Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
J Cardiovasc Pharmacol ; 59(6): 487-99, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22668799

RESUMEN

Nonsteroidal anti-inflammatory drugs (NSAIDs) have come under scrutiny because of the gastrointestinal, renal, and cardiovascular toxicity associated with prolonged use of these drugs. The purpose of this study was to identify molecular targets for NSAIDs related to cellular toxicity with a view to optimize drug efficacy in the clinic. Coronary artery smooth muscle cells and endothelial cells were treated with low (clinically achievable) and high (typically used in preclinical studies) concentrations of celecoxib, NS398, and ibuprofen for 24 hours. NSAIDs-induced gene expression changes were evaluated by microarray analysis and validated by real-time reverse-transcription polymerase chain reaction and western blotting. The functional significance of differentially expressed genes was evaluated by Ingenuity Pathway Analysis. At high concentrations, NSAIDs altered the expression of genes regulating cell proliferation and cell death. NSAIDs also altered genes associated with cardiovascular functions including inflammation, thrombosis, fibrinolysis, coronary artery disease, and hypertension. The gene expression was most impacted by ibuprofen, celecoxib, and NS398, in that order. This study revealed that NSAIDs altered expression of an array of genes associated with cardiovascular events and emphasizes the potential for fingerprinting drugs in preclinical studies to assess the potential drug toxicity and to optimize the drug efficacy in clinical settings.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Vasos Coronarios/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Antiinflamatorios no Esteroideos/administración & dosificación , Western Blotting , Celecoxib , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Vasos Coronarios/citología , Vasos Coronarios/metabolismo , Relación Dosis-Respuesta a Droga , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Perfilación de la Expresión Génica , Humanos , Ibuprofeno/farmacología , Análisis por Micromatrices , Terapia Molecular Dirigida , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Nitrobencenos/administración & dosificación , Nitrobencenos/farmacología , Pirazoles/administración & dosificación , Pirazoles/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sulfonamidas/administración & dosificación , Sulfonamidas/farmacología
2.
Mol Cancer Ther ; 8(1): 261-73, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19139136

RESUMEN

Cyclooxygenase-2 (COX-2) plays a significant role in tumor development and progression. Nonsteroidal anti-inflammatory drugs (NSAID) exhibit potent anticancer effects in vitro and in vivo by COX-2-dependent and COX-2-independent mechanisms. In this study, we used microarray analysis to identify the change of expression profile regulated by a COX-2-specific NSAID NS-398 (0.01 and 0.1 mmol/L), a nonspecific NSAID ibuprofen (0.1 and 1.5 mmol/L) and RNA interference (RNAi)-mediated COX-2 inhibition in PC3 prostate cancer cells. A total of 3,362 differentially expressed genes with 2-fold change and P<0.05 were identified. Low concentrations of NSAIDs and COX-2 RNAi altered very few genes (1-3%) compared with the higher concentration of NS-398 (17%) and ibuprofen (80%). Ingenuity Pathway Analysis was used for distributing the differentially expressed genes into biological networks and for evaluation of functional significance. The top 3 networks for both NSAIDs included functional categories of DNA replication, recombination and repair, and gastrointestinal disease. Immunoresponse function was specific to NS-398, and cell cycle and cellular movement were among the top functions for ibuprofen. Ingenuity Pathway Analysis also identified renal and urologic disease as a function specific for ibuprofen. This comprehensive study identified several COX-2-independent targets of NSAIDs, which may help explain the antitumor and radiosensitizing effects of NSAIDs. However, none of these categories were reflected in the identified networks in PC3 cells treated with clinically relevant low concentrations of NS-398 and ibuprofen or with COX-2 RNAi, suggesting the benefit to fingerprinting preclinical drug concentrations to improve their relevance to the clinical setting.


Asunto(s)
Inhibidores de la Ciclooxigenasa 2/farmacología , Ciclooxigenasa 2/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Ibuprofeno/farmacología , Nitrobencenos/farmacología , Neoplasias de la Próstata/genética , Interferencia de ARN , Sulfonamidas/farmacología , Proteína 4 Similar a la Angiopoyetina , Angiopoyetinas/genética , Angiopoyetinas/metabolismo , Antiinflamatorios no Esteroideos/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Ciclooxigenasa 2/metabolismo , Perfilación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Calor , Humanos , Masculino , Metaloproteinasa 3 de la Matriz/genética , Metaloproteinasa 3 de la Matriz/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología
3.
Int J Cancer ; 123(10): 2430-7, 2008 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-18729192

RESUMEN

Overexpression of hypoxia-inducible factor-1alpha (HIF-1alpha) in human tumors is associated with poor prognosis and poor outcome to radiation therapy. Inhibition of HIF-1alpha is considered as a promising approach in cancer therapy. The purpose of this study was to test the efficacy of a novel HIF-1alpha inhibitor PX-478 as a radiosensitizer under normoxic and hypoxic conditions in vitro. PC3 and DU 145 prostate carcinoma cells were treated with PX-478 for 20 hr, and HIF-1alpha protein level and clonogenic cell survival were determined under normoxia and hypoxia. Effects of PX-478 on cell cycle distribution and phosphorylation of H2AX histone were evaluated. PX-478 decreased HIF-1alpha protein in PC3 and DU 145 cells. PX-478 produced cytotoxicity in both cell lines with enhanced toxicity under hypoxia for DU-145. PX-478 (20 mumol/L) enhanced the radiosensitivity of PC3 cells irradiated under normoxic and hypoxic condition with enhancement factor (EF) 1.4 and 1.56, respectively. The drug was less effective in inhibiting HIF-1alpha and enhancing radiosensitivity of DU 145 cells compared to PC3 cells with EF 1.13 (normoxia) and 1.25 (hypoxia) at 50 mumol/L concentration. PX-478 induced S/G2M arrest in PC3 but not in DU 145 cells. Treatment of PC3 and DU 145 cells with the drug resulted in phosphorylation of H2AX histone and prolongation of gammaH2AX expression in the irradiated cells. PX-478 is now undergoing Phase I clinical trials as an oral agent. Although the precise mechanism of enhancement of radiosensitivity remains to be identified, this study suggests a potential role for PX-478 as a clinical radiation enhancer.


Asunto(s)
Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Compuestos de Mostaza/farmacología , Fenilpropionatos/farmacología , Neoplasias de la Próstata/patología , Tolerancia a Radiación , Western Blotting , Ciclo Celular/efectos de los fármacos , Hipoxia de la Célula , Línea Celular Tumoral , Supervivencia Celular , Técnica del Anticuerpo Fluorescente , Histonas/metabolismo , Humanos , Masculino , Fosforilación
4.
Mol Cancer Ther ; 17(2): 355-367, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28802252

RESUMEN

Implementing targeted drug therapy in radio-oncologic treatment regimens has greatly improved the outcome of cancer patients. However, the efficacy of molecular targeted drugs such as inhibitory antibodies or small molecule inhibitors essentially depends on target expression and activity, which both can change during the course of treatment. Radiotherapy has previously been shown to activate prosurvival pathways, which can help tumor cells to adapt and thereby survive treatment. Therefore, we aimed to identify changes in signaling induced by radiation and evaluate the potential of targeting these changes with small molecules to increase the therapeutic efficacy on cancer cell survival. Analysis of "The Cancer Genome Atlas" database disclosed a significant overexpression of AKT1, AKT2, and MTOR genes in human prostate cancer samples compared with normal prostate gland tissue. Multifractionated radiation of three-dimensional-cultured prostate cancer cell lines with a dose of 2 Gy/day as a clinically relevant schedule resulted in an increased protein phosphorylation and enhanced protein-protein interaction between AKT and mTOR, whereas gene expression of AKT, MTOR, and related kinases was not altered by radiation. Similar results were found in a xenograft model of prostate cancer. Pharmacologic inhibition of mTOR/AKT signaling after activation by multifractionated radiation was more effective than treatment prior to radiotherapy. Taken together, our findings provide a proof-of-concept that targeting signaling molecules after activation by radiotherapy may be a novel and promising treatment strategy for cancers treated with multifractionated radiation regimens such as prostate cancer to increase the sensitivity of tumor cells to molecular targeted drugs. Mol Cancer Ther; 17(2); 355-67. ©2017 AACRSee all articles in this MCT Focus section, "Developmental Therapeutics in Radiation Oncology."


Asunto(s)
Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/radioterapia , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Animales , Benzoxazoles/farmacología , Línea Celular Tumoral , Resistencia a Antineoplásicos , Humanos , Masculino , Ratones , Ratones Desnudos , Piperazinas/farmacología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Pirimidinas/farmacología , Distribución Aleatoria , Transducción de Señal/efectos de la radiación , Serina-Treonina Quinasas TOR/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Clin Cancer Res ; 11(19 Pt 1): 6980-6, 2005 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-16203791

RESUMEN

PURPOSE: Cyclooxygenase-2 (COX-2) is considered a potential target for cancer therapy, because COX-2 levels are elevated in the majority of human tumors compared with the normal tissues. COX-2 inhibitors inhibit tumor growth and enhance radiation response in vitro as well as in vivo. However, the precise role of COX-2 in radiation response is not clear. The purpose of the present study was to investigate the in vitro radiosensitivity of tumor cells as a function of COX-2 expression. EXPERIMENTAL DESIGN AND RESULTS: PC3 and HeLa cells express COX-2 protein constitutively. We silenced the COX-2 gene in these cells using small interfering RNA (siRNA). Transfection of PC3 cells with 100 nmol/L siRNA targeted against COX-2 resulted in reduction of COX-2 protein by 75% and inhibition of arachidonic acid-induced prostaglandin E2 synthesis by approximately 50% compared with the vehicle control. In HeLa cells, 100 nmol/L COX-2 siRNA inhibited COX-2 protein expression by 80%. Cell cycle analysis showed that transfection with COX-2 siRNA did not alter the cell cycle distribution. Radiosensitivity was determined by clonogenic cell survival assay. There was no significant difference in the radiosensitivity of cells in which COX-2 was silenced compared with the cells transfected vehicle or with negative control siRNAs (enhancement ratio = 1.1). CONCLUSIONS: These data indicate that the in vitro radiosensitivity of tumor cells is minimally dependent on the cellular COX-2 status. Given that a number of potential mechanisms are attributed to COX-2 inhibitors for radiosensitization, specific intervention of COX-2 by RNA interference could help elucidate the precise role of COX-2 in cancer therapy and to optimize strategies for COX-2 inhibition.


Asunto(s)
Carcinoma/radioterapia , Ciclooxigenasa 2/metabolismo , Neoplasias de la Próstata/radioterapia , ARN Interferente Pequeño/metabolismo , Western Blotting , Ciclo Celular , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Inhibidores de la Ciclooxigenasa/farmacología , Dinoprostona/metabolismo , Relación Dosis-Respuesta en la Radiación , Silenciador del Gen , Células HeLa , Humanos , Técnicas para Inmunoenzimas , Masculino , Nitrobencenos/farmacología , Prostaglandinas/metabolismo , Neoplasias de la Próstata/metabolismo , Interferencia de ARN , Tolerancia a Radiación , Sulfonamidas/farmacología , Transfección
6.
Clin Cancer Res ; 9(8): 3150-7, 2003 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-12912967

RESUMEN

PURPOSE: Hypoxia-inducible factors HIF-1alpha and HIF-2alpha are considered to be potential targets for antineoplastic therapy because they regulate the expression of genes that contribute to tumor cell survival, aggressiveness, and angiogenesis. Nonsteroidal anti-inflammatory drugs (NSAIDs) have gained considerable interest as anticancer agents because of their cytotoxic and antiangiogenic properties. The aim of this study was to investigate whether NSAIDs inhibit HIFs and HIF-regulated gene expression in prostate cancer cells. EXPERIMENTAL DESIGN: PC3 and DU-145 cells were treated with ibuprofen (Ibu) and other NSAIDs under normoxic and hypoxic (95% N(2), 5% CO(2); <10 ppm O(2)) conditions. The effect of NSAIDs on HIF proteins was analyzed by Western blot analysis. HIF-regulated proteins, vascular endothelial growth factor (VEGF) and glucose transporter-1 (Glut-1), were analyzed by ELISA and Western blot analysis, respectively. RESULTS: Exposure of PC3 and DU-145 cells to hypoxic condition up-regulated HIF-1alpha and HIF-2alpha proteins. Treatment with Ibu under normoxic and hypoxic conditions reduced the level of HIF-1alpha and HIF-2alpha. Ibu-mediated down-regulation of HIFs was associated with down-regulation of HIF-regulated proteins VEGF and Glut-1 in cells exposed to hypoxia. Other nonspecific NSAIDs, diclofenac and ketorolac, also inhibited HIF-1alpha and HIF-2alpha. The reduction in HIFs was observed in PC3 cells that expressed cyclooxygenase-2 (COX-2) protein as well as in DU-145 cells, which did not express COX-2 protein. COX-2-specific inhibitor NS-398 did not inhibit HIF-1alpha or VEGF and GLUT-1. CONCLUSIONS: These data indicate that one of the effects of NSAIDs is to reduce HIF protein levels. The inhibition of HIFs by NSAIDs was COX-2 independent.


Asunto(s)
Ibuprofeno/uso terapéutico , Neoplasias de la Próstata/metabolismo , Transactivadores/biosíntesis , Factores de Transcripción/biosíntesis , Antiinflamatorios no Esteroideos/uso terapéutico , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Línea Celular Tumoral , Ciclooxigenasa 2 , Inhibidores de la Ciclooxigenasa 2 , Inhibidores de la Ciclooxigenasa/uso terapéutico , Diclofenaco/uso terapéutico , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica , Transportador de Glucosa de Tipo 1 , Humanos , Hipoxia , Subunidad alfa del Factor 1 Inducible por Hipoxia , Isoenzimas/biosíntesis , Ketorolaco/uso terapéutico , Masculino , Proteínas de la Membrana , Proteínas de Transporte de Monosacáridos/biosíntesis , Nitrobencenos/uso terapéutico , Prostaglandina-Endoperóxido Sintasas/biosíntesis , Sulfonamidas/uso terapéutico , Factores de Tiempo , Regulación hacia Arriba , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Factor A de Crecimiento Endotelial Vascular/metabolismo
7.
Clin Cancer Res ; 10(12 Pt 1): 4158-64, 2004 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-15217953

RESUMEN

PURPOSE: Tumor hypoxia is a major limiting factor for radiation therapy. Hypoxia-inducible factors (HIFs) are overexpressed in several human cancers and are considered prognostic markers and potential targets for cancer therapy. The purpose of the present study was to investigate the impact of HIFs on radiosensitivity. EXPERIMENTAL DESIGN: Renal clear cell carcinoma (RCC) cell lines overexpressing HIFs under normoxic conditions because of inactivation of von Hippel-Lindau tumor suppressor gene function (VHL-ve) and their matched pairs in which overexpression of HIFs was abolished by expression of functional VHL (VHL+ve) were irradiated. Radiosensitivity was determined by clonogenic assay. HIF and VHL protein levels were evaluated by Western blot analysis. RCC cells were also treated with ibuprofen, a radiosensitizer and HIF inhibitor in prostate cancer cells. The effect of ibuprofen on radiosensitization and HIF and VHL proteins was compared in RCC matched-pair cell lines. RESULTS: The data showed only small differences in the radiosensitivity between the cells overexpressing HIFs and cells with basal HIF levels. The dose-modifying factors for C2, 786-0, and A498 RCC cells were 1.14, 1.14 and 1.15, respectively. Radiation did not alter HIF or VHL protein levels. Ibuprofen inhibited HIFs in VHL+ve cells expressing basal levels of HIFs. In VHL-ve cells overexpressing HIFs, the inhibition was very modest. Ibuprofen radiosensitized C2 RCC cells to the same extent irrespective of their HIF status. CONCLUSIONS: Overexpression of HIFs in RCC cells harboring VHL mutations has only a modest effect on the radiosensitivity. Radiosensitization by ibuprofen appears to be independent of HIF status.


Asunto(s)
Carcinoma/tratamiento farmacológico , Carcinoma/radioterapia , Ibuprofeno/farmacología , Neoplasias Renales/tratamiento farmacológico , Neoplasias Renales/radioterapia , Transactivadores/fisiología , Factores de Transcripción/fisiología , Proteínas Supresoras de Tumor/fisiología , Ubiquitina-Proteína Ligasas/fisiología , Antiinflamatorios no Esteroideos/farmacología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Western Blotting , Línea Celular , Supervivencia Celular , Relación Dosis-Respuesta a Droga , Relación Dosis-Respuesta en la Radiación , Ensayo de Inmunoadsorción Enzimática , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia , Immunoblotting , Mutación , Oxígeno/metabolismo , Pronóstico , Tolerancia a Radiación , Factores de Tiempo , Transactivadores/genética , Factores de Transcripción/genética , Transfección , Proteínas Supresoras de Tumor/genética , Ubiquitina-Proteína Ligasas/genética , Regulación hacia Arriba , Factor A de Crecimiento Endotelial Vascular/metabolismo , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau
8.
Mol Cancer Res ; 12(7): 1002-15, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24784841

RESUMEN

UNLABELLED: Although modern radiotherapy technologies can precisely deliver higher doses of radiation to tumors, thus, reducing overall radiation exposure to normal tissues, moderate dose, and normal tissue toxicity still remains a significant limitation. The present study profiled the global effects on transcript and miR expression in human coronary artery endothelial cells using single-dose irradiation (SD, 10 Gy) or multifractionated irradiation (MF, 2 Gy × 5) regimens. Longitudinal time points were collected after an SD or final dose of MF irradiation for analysis using Agilent Human Gene Expression and miRNA microarray platforms. Compared with SD, the exposure to MF resulted in robust transcript and miR expression changes in terms of the number and magnitude. For data analysis, statistically significant mRNAs (2-fold) and miRs (1.5-fold) were processed by Ingenuity Pathway Analysis to uncover miRs associated with target transcripts from several cellular pathways after irradiation. Interestingly, MF radiation induced a cohort of mRNAs and miRs that coordinate the induction of immune response pathway under tight regulation. In addition, mRNAs and miRs associated with DNA replication, recombination and repair, apoptosis, cardiovascular events, and angiogenesis were revealed. IMPLICATIONS: Radiation-induced alterations in stress and immune response genes in endothelial cells contribute to changes in normal tissue and tumor microenvironment, and affect the outcome of radiotherapy.


Asunto(s)
Células Endoteliales/metabolismo , Células Endoteliales/efectos de la radiación , MicroARNs/biosíntesis , Apoptosis , Fraccionamiento de la Dosis de Radiación , Relación Dosis-Respuesta en la Radiación , Perfilación de la Expresión Génica , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Análisis por Micromatrices , Estrés Fisiológico/inmunología
9.
Radiat Res ; 182(2): 139-48, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25003313

RESUMEN

To understand the impact of clinically relevant radiation therapy (RT) on tumor immune gene expression and to utilize the changes that occur during treatment to improve cancer treatment outcome, we examined how immune response genes are modulated in prostate cancer cells of varying p53 status. LNCaP (p53 wild-type), PC3 (p53 null) and DU145 (p53 mutant) cells received a 10 Gy single dose or 1 Gy × 10 multifractionated radiation dose to simulate hypofractionated and conventionally fractionated prostate radiotherapy. Total RNA was isolated 24 h after multifractionated radiation treatment and single-dose treatments and subjected to microarray analysis and later validated by RT-PCR. RT-PCR was utilized to identify total-dose inflection points for significantly upregulated genes in response to multifractionated radiation therapy. Radiation-induced damage-associated molecular pattern molecules (DAMPs) and cytokine analyses were performed using bioluminescence and ELISA. Multifractionated doses activated immune response genes more robustly than single-dose treatment, with a relatively larger number of immune genes upregulated in PC3 compared to DU145 and LNCaP cells. The inflection point of multifractionated radiation-induced immune genes in PC3 cells was observed in the range of 8-10 Gy total radiation dose. Although both multifractionated and single-dose radiation-induced proinflammatory DAMPs and positively modulated the cytokine environment, the changes were of higher magnitude with multifractionated therapy. The findings of this study together with the gene expression data suggest that cells subjected to multifractionated radiation treatment would promote productive immune cell-tumor cell interactions.


Asunto(s)
Inmunidad/efectos de la radiación , Neoplasias de la Próstata/patología , Radioterapia/métodos , Línea Celular Tumoral , Citocinas/metabolismo , Fraccionamiento de la Dosis de Radiación , Humanos , Inmunidad/genética , Masculino , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/inmunología , Neoplasias de la Próstata/radioterapia , Transcriptoma/inmunología , Transcriptoma/efectos de la radiación , Resultado del Tratamiento
10.
Mol Cancer Res ; 11(1): 5-12, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23175523

RESUMEN

Radiation oncology modalities such as intensity-modulated and image-guided radiation therapy can reduce the high dose to normal tissue and deliver a heterogeneous dose to tumors, focusing on areas deemed at highest risk for tumor persistence. Clinical radiation oncology produces daily doses ranging from 1 to 20 Gy, with tissues being exposed to 30 or more daily fractions. Hypothesizing the cells that survive fractionated radiation therapy have a substantially different phenotype than the untreated cells, which might be exploitable for targeting with molecular therapeutics or immunotherapy, three prostate cancer cell lines (PC3, DU145, and LNCaP) and normal endothelial cells were studied to understand the biology of differential effects of multifraction (MF) radiation of 0.5, 1, and/or 2 Gy fraction to 10 Gy total dose, and a single dose of 5 and 10 Gy. The resulting changes in mRNA, miRNA, and phosphoproteome were analyzed. Significant differences were observed in the MF radiation exposures including those from the 0.5 Gy MF that produces little cell killing. As expected, p53 function played a major role in response. Pathways modified by MF include immune response, DNA damage, cell-cycle arrest, TGF-ß, survival, and apoptotic signal transduction. The radiation-induced stress response will set forth a unique platform for exploiting the effects of radiation therapy as "focused biology" for cancer treatment in conjunction with molecular targeted or immunologically directed therapy. Given that more normal tissue is treated, albeit to lower doses with these newer techniques, the response of the normal tissue may also influence long-term treatment outcome.


Asunto(s)
Neoplasias/radioterapia , Línea Celular Tumoral , Relación Dosis-Respuesta en la Radiación , Humanos , Neoplasias/genética , Tolerancia a Radiación , Radioterapia Guiada por Imagen , Sobrevivientes
11.
Transl Oncol ; 6(5): 573-85, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24151538

RESUMEN

We assessed changes in cell lines of varying p53 status after various fractionation regimens to determine if p53 influences gene expression and if multifractionated (MF) irradiation can induce molecular pathway changes. LNCaP (p53 wild-type), PC3 (p53 null), and DU145 (p53 mutant) prostate carcinoma cells received 5 and 10 Gy as single-dose (SD) or MF (0.5 Gy x 10, 1 Gy x 10, and 2 Gy x 5) irradiation to simulate hypofractionated and conventionally fractionated prostate radiotherapies, respectively. mRNA analysis revealed 978 LNCaP genes differentially expressed (greater than two-fold change, P < .05) after irradiation. Most were altered with SD (69%) and downregulated (75%). Fewer PC3 (343) and DU145 (116) genes were induced, with most upregulated (87%, 89%) and altered with MF irradiation. Gene ontology revealed immune response and interferon genes most prominently expressed after irradiation in PC3 and DU145. Cell cycle regulatory (P = 9.23 x 10(-73), 14.2% of altered genes, nearly universally downregulated) and DNA replication/repair (P = 6.86 x 10(-30)) genes were most prominent in LNCaP. Stress response and proliferation genes were altered in all cell lines. p53-activated genes were only induced in LNCaP. Differences in gene expression exist between cell lines and after varying irradiation regimens that are p53 dependent. As the duration of changes is ≥24 hours, it may be possible to use radiation-inducible targeted therapy to enhance the efficacy of molecular targeted agents.

12.
Radiat Res ; 178(3): 105-17, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22827214

RESUMEN

We have previously demonstrated that prostate carcinoma cells exposed to fractionated radiation differentially expressed more genes compared to single-dose radiation. To understand the role of miRNA in regulation of radiation-induced gene expression, we analyzed miRNA expression in LNCaP, PC3 and DU145 prostate cancer cells treated with single-dose radiation and fractionated radiation by microarray. Selected miRNAs were studied in RWPE-1 normal prostate epithelial cells by RT-PCR. Fractionated radiation significantly altered more miRNAs as compared to single-dose radiation. Downregulation of oncomiR-17-92 cluster was observed only in the p53 positive LNCaP and RWPE-1 cells treated with single-dose radiation and fractionated radiation. Comparison of miRNA and mRNA data by IPA target filter analysis revealed an inverse correlation between miR-17-92 cluster and several targets including TP53INP1 in p53 signaling pathway. The base level expressions of these miRNAs were significantly different among the cell lines and did not predict the radiation outcome. Tumor suppressor miR-34a and let-7 miRNAs were upregulated by fractionated radiation in radiosensitive LNCaP (p53 positive) and PC3 (p53-null) cells indicating that radiation-induced miRNA expression may not be regulated by p53 alone. Our data support the potential for using fractionated radiation to induce molecular targets and radiation-induced miRNAs may have a significant role in predicting radiosensitivity.


Asunto(s)
Fraccionamiento de la Dosis de Radiación , MicroARNs/genética , MicroARNs/metabolismo , Neoplasias de la Próstata/patología , Línea Celular Tumoral , Células Epiteliales/metabolismo , Células Epiteliales/efectos de la radiación , Calor , Humanos , Inmunidad Innata/genética , Inmunidad Innata/efectos de la radiación , Masculino , Tolerancia a Radiación/genética , Tolerancia a Radiación/efectos de la radiación , Reproducibilidad de los Resultados , Transcriptoma/efectos de la radiación
13.
Radiat Res ; 174(4): 446-58, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20726711

RESUMEN

To examine the possibility of using fractionated radiation in a unique way with molecular targeted therapy, gene expression profiles of prostate carcinoma cells treated with 10 Gy radiation administered either as a single dose or as fractions of 2 Gy × 5 and 1 Gy × 10 were examined by microarray analysis. Compared to the single dose, the fractionated irradiation resulted in significant increases in differentially expressed genes in both cell lines, with more robust changes in PC3 cells than in DU145 cells. The differentially expressed genes (>twofold change; P < 0.05) were clustered and their ontological annotations evaluated. In PC3 cells genes regulating immune and stress response, cell cycle and apoptosis were significantly up-regulated by multifractionated radiation compared to single-dose radiation. Ingenuity Pathway Analysis (IPA) of the differentially expressed genes revealed that immune response and cardiovascular genes were in the top functional category in PC3 cells and cell-to-cell signaling in DU145 cells. RT-PCR analysis showed that a flexure point for gene expression occurred at the 6th-8th fraction and AKT inhibitor perifosine produced enhanced cell killing after 1 Gy × 8 fractionated radiation in PC3 and DU145 cells compared to single dose. This study suggests that fractionated radiation may be a uniquely exploitable, non-oncogene-addiction stress pathway for molecular therapeutic targeting.


Asunto(s)
Fraccionamiento de la Dosis de Radiación , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/radioterapia , Apoptosis/efectos de la radiación , Ciclo Celular/efectos de la radiación , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Perfilación de la Expresión Génica , Histonas/metabolismo , Humanos , Masculino , Análisis de Secuencia por Matrices de Oligonucleótidos , Fenotipo , Fosforilación/efectos de la radiación , Fosforilcolina/análogos & derivados , Fosforilcolina/farmacología , Fosforilcolina/uso terapéutico , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/genética , Dosificación Radioterapéutica , Reproducibilidad de los Resultados , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA