostate cancer. The cytotoxic effect of 212Pb-NG001 was tested in human prostate C4-2 spheroids. Biodistribution at various time points and therapeutic effects of different activities of the radioligand were investigated in male athymic nude mice bearing C4-2 tumours, while long-term toxicity was studied in immunocompetent BALB/c mice. The radioligand induced a selective cytotoxic effect in spheroids at activity concentrations of 3-10 kBq/mL. In mice, the radioligand accumulated rapidly in tumours and was retained over 24 h, while it rapidly cleared from nontargeted tissues. Treatment with 0.25, 0.30 or 0.40 MBq of 212Pb-NG001 significantly inhibited tumour growth and improved median survival with therapeutic indexes of 1.5, 2.3 and 2.7, respectively. In BALB/c mice, no signs of long-term radiation toxicity were observed at activities of 0.05 and 0.33 MBq. The obtained results warrant clinical studies to evaluate the biodistribution, therapeutic efficacy and toxicity of 212Pb-NG001.


Asunto(s)
Neoplasias de la Próstata Resistentes a la Castración/radioterapia , Ensayo de Unión Radioligante , Radiofármacos/farmacología , Esferoides Celulares/efectos de la radiación , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Xenoinjertos , Humanos , Plomo/farmacología , Ligandos , Masculino , Ratones , Metástasis de la Neoplasia , Neoplasias de la Próstata Resistentes a la Castración/patología , Radioisótopos/farmacología , Esferoides Celulares/patología , Distribución Tisular/efectos de la radiación
6.
Int J Radiat Oncol Biol Phys ; 110(3): 833-844, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-33545301

RESUMEN

PURPOSE: The differential response of normal and tumor tissues to ultrahigh-dose-rate radiation (FLASH) has raised new hope for treating solid tumors but, to date, the mechanism remains elusive. One leading hypothesis is that FLASH radiochemically depletes oxygen from irradiated tissues faster than it is replenished through diffusion. The purpose of this study was to investigate these effects within hypoxic multicellular tumor spheroids through simulations and experiments. METHODS AND MATERIALS: Physicobiological equations were derived to model (1) the diffusion and metabolism of oxygen within spheroids; (2) its depletion through reactions involving radiation-induced radicals; and (3) the increase in radioresistance of spheroids, modeled according to the classical oxygen enhancement ratio and linear-quadratic response. These predictions were then tested experimentally in A549 spheroids exposed to electron irradiation at conventional (0.075 Gy/s) or FLASH (90 Gy/s) dose rates. Clonogenic survival, cell viability, and spheroid growth were scored postradiation. Clonogenic survival of 2 other cell lines was also investigated. RESULTS: The existence of a hypoxic core in unirradiated tumor spheroids is predicted by simulations and visualized by fluorescence microscopy. Upon FLASH irradiation, this hypoxic core transiently expands, engulfing a large number of well-oxygenated cells. In contrast, oxygen is steadily replenished during slower conventional irradiation. Experimentally, clonogenic survival was around 3-fold higher in FLASH-irradiated spheroids compared with conventional irradiation, but no significant difference was observed for well-oxygenated 2-dimensional cultured cells. This differential survival is consistent with the predictions of the computational model. FLASH irradiation of spheroids resulted in a dose-modifying factor of around 1.3 for doses above 10 Gy. CONCLUSIONS: Tumor spheroids can be used as a model to study FLASH irradiation in vitro. The improved survival of tumor spheroids receiving FLASH radiation confirms that ultrafast radiochemical oxygen depletion and its slow replenishment are critical components of the FLASH effect.


Asunto(s)
Modelos Biológicos , Oxígeno/metabolismo , Esferoides Celulares/metabolismo , Esferoides Celulares/efectos de la radiación , Humanos , Lipoproteínas
7.
Biochem Biophys Res Commun ; 546: 150-154, 2021 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-33582558

RESUMEN

In this study, we examined the phenotypes of CD133-positive cells that were induced in a hypoxic microenvironment of spheroids formed using a glioblastoma cell line (T98G). Colony-formation assay showed that spheroid CD133-positive cells (SCPCs) were more resistant to X-rays and Temozolomide (TMZ) than spheroid CD133-negative cells (SCNCs) sorted from T98G spheroids. In contrast, the sensitivity to X-rays and TMZ was not different between hypoxic cells and normoxic cells of T98G spheroids in a colony-formation assay using green fluorescent protein (GFP) reporter-transfectants to monitor hypoxia. This result suggests that the difference in the sensitivity to X-rays and TMZ between SCPCs and SCNCs did not result from hypoxia. Transwell membrane assay indicated that the migration and inversion ability of SCPCs was higher than that of SCNCs. These results, including the findings obtained previously regarding nestin positivity in SCPCs, strongly suggest that SCPCs are cancer stem cell (CSC)-like cells. Additionally, based on experiments of monolayer culture of T98G cells, it was shown that hypoxia or low pH culture condition is not sufficient for the induction of SCPCs. The three-dimensional cell structure might be a critical factor for SCPC induction.


Asunto(s)
Hipoxia de la Célula , Glioblastoma/patología , Modelos Biológicos , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Esferoides Celulares/metabolismo , Esferoides Celulares/patología , Microambiente Tumoral , Antígeno AC133/metabolismo , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Movimiento Celular , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Citometría de Flujo , Genes Reporteros , Humanos , Concentración de Iones de Hidrógeno , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Células Madre Neoplásicas/efectos de los fármacos , Fenotipo , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/efectos de la radiación , Temozolomida/farmacología , Rayos X
8.
Int J Radiat Biol ; 97(5): 695-703, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33617430

RESUMEN

PURPOSE: The present study investigated the biological effects of spot scanning and passive scattering proton therapies at the distal end region of the spread-out Bragg peak (SOBP) using single cell and multicell spheroids. MATERIALS AND METHODS: The Geant4 Monte Carlo simulation was used to calculate linear energy transfer (LET) values in passive scattering and spot scanning beams. The biological doses of the two beam options at various points of the distal end region of SOBP were investigated using EMT6 single cells and 0.6-mm V79 spheroids irradiated with 6 and 15 Gy, respectively, by inserting the fractions surviving these doses onto dose-survival curves and reading the corresponding dose. RESULTS: LET values in the entrance region of SOBP were similar between the two beam options and increased at the distal end region of SOBP, where the LET value of spot scanning beams was higher than that of passive scattering beams. Increases in biological effects at the distal end region were similarly observed in single cells and spheroids; biological doses at 2-10 mm behind the distal end were 4.5-57% and 5.7-86% higher than physical doses in passive scattering and spot scanning beams, respectively, with the biological doses of spot scanning beams being higher than those of passive scattering beams (p < .05). CONCLUSIONS: In single cells and spheroids, the effects of proton irradiation were stronger than expected from measured physical doses at the distal end of SOBP and were correlated with LET increases.


Asunto(s)
Protones , Dispersión de Radiación , Esferoides Celulares/efectos de la radiación , Línea Celular , Transferencia Lineal de Energía , Método de Montecarlo , Efectividad Biológica Relativa , Análisis de la Célula Individual , Esferoides Celulares/citología
9.
PLoS One ; 16(1): e0244513, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33395433

RESUMEN

Although 2D cell cultures are commonly used to predict therapy response, it has become clear that 3D cultures may better mimic the in vivo situation and offer the possibility of tailoring translational clinical approaches. Here, we compared the response of 2D and 3D colorectal cancer (CRC) cell lines to irradiation and chemotherapy. Classic 2D cultures and 3D spheroids of CRC cell lines (CaCo2, Colo205, HCT116, SW480) were thoroughly established, then irradiated with doses of 1, 4, or 10 Gy, using a clinical-grade linear accelerator. The response was assessed by immunohistochemistry, flow cytometry, and TUNEL assays. Upon irradiation, CRC 3D spheroids were morphologically altered. After irradiation with 10 Gy, annexin V/PI staining revealed a 1.8- to 4-fold increase in the apoptosis rate in the 2D cell cultures (95% CI 3.24±0.96), and a 1.5- to 2.4-fold increase in the 3D spheroids (95% CI 1.56±0.41). Irradiation with 1 Gy caused 3- and 4-fold increases in TUNEL positive cells in the CaCo2 and HCT116 (p = 0.01) 2D cultures, respectively, compared with a 2-fold increase in the 3D spheroids. Furthermore, the 2D and 3D cultures responded differently to chemotherapy; the 3D cultures were more resistant to 5-FU and cisplatin, but not to doxorubicin and mitomycin C, than the 2D cultures. Taken together, CRC cells cultured as 3D spheroids displayed markedly higher resistance to irradiation therapy and selected chemotherapeutic drugs than 2D cultures. This in vitro difference must be considered in future approaches for determining the ideal in vitro systems that mimic human disease.


Asunto(s)
Antineoplásicos/farmacología , Técnicas de Cultivo de Célula/métodos , Resistencia a Antineoplásicos/efectos de los fármacos , Radiación Ionizante , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Doxorrubicina/farmacología , Fluorouracilo/farmacología , Humanos , Tolerancia a Radiación/efectos de la radiación , Esferoides Celulares/citología , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/efectos de la radiación
10.
Lasers Med Sci ; 36(2): 339-347, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32623604

RESUMEN

This study aims to evaluate the photodynamic efficacy of purpurin 18 (pu-18) on triple negative breast cancer both in vitro and in vivo. Two states of 4T1 cells, 2D culture and 3D spheroids, were used to evaluate the photodynamic action of pu-18 in vitro. The in vitro study results indicated that for the 4T1 2D cell culture, the photodynamic therapy (PDT) treatment showed significant photocytotoxicity at low pu-18 concentrations following light irradiation. Pu-18 was found to distribute on the lysosomes, mitochondria, Golgi apparatus, and endoplasmic reticulum. After irradiation, pu-18 can generate ROS to destroy the mitochondrial membrane potential (MMP) and eventually induce apoptosis in the 2D 4T1 cells. Light-activated pu-18 could also induce the destruction of the 3D 4T1 cell spheroids. The in vivo study was conducted by using a subcutaneous 4T1 breast cancer animal model. The results demonstrated that pu-18 could remain in the tumor for more than 4 days by direct intra-tumoral injection. The PDT treatment was performed every 2 days for a total of 3 times. The results showed that PDT treatment could significantly inhibit tumor growth in vivo, indicating a good photodynamic efficacy of pu-18 in the mouse breast cancer model, without influencing weight and major organ function. The survival pattern results showed that PDT treatment could largely extend the survival time of mice with breast cancer. The preliminary conclusion is that photodynamic treatment using pu-18 is effective at preventing the growth of triple negative breast cancer cells both in vitro and in vivo. A combination of light irradiation and pu-18 could therefore be a worthwhile approach for the treatment of triple negative breast cancer.


Asunto(s)
Apoptosis , Fotoquimioterapia , Porfirinas/uso terapéutico , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Peso Corporal/efectos de los fármacos , Peso Corporal/efectos de la radiación , Línea Celular Tumoral , Femenino , Humanos , Luz , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Potencial de la Membrana Mitocondrial/efectos de la radiación , Ratones Endogámicos BALB C , Mitocondrias/efectos de los fármacos , Imagen Óptica , Fármacos Fotosensibilizantes/farmacología , Fármacos Fotosensibilizantes/uso terapéutico , Porfirinas/farmacología , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/patología , Esferoides Celulares/efectos de la radiación , Fracciones Subcelulares/metabolismo , Neoplasias de la Mama Triple Negativas/patología , Carga Tumoral/efectos de los fármacos , Carga Tumoral/efectos de la radiación
11.
Strahlenther Onkol ; 197(1): 27-38, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-32705304

RESUMEN

INTRODUCTION: Focal adhesion kinase (FAK) is a nonreceptor tyrosine kinase protein frequently overexpressed in cancer and has been linked to an increase in the stem cell population of tumors, resistance to therapy, and metastatic spread. Pharmacological FAK inhibition in pancreatic cancer has received increased attention over the last few years, either alone or in combination with other therapeutics including chemotherapy and immunotherapy. However, its prognostic value and its role in radioresistance of pancreatic ducal adenocarcinoma (PDAC) is unknown. METHODS AND MATERIALS: Using the TCGA and GTEx databases, we investigated the genetic alterations and mRNA expression levels of PTK2 (the encoding-gene for FAK) in normal pancreatic tissue and pancreatic cancer and its impact on patient survival. Furthermore, we evaluated the expression of FAK and its tyrosine domain Ty-397 in three pancreatic cancer cell lines. We went further and evaluated the role of a commercial FAK tyrosine kinase inhibitor VS-4718 on the viability and radiosensitization of the pancreatic cell lines as well as its effect on the extracellular matrix (ECM) production from the pancreatic stellate cells. Furthermore, we tested the effect of combining radiation with VS-4718 in a three-dimensional (3D) multicellular pancreatic tumor spheroid model. RESULTS: A database analysis revealed a relevant increase in genetic alterations and mRNA expression of the PTK2 in PDAC, which were associated with lower progression-free survival. In vitro, there was only variation in the basal phosphorylation level of FAK in cell lines. VS-4718 radiosensitized pancreatic cell lines only in the presence of ECM-producing pancreatic stellate cells and markedly reduced the ECM production in the stromal cells. Finally, using a 3D multicellular tumor model, the combination of VS-4718 and radiotherapy significantly reduced the growth of tumor aggregates. CONCLUSION: Pharmacological inhibition of FAK in pancreatic cancer could be a novel therapeutic strategy as our results show a radiosensitization effect of VS-4718 in vitro in a multicellular 2D- and in a 3D-model of pancreatic cancer.


Asunto(s)
Aminopiridinas/farmacología , Antineoplásicos/farmacología , Carcinoma Ductal Pancreático/tratamiento farmacológico , Quinasa 1 de Adhesión Focal/antagonistas & inhibidores , Proteínas de Neoplasias/antagonistas & inhibidores , Neoplasias Pancreáticas/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Fármacos Sensibilizantes a Radiaciones/farmacología , Carcinoma Ductal Pancreático/enzimología , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Técnicas de Cocultivo , Colágeno/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Histonas/análisis , Humanos , Estimación de Kaplan-Meier , Neoplasias Pancreáticas/enzimología , Células Estrelladas Pancreáticas/efectos de los fármacos , Células Estrelladas Pancreáticas/metabolismo , Supervivencia sin Progresión , Interferencia de ARN , ARN Mensajero/biosíntesis , ARN Neoplásico/biosíntesis , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología , Tolerancia a Radiación/efectos de los fármacos , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/efectos de la radiación , Células del Estroma/efectos de los fármacos , Ensayo de Tumor de Célula Madre
12.
Int J Radiat Biol ; 96(12): 1534-1540, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33074046

RESUMEN

PURPOSE: Agents that increase tumor radiosensitivity are of interest in improving outcomes in radiotherapy (XRT). DNA-PK inhibitors radiosensitize and alter cell adhesion proteins. We investigated combination radiation and a DNA-PK inhibitor in monolayers vs spheroids. MATERIALS AND METHODS: Using HER2 positive mammary carcinoma cells, we investigated the impact of NU7441, a DNA-PK inhibitor, on irradiated monolayer and spheroid cultures. Colony formation assays were performed with monolayer culture cells and spheroids after irradiation with/without NU7441 (5 µM). RESULTS: In monolayer culture cells, α/ß increased from 3.0 ± 0.2 Gy (XRT alone) to 6.9 ± 0.2 Gy (XRT+NU7441). Corresponding α/ß values for cells obtained by disaggregating treated spheroids were 3.6 ± 0.7 Gy (XRT alone) and 3.5 ± 0.2 Gy (XRT+NU7441). However, spheroid survival was highly sensitive to NU7441 incubation. After 4 Gy XRT alone 75% of the irradiated spheroids remained intact; when NU7441 treatment was involved, 13% remained intact. No spheroids survived to 3 weeks at 6 Gy or more. The discrepancy between the minimal change in α/ß from cells derived from spheroids and the spheroid growth response was not related to poor penetration of NU7441. CONCLUSIONS: DNA-PK inhibitor NU7441 radiosensitized monolayer cells but not cells obtained from spheroids. NU7441 and radiation increased spheroid fragmentation.


Asunto(s)
Neoplasias de la Mama/patología , Proteína Quinasa Activada por ADN/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/efectos de la radiación , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/radioterapia , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Humanos , Inhibidores de Proteínas Quinasas/uso terapéutico , Esferoides Celulares/patología
13.
Int J Mol Sci ; 21(19)2020 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-33027908

RESUMEN

Breast cancer is the leading cause of cancer death in females. The incidence has risen dramatically during recent decades. Dismissed as an "unsolved problem of the last century", breast cancer still represents a health burden with no effective solution identified so far. Microgravity (µg) research might be an unusual method to combat the disease, but cancer biologists decided to harness the power of µg as an exceptional method to increase efficacy and precision of future breast cancer therapies. Numerous studies have indicated that µg has a great impact on cancer cells; by influencing proliferation, survival, and migration, it shifts breast cancer cells toward a less aggressive phenotype. In addition, through the de novo generation of tumor spheroids, µg research provides a reliable in vitro 3D tumor model for preclinical cancer drug development and to study various processes of cancer progression. In summary, µg has become an important tool in understanding and influencing breast cancer biology.


Asunto(s)
Neoplasias de la Mama/terapia , Movimiento Celular/efectos de la radiación , Proliferación Celular/efectos de la radiación , Ingravidez , Neoplasias de la Mama/patología , Supervivencia Celular/efectos de la radiación , Femenino , Humanos , Esferoides Celulares/efectos de la radiación
14.
Int J Radiat Biol ; 96(11): 1413-1422, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32856972

RESUMEN

PURPOSE: Cancer stem cells (CSCs) are relatively resistant to radiation compared to their non-tumorigenic progeny. Ionizing radiation (IR) can expand the pool of CSCs that leads to more aggressive cancers, but the reason underlying CSC-induced cancer aggressiveness after radiation therapy remains unclear. To understand this, we investigated the phenotypic and molecular characteristics of sphere cells formed from IR-treated patient-derived xenograft (PDX) lung adenocarcinoma tumors. MATERIALS AND METHODS: After treatment with various modes of IR, we collected tumors from PDX mice and successfully obtained sphere cells. To compare tumorigenicity, we performed migration, invasion, and mouse transplantation assays with sphere cells from each group. To investigate the molecular features, we used a cDNA microarray and compared gene expression among groups. RESULTS AND CONCLUSIONS: Tumorigenicity assays revealed that sphere cells from 2- or 5-Gy IR-treated tumors more aggressive than sphere cells from non-IR treated tumors. Microarray results showed that SERPIB4 and CCL2 were upregulated in sphere cells from IR-treated tumors compared to that in sphere cells from non-IR treated tumors. Interestingly, these genes are related to immune reactions in cancer. Taken together, our results suggest that the aggressiveness of sphere cells obtained after IR treatment is related to resistance, and provide new opportunities for exploring targeted therapies to overcome common radioresistance.


Asunto(s)
Adenocarcinoma del Pulmón/patología , Transformación Celular Neoplásica , Esferoides Celulares/efectos de la radiación , Adenocarcinoma del Pulmón/radioterapia , Animales , Biomarcadores de Tumor/metabolismo , Humanos , Ratones , Esferoides Celulares/metabolismo , Esferoides Celulares/patología
15.
Biochem Biophys Res Commun ; 531(4): 535-542, 2020 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-32807492

RESUMEN

INTRODUCTION: Pre-clinical testing of small molecules for therapeutic development across many pathologies relies on the use of in-vitro and in-vivo models. When designed and implemented well, these models serve to predict the clinical outcome as well as the toxicity of the evaluated therapies. The two-dimensional (2D) reductionist approach where cells are incubated in a mono-layer on hard plastic microtiter plates is relatively inexpensive but not physiologically relevant. In contrast, well developed and applied three dimensional (3D) in vitro models could be employed to bridge the gap between 2D in vitro primary screening and expensive in vivo rodent models by incorporating key features of the tissue microenvironment to explore differentiation, cortical development, cancers and various neuronal dysfunctions. These features include an extracellular matrix, co-culture, tension and perfusion and could replace several hundred rodents in the drug screening validation cascade. METHODS: Human neural progenitor cells from middle brain (ReN VM, Merck Millipore, UK) were expanded as instructed by the supplier (Merck Millipore, UK), and then seeded in 96-well low-attachment plates (Corning, UK) to form multicellular spheroids followed by adding a Matrigel layer to mimic extracellular matrix around neural stem cell niche. ReN VM cells were then differentiated via EGF and bFGF deprivation for 7 days and were imaged at day 7. Radiotherapy was mimicked via gamma-radiation at 2Gy in the absence and presence of selected DYRK1A inhibitors Harmine, INDY and Leucettine 41 (L41). Cell viability was measured by AlamarBlue assay. Immunofluorescence staining was used to assess cell pluripotency marker SOX2 and differentiation marker GFAP. RESULTS: After 7 days of differentiation, neuron early differentiation marker (GFAP, red) started to be expressed among the cells expressing neural stem cell marker SOX2 (green). Radiation treatment caused significant morphology change including the reduced viability of the spheroids. These spheroids also revealed sensitizing potential of DYRK1A inhibitors tested in this study, including Harmine, INDY and L41. DISCUSSION & CONCLUSIONS: Combined with the benefit of greatly reducing the issues associated with in vivo rodent models, including reducing numbers of animals used in a drug screening cascade, cost, ethics, and potential animal welfare burden, we feel the well-developed and applied 3D neural spheroid model presented in this study will provide a crucial tool to evaluate combinatorial therapies, optimal drug concentrations and treatment dosages.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Células-Madre Neurales/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Esferoides Celulares/efectos de los fármacos , Línea Celular , Colágeno , Dioxoles/farmacología , Combinación de Medicamentos , Matriz Extracelular , Rayos gamma , Harmina/farmacología , Humanos , Imidazoles/farmacología , Laminina , Células-Madre Neurales/efectos de la radiación , Neuritas/efectos de los fármacos , Proteoglicanos , Fármacos Sensibilizantes a Radiaciones/farmacología , Factores de Transcripción SOXB1/metabolismo , Esferoides Celulares/efectos de la radiación , Quinasas DyrK
16.
Oxid Med Cell Longev ; 2020: 2534643, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32617133

RESUMEN

The presence of glioma stem cells (GSCs), which are enriched in neurospheres, may be connected to the radioresistance of glioblastoma (GBM) due to their enhanced antioxidant defense and elevated DNA repair capacity. The aim was to evaluate the responses to different radiation qualities and to reduce radioresistance of U87MG cells, a GBM cell line. U87MG cells were cultured in a 3D model and irradiated with low (24 mGy/h) and high (0.39 Gy/min) dose rates of low LET gamma and high LET carbon ions (1-2 Gy/min). Thereafter, expression of proteins related to oxidative stress response, extracellular 8-oxo-dG, and neurospheres were determined. LD50 for carbon ions was significantly lower compared to LD50 of high and low dose rate gamma radiation. A significantly higher level of 8-oxo-dG was detected in the media of cells exposed to a low dose rate as compared to a high dose rate of gamma or carbon ions. A downregulation of oxidative stress proteins was also observed (NRF2, hMTH1, and SOD1). The NRF2 gene was knocked down by CRISPR/Cas9 in neurosphere cells, resulting in less self-renewal, more differentiated cells, and less proliferation capacity after irradiation with low and high dose rate gamma rays. Overall, U87MG glioma neurospheres presented differential responses to distinct radiation qualities and NRF2 plays an important role in cellular sensitivity to radiation.


Asunto(s)
Antioxidantes/metabolismo , Rayos gamma , Glioblastoma/patología , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo/efectos de la radiación , Esferoides Celulares/patología , Sistemas CRISPR-Cas/genética , Línea Celular Tumoral , Supervivencia Celular/efectos de la radiación , Relación Dosis-Respuesta en la Radiación , Humanos , Modelos Biológicos , Proteínas de Neoplasias/metabolismo , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Proteínas del Tejido Nervioso/metabolismo , Proteínas de Unión al ARN/metabolismo , Tolerancia a Radiación/efectos de la radiación , Esferoides Celulares/efectos de la radiación
17.
Stem Cell Reports ; 15(2): 374-388, 2020 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-32649903

RESUMEN

Intestinal regeneration is crucial for functional restoration after injury, and nutritional molecules can play an important role in this process. Here, we found that arachidonic acid (AA) serves as a direct proliferation promoter of intestinal epithelial cells that facilitates small intestinal regeneration in both three-dimensional cultured organoids and mouse models. As shown in the study, during post-irradiation regeneration, AA positively regulates intestinal epithelial cell proliferation by upregulating the expression of Ascl2 and activating WNT signaling, but negatively regulates intestinal epithelial cell differentiation. AA acts as a delicate regulator that efficiently facilitates epithelial tissue repair by activating radiation-resistant Msi1+ cells rather than Lgr5+ cells, which are extensively considered WNT-activated crypt base stem cells. Additionally, short-term AA treatment maintains optimal intestinal epithelial homeostasis under physiological conditions. As a result, AA treatment can be considered a potential therapy for irradiation injury repair and tissue regeneration.


Asunto(s)
Ácido Araquidónico/farmacología , Intestino Delgado/fisiología , Regeneración/efectos de los fármacos , Vía de Señalización Wnt , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/efectos de la radiación , Línea Celular , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/efectos de la radiación , Intestino Delgado/citología , Masculino , Ratones Endogámicos C57BL , Organoides/citología , Radiación Ionizante , Regeneración/efectos de la radiación , Esferoides Celulares/citología , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/efectos de la radiación , Transcriptoma/genética , Vía de Señalización Wnt/efectos de los fármacos , Vía de Señalización Wnt/efectos de la radiación
18.
Cells ; 9(5)2020 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-32397212

RESUMEN

Compact chromatin is linked to a poor tumour prognosis and resistance to radiotherapy from photons. We investigated DNA damage induction and repair in the context of chromatin structure for densely ionising alpha radiation as well as its therapeutic potential. Chromatin opening by histone deacetylase inhibitor trichostatin A (TSA) pretreatment reduced clonogenic survival and increased γH2AX foci in MDA-MB-231 cells, indicative of increased damage induction by free radicals using gamma radiation. In contrast, TSA pretreatment tended to improve survival after alpha radiation while γH2AX foci were similar or lower; therefore, an increased DNA repair is suggested due to increased access of repair proteins. MDA-MB-231 cells exposed to fractionated gamma radiation (2 Gy × 6) expressed high levels of stem cell markers, elevated heterochromatin H3K9me3 marker, and a trend towards reduced clonogenic survival in response to alpha radiation. There was a higher level of H3K9me3 at baseline, and the ratio of DNA damage induced by alpha vs. gamma radiation was higher in the aggressive MDA-MB-231 cells compared to hormone receptor-positive MCF7 cells. We demonstrate that heterochromatin structure and stemness properties are induced by fractionated radiation exposure. Gamma radiation-exposed cells may be targeted using alpha radiation, and we provide a mechanistic basis for the involvement of chromatin in these effects.


Asunto(s)
Partículas alfa , Neoplasias de la Mama/metabolismo , Rayos gamma , Heterocromatina/efectos de la radiación , Acetilación , Neoplasias de la Mama/patología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Células Clonales , Femenino , Histonas/metabolismo , Humanos , Ácidos Hidroxámicos/farmacología , Lisina/metabolismo , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Células Madre Neoplásicas/efectos de la radiación , Exposición a la Radiación , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/patología , Esferoides Celulares/efectos de la radiación
19.
Sci Rep ; 10(1): 1653, 2020 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-32015396

RESUMEN

For multimodality therapies such as the combination of hyperthermia and radiation, quantification of biological effects is key for dose prescription and response prediction. Tumour spheroids have a microenvironment that more closely resembles that of tumours in vivo and may thus be a superior in vitro cancer model than monolayer cultures. Here, the response of tumour spheroids formed from two established human cancer cell lines (HCT116 and CAL27) to single and combination treatments of radiation (0-20 Gy), and hyperthermia at 47 °C (0-780 CEM43) has been evaluated. Response was analysed in terms of spheroid growth, cell viability and the distribution of live/dead cells. Time-lapse imaging was used to evaluate mechanisms of cell death and cell detachment. It was found that sensitivity to heat in spheroids was significantly less than that seen in monolayer cultures. Spheroids showed different patterns of shrinkage and regrowth when exposed to heat or radiation: heated spheroids shed dead cells within four days of heating and displayed faster growth post-exposure than samples that received radiation or no treatment. Irradiated spheroids maintained a dense structure and exhibited a longer growth delay than spheroids receiving hyperthermia or combination treatment at (thermal) doses that yielded equivalent levels of clonogenic cell survival. We suggest that, unlike radiation, which kills dividing cells, hyperthermia-induced cell death affects cells independent of their proliferation status. This induces microenvironmental changes that promote spheroid growth. In conclusion, 3D tumour spheroid growth studies reveal differences in response to heat and/or radiation that were not apparent in 2D clonogenic assays but that may significantly influence treatment efficacy.


Asunto(s)
Hipertermia Inducida , Neoplasias/radioterapia , Neoplasias/terapia , Línea Celular Tumoral , Proliferación Celular/efectos de la radiación , Supervivencia Celular/efectos de la radiación , Terapia Combinada , Relación Dosis-Respuesta en la Radiación , Células HCT116 , Humanos , Modelos Biológicos , Neoplasias/patología , Esferoides Celulares/patología , Esferoides Celulares/efectos de la radiación , Microambiente Tumoral/efectos de la radiación , Ensayo de Tumor de Célula Madre
20.
Mol Pharm ; 17(1): 301-315, 2020 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-31765570

RESUMEN

The controversy surrounding the use of diphtheria toxin (DT) as a therapeutic agent against tumor cells arises mainly from its unexpected harmfulness to healthy tissues. We encoded the cytotoxic fragment A of DT (DTA) as an objective gene in the Light-On gene-expression system to construct plasmids pGAVPO (pG) and pU5-DTA (pDTA). Meanwhile, a cRGD-modified ternary complex comprising plasmids, chitosan, and liposome (pG&pDTA@cRGD-CL) was prepared as a nanocarrier to ensure transfection efficiency. Benefiting from spatiotemporal control of this light-switchable transgene system and the superior tumor targeting of the carrier, toxins were designed to be expressed selectively in illuminated lesions. In vitro studies suggested that pG&pDTA@cRGD-CL exerted arrest of the S phase in B16F10 cells upon blue light irradiation and, ultimately, induced the apoptosis and necrosis of tumor cells. Such DTA-based treatment exerted enhanced antitumor activity in mice bearing B16F10 xenografts and displayed prolonged survival time with minimal side effects. Hence, we described novel DTA-based therapy combined with nanotechnology and the Light-On gene-expression system: such treatment could be a promising strategy against melanoma.


Asunto(s)
Toxina Diftérica/genética , Expresión Génica/efectos de la radiación , Terapia Genética , Liposomas/química , Melanoma Experimental/terapia , Nanotecnología/métodos , Fragmentos de Péptidos/genética , Animales , Apoptosis/genética , Apoptosis/efectos de la radiación , Línea Celular Tumoral , Quitosano/química , Expresión Génica/genética , Liposomas/ultraestructura , Masculino , Melanoma Experimental/genética , Melanoma Experimental/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Desnudos , Microscopía Electrónica de Transmisión , Tamaño de la Partícula , Péptidos Cíclicos/química , Puntos de Control de la Fase S del Ciclo Celular/efectos de los fármacos , Puntos de Control de la Fase S del Ciclo Celular/genética , Puntos de Control de la Fase S del Ciclo Celular/efectos de la radiación , Esferoides Celulares/efectos de la radiación , Distribución Tisular , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA