Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Nat Cell Biol ; 24(6): 940-953, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35654839

RESUMEN

Bidirectional signalling between the tumour and stroma shapes tumour aggressiveness and metastasis. ATF4 is a major effector of the Integrated Stress Response, a homeostatic mechanism that couples cell growth and survival to bioenergetic demands. Using conditional knockout ATF4 mice, we show that global, or fibroblast-specific loss of host ATF4, results in deficient vascularization and a pronounced growth delay of syngeneic melanoma and pancreatic tumours. Single-cell transcriptomics of tumours grown in Atf4Δ/Δ mice uncovered a reduction in activation markers in perivascular cancer-associated fibroblasts (CAFs). Atf4Δ/Δ fibroblasts displayed significant defects in collagen biosynthesis and deposition and a reduced ability to support angiogenesis. Mechanistically, ATF4 regulates the expression of the Col1a1 gene and levels of glycine and proline, the major amino acids of collagen. Analyses of human melanoma and pancreatic tumours revealed a strong correlation between ATF4 and collagen levels. Our findings establish stromal ATF4 as a key driver of CAF functionality, malignant progression and metastasis.


Asunto(s)
Fibroblastos Asociados al Cáncer , Melanoma , Neoplasias Pancreáticas , Animales , Fibroblastos Asociados al Cáncer/metabolismo , Colágeno/metabolismo , Fibroblastos/metabolismo , Regulación Neoplásica de la Expresión Génica , Melanoma/genética , Ratones , Ratones Noqueados , Neovascularización Patológica/metabolismo , Neoplasias Pancreáticas/patología
2.
Radiat Res ; 198(2): 181-189, 2022 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-35640166

RESUMEN

FLASH is a high-dose-rate form of radiation therapy that has the reported ability, compared with conventional dose rates, to spare normal tissues while being equipotent in tumor control, thereby increasing the therapeutic ratio. The mechanism underlying this normal tissue sparing effect is currently unknown, however one possibility is radiochemical oxygen depletion (ROD) during dose delivery in tissue at FLASH dose rates. In order to investigate this possibility, we used the phosphorescence quenching method to measure oxygen partial pressure before, during and after proton radiation delivery in model solutions and in normal muscle and sarcoma tumors in mice, at both conventional (Conv) (∼0.5 Gy/s) and FLASH (∼100 Gy/s) dose rates. Radiation dosimetry was determined by Advanced Markus Chamber and EBT-XL film. For solutions contained in sealed glass vials, phosphorescent probe Oxyphor PtG4 (1 µM) was dissolved in a buffer (10 mM HEPES) containing glycerol (1 M), glucose (5 mM) and glutathione (5 mM), designed to mimic the reducing and free radical-scavenging nature of the intracellular environment. In vivo oxygen measurements were performed 24 h after injection of PtG4 into the interstitial space of either normal thigh muscle or subcutaneous sarcoma tumors in mice. The "g-value" for ROD is reported in mmHg/Gy, which represents a slight modification of the more standard chemical definition (µM/Gy). In solutions, proton irradiation at conventional dose rates resulted in a g-value for ROD of up to 0.55 mmHg/Gy, consistent with earlier studies using X or gamma rays. At FLASH dose rates, the g-value for ROD was ∼25% lower, 0.37 mmHg/Gy. pO2 levels were stable after each dose delivery. For normal muscle in vivo, oxygen depletion during irradiation was counterbalanced by resupply from the vasculature. This process was fast enough to maintain tissue pO2 virtually unchanged at Conv dose rates. However, during FLASH irradiation there was a stepwise decrease in pO2 (g-value ∼0.28 mmHg/Gy), followed by a rebound to the initial level after ∼8 s. The g-values were smaller and recovery times longer in tumor tissue when compared to muscle and may be related to the lower initial endogenous pO2 levels in the former. Considering that the FLASH effect is seen in vivo even at doses as low as 10 Gy, it is difficult to reconcile the amount of protection seen by oxygen depletion alone. However, the phosphorescence probe in our experiments was confined to the extracellular space, and it remains possible that intracellular oxygen depletion was greater than observed herein. In cell-mimicking solutions the oxygen depletion g-vales were indeed significantly higher than observed in vivo.


Asunto(s)
Protones , Sarcoma , Animales , Rayos gamma , Ratones , Oxígeno , Radiometría/métodos , Dosificación Radioterapéutica , Sarcoma/radioterapia
3.
Cancer Res ; 81(18): 4808-4821, 2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34321243

RESUMEN

In studies of electron and proton radiotherapy, ultrahigh dose rates of FLASH radiotherapy appear to produce fewer toxicities than standard dose rates while maintaining local tumor control. FLASH-proton radiotherapy (F-PRT) brings the spatial advantages of PRT to FLASH dose rates (>40 Gy/second), making it important to understand if and how F-PRT spares normal tissues while providing antitumor efficacy that is equivalent to standard-proton radiotherapy (S-PRT). Here we studied PRT damage to skin and mesenchymal tissues of muscle and bone and found that F-PRT of the C57BL/6 murine hind leg produced fewer severe toxicities leading to death or requiring euthanasia than S-PRT of the same dose. RNA-seq analyses of murine skin and bone revealed pathways upregulated by S-PRT yet unaltered by F-PRT, such as apoptosis signaling and keratinocyte differentiation in skin, as well as osteoclast differentiation and chondrocyte development in bone. Corroborating these findings, F-PRT reduced skin injury, stem cell depletion, and inflammation, mitigated late effects including lymphedema, and decreased histopathologically detected myofiber atrophy, bone resorption, hair follicle atrophy, and epidermal hyperplasia. F-PRT was equipotent to S-PRT in control of two murine sarcoma models, including at an orthotopic intramuscular site, thereby establishing its relevance to mesenchymal cancers. Finally, S-PRT produced greater increases in TGFß1 in murine skin and the skin of canines enrolled in a phase I study of F-PRT versus S-PRT. Collectively, these data provide novel insights into F-PRT-mediated tissue sparing and support its ongoing investigation in applications that would benefit from this sparing of skin and mesenchymal tissues. SIGNIFICANCE: These findings will spur investigation of FLASH radiotherapy in sarcoma and additional cancers where mesenchymal tissues are at risk, including head and neck cancer, breast cancer, and pelvic malignancies.


Asunto(s)
Epitelio , Tratamientos Conservadores del Órgano , Terapia de Protones , Sarcoma/patología , Sarcoma/radioterapia , Animales , Huesos/patología , Huesos/efectos de la radiación , Modelos Animales de Enfermedad , Perros , Epitelio/efectos de la radiación , Femenino , Perfilación de la Expresión Génica , Humanos , Ratones , Morbilidad , Músculos/patología , Músculos/efectos de la radiación , Tratamientos Conservadores del Órgano/métodos , Terapia de Protones/efectos adversos , Terapia de Protones/métodos , Traumatismos por Radiación/diagnóstico , Traumatismos por Radiación/etiología , Dosificación Radioterapéutica , Sarcoma/metabolismo , Piel/efectos de la radiación , Resultado del Tratamiento
4.
Cancer Res ; 81(9): 2399-2414, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33514514

RESUMEN

Inflammatory breast cancer (IBC) is a highly metastatic breast carcinoma with high frequency of estrogen receptor α (ERα) negativity. Here we explored the role of the second ER subtype, ERß, and report expression in IBC tumors and its correlation with reduced metastasis. Ablation of ERß in IBC cells promoted cell migration and activated gene networks that control actin reorganization, including G-protein-coupled receptors and downstream effectors that activate Rho GTPases. Analysis of preclinical mouse models of IBC revealed decreased metastasis of IBC tumors when ERß was expressed or activated by chemical agonists. Our findings support a tumor-suppressive role of ERß by demonstrating the ability of the receptor to inhibit dissemination of IBC cells and prevent metastasis. On the basis of these findings, we propose ERß as a potentially novel biomarker and therapeutic target that can inhibit IBC metastasis and reduce its associated mortality. SIGNIFICANCE: These findings demonstrate the capacity of ERß to elicit antimetastatic effects in highly aggressive inflammatory breast cancer and propose ERß and the identified associated genes as potential therapeutic targets in this disease.


Asunto(s)
Actinas/metabolismo , Movimiento Celular/genética , Receptor beta de Estrógeno/metabolismo , Neoplasias Inflamatorias de la Mama/metabolismo , Transducción de Señal/genética , Citoesqueleto de Actina/metabolismo , Animales , Estudios de Cohortes , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Receptor beta de Estrógeno/genética , Femenino , Técnicas de Inactivación de Genes , Células HEK293 , Humanos , Neoplasias Inflamatorias de la Mama/genética , Neoplasias Inflamatorias de la Mama/patología , Células MCF-7 , Ratones , Metástasis de la Neoplasia/genética , Transfección , Carga Tumoral/genética , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Phys Med Biol ; 65(16): 165002, 2020 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-32413889

RESUMEN

This paper aims to demonstrate the difference in biological effectiveness of proton monoenergetic arc therapy (PMAT) compared to intensity modulated proton therapy (IMPT) and conventional 6 MV photon therapy, and to quantify this difference when exposing cells of different radiosensitivity to the same experimental conditions for each modality. V79, H1299 and H460 cells were cultured in petri dishes placed in the central axis of a cylindrical and homogeneous solid water phantom of 20 cm in diameter. For the PMAT plan, cells were exposed to 13 mono-energetic proton beams separated every 15° over a 180° arc, designed to deliver a uniform dose of higher LET to the petri dishes. For the IMPT plans, 3 fields were used, where each field was modulated to cover the full target. Cells were also exposed to 6 MV photon beams in petri dishes to characterize their radiosensitivity. The relative biological effectiveness of the PMAT plans compared with those of IMPT was measured using clonogenic assays. Similarly, in order to study the quantity and quality of the DNA damage induced by the PMAT plans compared to that of IMPT and photons, γ-H2AX assays were conducted to study the relative amount of DNA damage induced by each modality, and their repair rate over time. The clonogenic assay revealed similar survival levels to the same dose delivered with IMPT or x-rays. However, a systematic average of up to a 43% increase in effectiveness in PMAT plans was observed when compared with IMPT. In addition, the repair kinetic assays proved that PMAT induces larger and more complex DNA damage (evidenced by a slower repair rate and a larger proportion of unrepaired DNA damage) than IMPT. The repair kinetics of IMPT and 6 MV photon therapy were similar. Mono-energetic arc beams offer the possibility of taking advantage of the enhanced LET of proton beams to increase TCP. This study presents initial results based on exposing cells with different radiosensitivity to other modalities under the same experimental conditions, but more extensive clonogenic and in-vivo studies will be required to confirm the validity of these results.


Asunto(s)
Fantasmas de Imagen , Fotones , Terapia de Protones , Radiobiología , Planificación de la Radioterapia Asistida por Computador/métodos , Humanos , Fotones/uso terapéutico , Dosificación Radioterapéutica , Efectividad Biológica Relativa
6.
Oncogene ; 39(22): 4344-4357, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32335582

RESUMEN

We explore a novel strategy of activating immune signaling through increased micronuclei formation utilizing a cell cycle checkpoint inhibitor to drive cell cycle progression following ionizing radiation. The Chk1/2 inhibitor AZD7762 is used to abrogate radiation therapy (RT)-induced G2/M cell cycle arrest in multiple cell lines and, we find that this therapeutic combination promotes increased micronuclei formation in vitro and subsequently drives increased type I interferon signaling and cytotoxic T-cell activation. In vivo studies using B16-F10 melanoma cancer cells implanted in C57/BL6 mice demonstrate improved rates of tumor control at the abscopal (unirradiated) site, located outside of the radiation field, only in the AZD7762 + RT group, with a corresponding reduction in mean tumor volume, increase in the CD8 T-cell population, and immune activated gene signaling. Our results demonstrate that targeted inhibition of cell cycle checkpoint activation following ionizing radiation drives increased production of immunogenic micronuclei, leading to systemic tumor response with potential future clinical benefit.


Asunto(s)
Núcleo Celular/efectos de los fármacos , Núcleo Celular/efectos de la radiación , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/antagonistas & inhibidores , Quinasa de Punto de Control 2/antagonistas & inhibidores , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de la radiación , Melanoma Experimental/inmunología , Proteínas de Neoplasias/antagonistas & inhibidores , Tiofenos/farmacología , Urea/análogos & derivados , Animales , Línea Celular Tumoral , Femenino , Humanos , Interferón beta/biosíntesis , Interferón beta/genética , Melanoma Experimental/tratamiento farmacológico , Melanoma Experimental/patología , Melanoma Experimental/radioterapia , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/genética , Ratones Endogámicos C57BL , Pruebas de Micronúcleos , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Interferencia de ARN , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología , Factor de Transcripción STAT1/biosíntesis , Factor de Transcripción STAT1/genética , Carga Tumoral/efectos de los fármacos , Carga Tumoral/efectos de la radiación , Urea/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Invest New Drugs ; 36(5): 773-781, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29387992

RESUMEN

Effective cytoprotectors that are selective for normal tissues could decrease radiotherapy and chemotherapy sequelae and facilitate the safe administration of higher radiation doses. This could improve the cure rates of radiotherapy for cancer patients. Autophagy is a cytoplasmic cellular process that is necessary for the clearance of damaged or aged proteins and organelles. It is a strong determinant of post-irradiation cell fate. In this study, we investigated the effect of the mTOR-independent small molecule enhancer of autophagy (SMER28) on mouse liver autophagy and post-irradiation recovery of mouse bone marrow and liver. SMER28 enhanced the autophagy flux and improved the survival of normal hepatocytes. This effect was specific for normal cells because SMER28 had no protective effect on hepatoma or other cancer cell line survival in vitro. In vivo subcutaneous administration of SMER28 protected mouse liver and bone marrow against radiation damage and facilitated survival of mice after lethal whole body or abdominal irradiation. These findings open a new field of research on autophagy-targeting radioprotectors with clinical applications in oncology, occupational, and space medicine.


Asunto(s)
Compuestos Alílicos/farmacología , Autofagia/efectos de los fármacos , Médula Ósea/efectos de los fármacos , Hígado/efectos de los fármacos , Quinazolinas/farmacología , Protectores contra Radiación/farmacología , Animales , Autofagia/efectos de la radiación , Médula Ósea/efectos de la radiación , Línea Celular , Humanos , Hígado/efectos de la radiación , Masculino , Ratones Endogámicos BALB C , Neoplasias/radioterapia , Serina-Treonina Quinasas TOR , Irradiación Corporal Total
9.
Anticancer Res ; 38(1): 227-238, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29277777

RESUMEN

BACKGROUND/AIM: Amifostine is the only selective normal tissue cytoprotector, approved for the protection against platinum toxicities and radiotherapy-induced xerostomia. Free radical scavenger and DNA repair activities have been attributed to the drug. MATERIALS AND METHODS: We investigated the effect of amifostine on autophagy, lysosomal biogenesis and lipophagy of normal mouse liver exposed to clinically relevant doses of radiation. RESULTS: The study provides evidence that ionizing radiation blocks autophagy activity and lysosomal biogenesis in normal mouse liver. Amifostine, protects the liver autophagic machinery and induces lysosomal biogenesis. By suppressing autophagy, ionizing radiation induces lipid droplet accumulation, while pre-treatment with amifostine protects lipophagy and up-regulates the TIP47 protein and mRNA levels, showing a maintenance of lipid metabolism in the liver cells. CONCLUSION: It is concluded that amifostine, aside to DNA protection activity, exerts its cytoprotective function by preventing radiation-induced blockage of autophagy, lysosomal biogenesis and lipophagy.


Asunto(s)
Amifostina/farmacología , Hígado/efectos de los fármacos , Protectores contra Radiación/farmacología , Animales , Autofagia/efectos de los fármacos , Rayos gamma , Metabolismo de los Lípidos/efectos de los fármacos , Hígado/metabolismo , Hígado/efectos de la radiación , Hígado/ultraestructura , Lisosomas/metabolismo , Masculino , Ratones Endogámicos BALB C
10.
In Vivo ; 31(2): 175-179, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28358697

RESUMEN

BACKGROUND/AIM: Altered fractionation is an area of intense clinical research in radiation oncology. Estimation of the α/ß ratio of individual carcinomas after establishment of primary cell cultures from tumor biopsies may prove of importance in the individualization of radiotherapy schemes. MATERIALS AND METHODS: Here we proposed a simple method to estimate the α/ß ratio in cultured cell lines (two lung carcinomas: A549 and H1299; one lung fibroblast cell line: MRC5), using viability assays. RESULTS: For the A549 cell line, the α/ß ratio ranged from 14-25 Gy, for H1299 from 11-43 Gy and for the MRC5 fibroblast cell line this was far lower, ranging from 0.69 to 6 Gy. The α/ß ratio decreased when extracted from comparisons of lower dose per fraction schemes. CONCLUSION: The α/ß ratio of a cell line can be easily defined after simple viability/dose fractionation experiments.


Asunto(s)
Apoptosis/efectos de la radiación , Proliferación Celular/efectos de la radiación , Relación Dosis-Respuesta en la Radiación , Fibroblastos/efectos de la radiación , Células A549 , Línea Celular , Línea Celular Tumoral , Supervivencia Celular/efectos de la radiación , Fraccionamiento de la Dosis de Radiación , Fibroblastos/citología , Humanos , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/radioterapia , Radioterapia/métodos
11.
Sci Rep ; 6: 30986, 2016 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-27507219

RESUMEN

The mechanism of Amifostine (WR-2721) mediated radioprotection is poorly understood. The effects of amifostine on human basal metabolism, mouse liver metabolism and on normal and tumor hepatic cells were studied. Indirect calorimetric canopy tests showed significant reductions in oxygen consumption and of carbon dioxide emission in cancer patients receiving amifostine. Glucose levels significantly decreased and lactate levels increased in patient venous blood. Although amifostine in vitro did not inhibit the activity of the prolyl-hydroxylase PHD2, experiments with mouse liver showed that on a short timescale WR-1065 induced expression of the Hypoxia Inducible Factor HIF1α, lactate dehydrogenase LDH5, glucose transporter GLUT2, phosphorylated pyruvate dehydrogenase pPDH and PDH-kinase. This effect was confirmed on normal mouse NCTC hepatocytes, but not on hepatoma cells. A sharp reduction of acetyl-CoA and ATP levels in NCTC cells indicated reduced mitochondrial usage of pyruvate. Transient changes of mitochondrial membrane potential and reactive oxygen species ROS production were evident. Amifostine selectively protects NCTC cells against radiation, whilst HepG2 neoplastic cells are sensitized. The radiation protection was correlates with HIF levels. These findings shed new light on the mechanism of amifostine cytoprotection and encourage clinical research with this agent for the treatment of primary and metastatic liver cancer.


Asunto(s)
Amifostina/farmacología , Neoplasias de la Mama/radioterapia , Protectores contra Radiación/farmacología , Adenosina Trifosfato/metabolismo , Animales , Metabolismo Basal/efectos de los fármacos , Glucemia/metabolismo , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Femenino , Transportador de Glucosa de Tipo 2/metabolismo , Glucólisis/efectos de los fármacos , Glucólisis/efectos de la radiación , Hepatocitos/efectos de los fármacos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Potencial de la Membrana Mitocondrial/efectos de la radiación , Ratones Endogámicos BALB C , Oxígeno/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora
12.
Br J Cancer ; 115(3): 312-21, 2016 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-27380135

RESUMEN

BACKGROUND: The cellular autophagic response to radiation is complex. Various cells and tissues respond differentially to radiation, depending on both the dose of exposure and the time post irradiation. In the current study, we determined the autophagosomal and lysosomal response to radiation in lung cancer cell lines by evaluating the expression of the associated proteins, as well as the effect of relevant gene silencing in radio and chemosensitisation. Furthermore, tumour sensitisation was evaluated in in vivo autophagic gene silencing model after irradiation. METHODS: A549 and H1299 cell lines were utilised as in vitro cancer models. Both cell lines were transfected with various small-interfering RNAs, silencing auto-lysosomal genes, and irradiated with 4 Gy. Cell growth response was evaluated with AlamarBlue assay. Western blot and confocal microscopy were utilised for the characterisation of the auto-lysosomal flux. Also, the H1299 cell line was stable transfected with small-hairpin RNA of the MAP1LC3A gene, and the tumour radiosensitisation in Athymic Nude-Foxn1(nu) was evaluated. RESULTS: Following exposure to 4 Gy of radiation, A549 cells exhibited a significant induction of the autophagic flux, which was not supported by transcriptional activation of auto-lysosomal genes (LC3A, LC3B, p62, TFEB and LAMP2a), resulting in aggresome accumulation. Recovery of transcriptional activity and autophagy efficacy occurred 7 days post irradiation. Alternatively, H1299 cells, a relatively radio-resistant cell line, sharply responded with an early (at 2 days) transcriptional activation of auto-lysosomal genes that sustained an effective autophagosomal flux, resulting in adequate aggresome clearance. Subsequently, we tested the silencing of four genes (LC3A, LC3B, TFEB and LAMP2a), confirming a significant radiosensitisation and chemosensitisation to various chemotherapeutic agents, including cisplatin and taxanes. In mouse xenografts, exposure to radiation significantly reduced tumour growth (P<0.001), which was exacerbated among shLC3A-H1299 transfected tumours. CONCLUSIONS: The ability of lung cancer cells to survive after irradiation at 4 Gy depends on their ability to sustain a functional autophagic flux. Abrogation of such ability results in increased radiosensitivity and susceptibility to various chemotherapy agents. Selective inhibitors of cancer cell autophagic function may prove important for the eradication of lung cancer.


Asunto(s)
Autofagia , Neoplasias Pulmonares/patología , Animales , Línea Celular Tumoral , Silenciador del Gen , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/radioterapia , Ratones , Ratones Desnudos , Tolerancia a Radiación , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Med Phys ; 43(5): 2200, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27147332

RESUMEN

PURPOSE: In vivo radiobiology experiments involving partial body irradiation (PBI) of mice are of major importance because they allow for the evaluation of individual organ tolerance; overcoming current limitations of experiments using lower dose, whole body irradiation. In the current study, the authors characterize and validate an effective and efficient apparatus for multiple animal PBI, directed to the head, thorax, or abdomen of mice. METHODS: The apparatus is made of polymethylmethacrylate and consists of a rectangular parallelepiped prism (40 cm × 16 cm × 8 cm), in which five holes were drilled to accomodate standard 60 ml syringes, each housing an unanesthetized, fully immobilized mouse. Following CT-scanning and radiotherapy treatment planning, radiation fields were designed to irradiate the head, thorax, or abdomen of the animal. Thermoluminescent dosimeters (TLDs) were used to confirm the treatment planning dosimetry for primary beam and scattered radiation. RESULTS: Mice are efficiently placed into 60 ml syringes and immobilized, without the use of anesthetics. Although partial rotational movement around the longitudinal axis and a minor 2 mm forward/backward movement are permitted, this does not compromise the irradiation of the chosen body area. TLDs confirmed the dose values predicted by the treatment planning dosimetry, both for primary beam and scattered radiation. CONCLUSIONS: The customized PMMA apparatus described and validated is cost-effective, convenient to use, and efficient in performing PBI without the use of anesthesia. The developed apparatus permits the isolated irradiation of the mouse head, thorax, and abdomen. Importantly, the apparatus allows the delivery of PBI to five mice, simultaneously, representing an efficient way to effectively expose a large number of animals to PBI through multiple daily fractions, simulating clinical radiotherapy treatment schedules.


Asunto(s)
Imagenología Tridimensional/instrumentación , Inmovilización/instrumentación , Radiocirugia/instrumentación , Radioterapia Guiada por Imagen/instrumentación , Tomografía Computarizada por Rayos X/instrumentación , Abdomen/efectos de la radiación , Animales , Conducta Animal , Diseño de Equipo , Cabeza/efectos de la radiación , Imagenología Tridimensional/métodos , Inmovilización/métodos , Inmovilización/psicología , Masculino , Ratones Endogámicos BALB C , Movimiento , Polimetil Metacrilato , Radiocirugia/métodos , Dosificación Radioterapéutica , Planificación de la Radioterapia Asistida por Computador/instrumentación , Planificación de la Radioterapia Asistida por Computador/métodos , Radioterapia Guiada por Imagen/métodos , Dosimetría Termoluminiscente/instrumentación , Dosimetría Termoluminiscente/métodos , Tórax/efectos de la radiación , Tomografía Computarizada por Rayos X/métodos
14.
Lung Cancer ; 90(1): 98-105, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26264650

RESUMEN

OBJECTIVES: We investigated the role of lysosomal biogenesis and hydrolase activity in the clinical behavior and postoperative outcome of lung cancer. MATERIALS AND METHODS: Using immunohistochemistry we investigated the expression of the transcription factor EB (TFEB) which orchestrates lysosomal biogenesis, the lysosome membrane protein LAMP2a and of the lysosomal hydrolase cathepsin D in a series of 98 non-small cell lung carcinomas (NSCLC) treated with surgery alone. In vitro experiments with the A549 and H1299 lung cancer cell lines were also performed. RESULTS: Overexpression of TFEB, LAMP2a and Cathepsin D was noted in 47/98 (47.9%), 43/98 (43.9%) and 39/98 (39.8%) cases, respectively, and were significantly correlated with each other and with adenocarcinomas. High LAMP2a was related to high histology grade. Linear regression analysis confirmed significant association of TFEB with BNIP3 (p=0.0003, r=0.35) and LC3A with LAMP2a expression (p=0.0002, r=0.37). An inverse association of Cathepsin D expression with stone-like structures (SLS) was recorded (p=0.02, r=0.22). On univariate analysis all three lyososomal variables were associated with poor prognosis (p=0.05, 0.04 and 0.01, for TFEB, Cathepsin D and LAMP2a, respectively). Multivariate analysis showed that the SLS number (p=0.0001, HR5.37), Cathepsin D expression (p=0.01, HR=2.2) and stage (p=0.01, HR=1.5) were independent prognostic variables. Silencing of TFEB with siRNAs in the A549 and H1299 lung cancer cell lines did not affect proliferation but resulted in reduced migration ability. CONCLUSION: Lysosomal biogenesis is linked to autophagosomal protein expression in NSCLC and characterizes subgroups of high risk patients after complete surgical lung tumor resection.


Asunto(s)
Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/biosíntesis , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Movimiento Celular/fisiología , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Adulto , Anciano , Anciano de 80 o más Años , Autofagia/fisiología , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/cirugía , Catepsina D/biosíntesis , Catepsina D/genética , Línea Celular Tumoral , Femenino , Humanos , Inmunohistoquímica , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/cirugía , Proteína 2 de la Membrana Asociada a los Lisosomas/biosíntesis , Proteína 2 de la Membrana Asociada a los Lisosomas/genética , Lisosomas/enzimología , Lisosomas/metabolismo , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Fenotipo , Pronóstico , Resultado del Tratamiento
15.
PLoS One ; 10(1): e0116021, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25635828

RESUMEN

PURPOSE: The current study examines the effect of fever-range hyperthermia and mild hypothermia on human cancer cells focusing on cell viability, proliferation and HSP90 expression. MATERIALS AND METHODS: A549 and H1299 lung carcinoma, MCF7 breast adenocarcinoma, U87MG and T98G glioblastoma, DU145 and PC3 prostate carcinoma and MRC5 normal fetal lung fibroblasts cell lines were studied. After 3-day exposure to 34°C, 37°C and 40°C, cell viability was determined. Cell proliferation (ki67 index), apoptosis (Caspase 9) and HSP90 expression was studied by confocal microscopy. RESULTS: Viability/proliferation experiments demonstrated that MRC5 fibroblasts were extremely sensitive to hyperthermia, while they were the most resistant to hypothermia. T98G and A549 were thermo-tolerant, the remaining being thermo-sensitive to a varying degree. Nonetheless, as a universal effect, hypothermia reduced viability/proliferation in all cell lines. Hyperthermia sharply induced Caspase 9 in the U87MG most thermo-sensitive cell line. In T98G and A549 thermo-tolerant cell lines, the levels of Caspase 9 declined. Moreover, hyperthermia strongly induced the HSP90 levels in T98G, whilst a sharp decrease was recorded in the thermo-sensitive PC3 and U87MG cell lines. Hyperthermia sensitized thermo-sensitive cancer cell lines to cisplatin and temozolomide, whilst its sensitizing effect was diminished in thermo-tolerant cell lines. CONCLUSIONS: The existence of thermo-tolerant and thermo-sensitive cancer cell lines was confirmed, which further encourages research to classify human tumor thermic predilection for patient stratification in clinical trials. Of interest, mild hypothermia had a universal suppressing effect on cancer cell proliferation, further supporting the radio-sensitization hypothesis through reduction of oxygen and metabolic demands.


Asunto(s)
Proliferación Celular , Proteínas HSP90 de Choque Térmico/metabolismo , Antineoplásicos Alquilantes/farmacología , Línea Celular Tumoral , Supervivencia Celular , Cisplatino/farmacología , Frío , Respuesta al Choque por Frío , Dacarbazina/análogos & derivados , Dacarbazina/farmacología , Ensayos de Selección de Medicamentos Antitumorales , Respuesta al Choque Térmico , Calor , Humanos , Temozolomida
16.
PLoS One ; 9(7): e102408, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25010689

RESUMEN

OBJECTIVES: Vasculature damage is an important contributor to the side-effects of radiotherapy. The aim of this study is to provide insights into the radiobiology of the autophagic response of endothelial cells. METHODS AND MATERIALS: Human umbilical vascular endothelial cells (HUVEC) were exposed to 2 Gy of ionizing radiation (IR) and studied using confocal microscopy and western blot analysis, at 4 and 8 days post-irradiation. The role of autophagy flux in HUVEC radio-sensitivity was also examined. RESULTS: IR-induced accumulation of LC3A(+), LC3B(+) and p62 cytoplasmic vacuoles, while in double immunostaining with lysosomal markers (LAMP2a and CathepsinD) repression of the autophagolysosomal flux was evident. Autophagy-related proteins (ATF4, HIF1α., HIF2α, Beclin1) were, however, induced excluding an eventual repressive effect of radiation on autophagy initiating protein expression. Exposure of HUVEC to SMER28, an mTOR-independent inducer of autophagy, enhanced proLC3 and LC3A, B-I protein expression and accelerated the autophagic flux. Pre-treatment of HUVEC with SMER28 protected against the blockage of autophagic flux induced by IR and conferred radio-resistance. Suppression of LC3A/LC3B proteins with siRNAs resulted in radio-sensitization. CONCLUSIONS: The current data provide a rationale for the development of novel radioprotection policies targeting the autophagic pathway.


Asunto(s)
Autofagia/genética , Células Endoteliales/efectos de la radiación , Lisosomas/efectos de la radiación , Radiación Ionizante , Compuestos Alílicos/administración & dosificación , Autofagia/efectos de la radiación , Catepsina D/biosíntesis , Citoplasma/efectos de los fármacos , Citoplasma/efectos de la radiación , Citoplasma/ultraestructura , Células Endoteliales/efectos de los fármacos , Células Endoteliales/ultraestructura , Células Endoteliales de la Vena Umbilical Humana , Humanos , Proteína 2 de la Membrana Asociada a los Lisosomas/biosíntesis , Lisosomas/efectos de los fármacos , Lisosomas/ultraestructura , Proteínas de la Membrana/efectos de los fármacos , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/efectos de la radiación , Microscopía Confocal , Quinazolinas/administración & dosificación , ARN Interferente Pequeño
17.
Redox Rep ; 17(5): 181-6, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22889828

RESUMEN

OBJECTIVES: The objective of this study was to investigate the effects of catechin and epicatechin on the activity of the endogenous antioxidant enzymes superoxide dismutase (SOD) and glutathione peroxidase (GPx) (as well as the total antioxidant capacity (TAC)) of rats after intra-peritoneal (i.p.) administration. METHODS: Twenty-four Wistar rats were randomly divided into two groups: the experimental group which was administered daily with a 1:1 mixture of epicatechin and catechin at a concentration of 23 mg/kg body weight for 10 days and the control group which was injected daily with an equal amount of saline. Blood and urine samples were collected before and after the administration period, as well as 10 days after (follow-up). RESULTS: Intra-peritoneal administration of catechins led to a potent decrease in GPx levels and a significant increase in SOD levels. TAC was significantly increased in plasma and urine. Malonaldehyde levels in urine remained stable. In the animals treated with catechins, SOD activity showed a moderate negative correlation with GPx activity. DISCUSSION: Boosting the activity of the antioxidant enzymes could be a potential adjuvant approach for the treatment of the oxidative stress-related diseases.


Asunto(s)
Catequina/farmacología , Glutatión Peroxidasa/metabolismo , Superóxido Dismutasa/metabolismo , Animales , Catequina/administración & dosificación , Activación Enzimática/efectos de los fármacos , Femenino , Inyecciones Intraperitoneales , Distribución Aleatoria , Ratas , Ratas Wistar
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA