Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
J Biol Chem ; 290(41): 24858-74, 2015 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-26311851

RESUMO

Modulating tissue responses to stress is an important therapeutic objective. Oxidative and genotoxic stresses caused by ionizing radiation are detrimental to healthy tissues but beneficial for treatment of cancer. CD47 is a signaling receptor for thrombospondin-1 and an attractive therapeutic target because blocking CD47 signaling protects normal tissues while sensitizing tumors to ionizing radiation. Here we utilized a metabolomic approach to define molecular mechanisms underlying this radioprotective activity. CD47-deficient cells and cd47-null mice exhibited global advantages in preserving metabolite levels after irradiation. Metabolic pathways required for controlling oxidative stress and mediating DNA repair were enhanced. Some cellular energetics pathways differed basally in CD47-deficient cells, and the global declines in the glycolytic and tricarboxylic acid cycle metabolites characteristic of normal cell and tissue responses to irradiation were prevented in the absence of CD47. Thus, CD47 mediates signaling from the extracellular matrix that coordinately regulates basal metabolism and cytoprotective responses to radiation injury.


Assuntos
Antígeno CD47/metabolismo , Redes e Vias Metabólicas/efeitos da radiação , Tolerância a Radiação , Animais , Antígeno CD47/genética , Ciclo do Ácido Cítrico/efeitos da radiação , Metabolismo Energético/efeitos da radiação , Deleção de Genes , Homeostase/efeitos da radiação , Humanos , Células Jurkat , Metabolômica , Camundongos , Nucleotídeos/biossíntese , Estresse Oxidativo/efeitos da radiação , Via de Pentose Fosfato/efeitos da radiação
2.
Am J Pathol ; 173(4): 1100-12, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18787106

RESUMO

Radiation, a primary mode of cancer therapy, acutely damages cellular macromolecules and DNA and elicits stress responses that lead to cell death. The known cytoprotective activity of nitric oxide (NO) is blocked by thrombospondin-1, a potent antagonist of NO/cGMP signaling in ischemic soft tissues, suggesting that thrombospondin-1 signaling via its receptor CD47 could correspondingly increase radiosensitivity. We show here that soft tissues in thrombospondin-1-null mice are remarkably resistant to radiation injury. Twelve hours after 25-Gy hindlimb irradiation, thrombospondin-1-null mice showed significantly less cell death in both muscle and bone marrow. Two months after irradiation, skin and muscle units in null mice showed minimal histological evidence of radiation injury and near full retention of mitochondrial function. Additionally, both tissue perfusion and acute vascular responses to NO were preserved in irradiated thrombospondin-1-null hindlimbs. The role of thrombospondin-1 in radiosensitization is specific because thrombospondin-2-null mice were not protected. However, mice lacking CD47 showed radioresistance similar to thrombospondin-1-null mice. Both thrombospondin-1- and CD47-dependent radiosensitization is cell autonomous because vascular cells isolated from the respective null mice showed dramatically increased survival and improved proliferative capacity after irradiation in vitro. Therefore, thrombospondin-1/CD47 antagonists may have selective radioprotective activity for normal tissues.


Assuntos
Antígeno CD47/metabolismo , Lesões Experimentais por Radiação/metabolismo , Lesões Experimentais por Radiação/patologia , Trombospondina 1/metabolismo , Sobrevivência de Tecidos/efeitos da radiação , Animais , Apoptose/efeitos da radiação , Vasos Sanguíneos/patologia , Vasos Sanguíneos/efeitos da radiação , Proliferação de Células/efeitos da radiação , Sobrevivência Celular/efeitos da radiação , Células Cultivadas , Células Endoteliais/patologia , Células Endoteliais/efeitos da radiação , Membro Posterior/patologia , Membro Posterior/efeitos da radiação , Hipertrofia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Transplante de Neoplasias , Neoplasias/patologia , Tolerância a Radiação/efeitos da radiação , Trombospondinas/metabolismo , Raios X
3.
Cancer Res ; 67(8): 3845-52, 2007 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-17440099

RESUMO

Studies were conducted to determine whether gene expression profiles following a single dose of radiation would yield equivalent profiles following fractionated radiation in different tumor cell lines. MCF7 (breast), DU145 (prostate), and SF539 (gliosarcoma) cells were exposed to a total radiation dose of 10 Gy administered as a single dose (SD) or by daily multifractions (MF) of 5 x 2 Gy. Following radiation treatment, mRNA was isolated at 1, 4, 10, and 24 h and processed for cDNA microarray analysis. To determine the influence of the tumor microenvironment on gene expression, one cell type (DU145) was evaluated growing as a solid tumor in athymic nude mice for both radiation protocols. Unsupervised hierarchical cluster map analysis showed significant differences in gene expression profiles between SD and MF treatments for cells treated in vitro, with MF yielding a more robust induction compared with SD. Several genes were uniquely up-regulated by MF treatment, including multiple IFN-related genes (STAT1, G1P2, OAS1, OAS3, G1P3, IFITM1) and TGF-beta-associated genes (EGR1, VEGF, THBS1, and TGFB2). DU145 cells grown in vivo exhibited a completely different set of genes induced by both SD and MF compared with the same cells exposed in vitro. The results of the study clearly show distinct differences in the molecular response of cells between SD and MF radiation exposures and show that the tumor microenvironment can significantly influence the pattern of gene expression after radiation exposures.


Assuntos
Neoplasias da Mama/genética , Neoplasias da Mama/radioterapia , Glioma/genética , Glioma/radioterapia , Neoplasias da Próstata/genética , Neoplasias da Próstata/radioterapia , Animais , Neoplasias da Mama/metabolismo , Ciclo Celular/efeitos da radiação , Linhagem Celular Tumoral , Análise por Conglomerados , Fracionamento da Dose de Radiação , Feminino , Expressão Gênica/efeitos da radiação , Perfilação da Expressão Gênica , Genes p53/efeitos da radiação , Glioma/metabolismo , Humanos , Interferons/biossíntese , Interferons/genética , Masculino , Camundongos , Camundongos Nus , Transplante de Neoplasias , Análise de Sequência com Séries de Oligonucleotídeos , Neoplasias da Próstata/metabolismo , Fator de Crescimento Transformador beta/genética , Transplante Heterólogo , Regulação para Cima/efeitos da radiação
4.
Int J Cancer ; 123(10): 2430-7, 2008 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-18729192

RESUMO

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.


Assuntos
Subunidade alfa do Fator 1 Induzível por Hipóxia/antagonistas & inibidores , Compostos de Mostarda/farmacologia , Fenilpropionatos/farmacologia , Neoplasias da Próstata/patologia , Tolerância a Radiação , Western Blotting , Ciclo Celular/efeitos dos fármacos , Hipóxia Celular , Linhagem Celular Tumoral , Sobrevivência Celular , Imunofluorescência , Histonas/metabolismo , Humanos , Masculino , Fosforilação
5.
J Dermatol Sci ; 48(1): 35-42, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17624738

RESUMO

BACKGROUND: Transforming growth factor-beta1 (TGF-beta1), a key biological mediator following ionizing radiation, plays a role in a complex tissue reaction involved in local radiation-induced pathological damage. Knocking out Smad3 (S3KO), a downstream signaling intermediate in the TGF-beta pathway, in mice protects their skin from radiation damage as demonstrated by decreased epithelial acanthosis and dermal fibrosis as compared to Smad3 wild-type (S3WT) mice. OBJECTIVE: The present study was designed to investigate the molecular mechanisms contributing to increased radioprotection in the absence of Smad3. METHODS: Primary dermal fibroblasts derived from S3WT and KO mice were exposed to 5Gy ionizing radiation in vitro. Western blot analyses, immunocytochemistry, and reporter transfections were used to dissect the radiation-induced events. RESULTS: There was increased phosphorylation of ERK-MAPK, p53 and H2A.X in S3KO compared to the S3WT fibroblasts, implicating them in a key signaling cascade in response of these cells to radiation. Pro-fibrotic gene expression was decreased in S3KO fibroblasts post-irradiation. CONCLUSION: The absence of Smad3 may decrease radio-responsiveness by increasing activation of DNA damage sensing mechanisms and decreasing induction of pro-fibrotic genes.


Assuntos
Derme/citologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fibroblastos/metabolismo , Fibroblastos/efeitos da radiação , Radiação Ionizante , Proteína Smad3/metabolismo , Animais , Proliferação de Células/efeitos da radiação , Células Cultivadas , Dano ao DNA/efeitos da radiação , Histonas/metabolismo , Camundongos , Camundongos Knockout , Lesões por Radiação/prevenção & controle , Proteína Smad3/genética , Transfecção , Proteína Supressora de Tumor p53/metabolismo
6.
Cancer Res ; 65(2): 579-86, 2005 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-15695402

RESUMO

Despite significant evidence of a role of hypoxia in cellular resistance to ionizing radiation-induced toxicity, the underlying molecular mechanisms remain unclear. This study focused on the influence of hypoxia on radiation-induced signals in TK6 human lymphoblastoid cells. Hypoxic (<10 ppm oxygen) and aerobic cells were exposed to equilethal doses of ionizing radiation, radiation dose ratio, 3:1 (hypoxia:air). Hypoxia alone or radiation treatment under aerobic or hypoxic conditions led to increased levels of phospho-p44/42 mitogen-activated protein kinase. Levels of phospho-p38 mitogen-activated protein kinase did not change as a result of either hypoxia or irradiation. Hypoxia alone had no effect on expression of phospho-stress-activated protein kinase (SAPK), wild-type p53, or cleaved caspase 3. Irradiation under aerobic conditions resulted in an increase in the phospho-SAPK signal, whereas hypoxia suppressed the irradiation-induced increase in the level of phospho-SAPK. Both hypoxic and aerobic cells showed increases in p53 levels in response to radiation. Hypoxia blocked radiation-induced cleavage of caspase 3 and poly-ADP-ribose polymerase. Irradiation of aerobic and hypoxic TK6 cells using 6 and 18 Gy, respectively, resulted in a similar and significant increase in fraction of apoptotic cells within 24 hours postirradiation. In contrast, basal levels of apoptosis were observed at 24 hours postirradiation in aerobic and hypoxic NH32 cells, a p53 null derivative of TK6 cells. These results suggest that radiation-induced apoptosis under hypoxia occurs independent of phospho-SAPK and caspase 3, and the p53 response is an obligatory apoptotic signal in TK6 cells.


Assuntos
Caspases/metabolismo , Linfócitos/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos da radiação , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Aerobiose , Apoptose/fisiologia , Apoptose/efeitos da radiação , Caspase 3 , Caspases/efeitos da radiação , Hipóxia Celular/fisiologia , Linhagem Celular , Citometria de Fluxo , Humanos , Linfócitos/enzimologia , Linfócitos/efeitos da radiação , Sistema de Sinalização das MAP Quinases/fisiologia , Proteína Quinase 1 Ativada por Mitógeno/efeitos da radiação , Proteína Quinase 3 Ativada por Mitógeno/efeitos da radiação , Fosforilação/efeitos da radiação , Poli(ADP-Ribose) Polimerases/metabolismo , Proteína Supressora de Tumor p53/biossíntese , Proteína Supressora de Tumor p53/genética
7.
Antioxid Redox Signal ; 8(7-8): 1263-72, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16910774

RESUMO

SCCVII tumor cells that grow in vitro or in vivo as a solid tumor were used to compare and contrast geneexpression profiles with or without exposure to two doses of ionizing radiation. Exponentially growing SCCVII cell cultures and tumors (1 cm diameter) were treated with 0, 2, or 10 Gy, and RNA was collected 1, 3, 6, 12, and 24 h after treatment. Growth under in vitro conditions increased the expression of genes associated with the unfolded protein response (UPR) including ATF4, Ero-1 like, and cystathionase. Growth in vivo indicated that the HIF-1a genes were not upregulated, whereas genes such as hemoglobin alpha and crystallin alpha B were significantly upregulated. Ninety genes of 16K were found to be significantly modulated under either growth condition by radiation treatment. Gene expression was not dose dependent. Sixty percent of these genes exhibited similar modulation under both in vitro and in vivo conditions; however, 29% of the genes were modulated by radiation under in vivo conditions only. Gene-expression profiles for the same tumor cells can differ, dependent on growth conditions, underscoring the influence that the tumor microenvironment exerts on gene expression for both growth of solid tumors and their response to radiation treatment.


Assuntos
Carcinoma de Células Escamosas/radioterapia , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Neoplasias Experimentais/radioterapia , Ensaios Antitumorais Modelo de Xenoenxerto , Animais , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patologia , Técnicas de Cultura de Células , Ciclo Celular/efeitos da radiação , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos da radiação , Feminino , Camundongos , Camundongos Endogâmicos C3H , Transplante de Neoplasias , Neoplasias Experimentais/genética , Neoplasias Experimentais/patologia , Doses de Radiação , Radiação Ionizante , Fatores de Tempo , Proteína Supressora de Tumor p53/análise
8.
Cancer Res ; 62(21): 6246-54, 2002 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-12414654

RESUMO

Global gene expression patterns in breast cancer cells after treatment with oxidants (hydrogen peroxide, menadione, and t-butyl hydroperoxide) were investigated in three replicate experiments. RNA collected after treatment (at 1, 3, 7, and 24 h) rather than after a single time point, enabled an analysis of gene expression patterns. Using a 17,000 microarray, template-based clustering and multidimensional scaling analysis of the gene expression over the entire time course identified 421 genes as being either up- or down-regulated by the three oxidants. In contrast, only 127 genes were identified for any single time point and a 2-fold change criteria. Surprisingly, the patterns of gene induction were highly similar among the three oxidants; however, differences were observed, particularly with respect to p53, IL-6, and heat-shock related genes. Replicate experiments increased the statistical confidence of the study, whereas changes in gene expression patterns over a time course demonstrated significant additional information versus a single time point. Analyzing the three oxidants simultaneously by template cluster analysis identified genes that heretofore have not been associated with oxidative stress.


Assuntos
Neoplasias da Mama/genética , Expressão Gênica/efeitos dos fármacos , Oxidantes/farmacologia , Neoplasias da Mama/metabolismo , Análise por Conglomerados , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Peróxido de Hidrogênio/farmacologia , Análise de Sequência com Séries de Oligonucleotídeos , Estresse Oxidativo/genética , Reprodutibilidade dos Testes , Ativação Transcricional , Células Tumorais Cultivadas , Vitamina K 3/farmacologia , terc-Butil Hidroperóxido/farmacologia
9.
Biochim Biophys Acta ; 1573(2): 109-20, 2002 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-12399020

RESUMO

Modulation of radiation- and metal ion-catalyzed oxidative-induced damage using plasmid DNA, genomic DNA, and cell survival, by three nitroxides and their corresponding hydroxylamines, were examined. The antioxidant property of each compound was independently determined by reacting supercoiled DNA with copper II/1,10-phenanthroline complex fueled by the products of hypoxanthine/xanthine oxidase (HX/XO) and noting the protective effect as assessed by agarose gel electrophoresis. The nitroxides and their corresponding hydroxylamines protected approximately to the same degree (33-47% relaxed form) when compared to 76.7% relaxed form in the absence of protectors. Likewise, protection by both the nitroxide and corresponding hydroxylamine were observed for Chinese hamster V79 cells exposed to hydrogen peroxide. In contrast, when plasmid DNA damage was induced by ionizing radiation (100 Gy), only nitroxides (10 mM) provide protection (32.4-38.5% relaxed form) when compared to radiation alone or in the presence of hydroxylamines (10 mM) (79.8% relaxed form). Nitroxide protection was concentration dependent. Radiation cell survival studies and DNA double-strand break (DBS) assessment (pulse field electrophoresis) showed that only the nitroxide protected or prevented damage, respectively. Collectively, the results show that nitroxides and hydroxylamines protect equally against the damage mediated by oxidants generated by the metal ion-catalyzed Haber-Weiss reaction, but only nitroxides protect against radiation damage, suggesting that nitroxides may more readily react with intermediate radical species produced by radiation than hydroxylamines.


Assuntos
Antioxidantes/farmacologia , Dano ao DNA , Hidroxilaminas/farmacologia , Óxidos de Nitrogênio/farmacologia , Protetores contra Radiação/farmacologia , Animais , Linhagem Celular , Sobrevivência Celular , Cobre , Óxidos N-Cíclicos/farmacologia , DNA/análise , DNA/efeitos dos fármacos , DNA/efeitos da radiação , Relação Dose-Resposta a Droga , Relação Dose-Resposta à Radiação , Eletroforese/métodos , Peróxido de Hidrogênio , Oxirredução , Fenantrolinas , Plasmídeos , Marcadores de Spin , Triacetonamina-N-Oxil/farmacologia
10.
Biochim Biophys Acta ; 1571(3): 211-8, 2002 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-12090935

RESUMO

Tachpyr (N,N'N"-tris(2-pyridylmethyl)-cis,cis-1,3,5-triaminocyclohexane), a novel metal chelator, was previously shown to deplete intracellular iron and exert a cytotoxic effect on cultured bladder cancer cells. Tachpyr binds Fe(II) and readily reduces Fe(III). The iron(II)-Tachpyr chelate undergoes intramolecular oxidative dehydrogenation resulting in mono- and diimino Fe(II) complexes. The present study investigates the redox-activity of the Tachpyr-iron complex to better define the mechanism of Tachpyr's cytotoxicity. Tachpyr's mechanism of cytotoxicity was studied using cell-free solutions, isolated DNA, and cultured mammalian cells by employing UV-VIS spectrophotometry, oximetry, spin-trapping technique, and electron paramagnetic resonance (EPR) spectrometry. The results show that: (1) Tachpyr by itself after 24 h of incubation had a cytotoxic effect on cultured cells; (2) fully oxidized Tachpyr had no cytotoxic effects on cultured cells even after 24 h of incubation; (3) Tachpyr protected isolated DNA against H(2)O(2)-induced damage, but not against HX/XO-induced damage; and (4) Tachpyr-Fe(II) chelate slows down but does not block oxidation of Fe(II), allows O*(-)(2)-induced or Tachpyr-induced reduction of Fe(III), and consequently promotes production of *OH through the Haber-Weiss reaction cycle. The results indicate that Tachpyr can protect cells against short-term, metal-mediated damage. However, upon prolonged incubation, Tachpyr exerts cytotoxic effects. Therefore, in addition to iron depletion, low-level oxidative stress, which in part occurs because of redox cycling of the coordinated iron ion, may contribute to the cytotoxic effects of Tachpyr.


Assuntos
Carcinoma/tratamento farmacológico , Quelantes/farmacologia , Cicloexilaminas/toxicidade , Neoplasias Pulmonares/tratamento farmacológico , Piridinas/toxicidade , Antineoplásicos/farmacologia , Carcinoma/patologia , Catalase/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Sistema Livre de Células , DNA/química , DNA/efeitos dos fármacos , DNA de Cadeia Simples/química , DNA de Cadeia Simples/efeitos dos fármacos , Relação Dose-Resposta a Droga , Humanos , Ferro/química , Neoplasias Pulmonares/patologia , Substâncias Macromoleculares , Oxirredução/efeitos dos fármacos , Oxigênio/química , Consumo de Oxigênio/efeitos dos fármacos , Plasmídeos/genética , Superóxido Dismutase/farmacologia , Células Tumorais Cultivadas , Ensaio Tumoral de Célula-Tronco
11.
Free Radic Biol Med ; 39(12): 1539-48, 2005 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-16298679

RESUMO

The mechanism(s) responsible for sudden cytolysis observed when cells are exposed to ultrasound could be mechanical and/or free radical in nature. Free radical reactions are initiated in the core and in the interfacial regions of collapsing acoustic cavitation bubbles. Because cyclic sugars are known to inhibit free radical chain reactions, we investigated the effects of n-alkyl-beta-d-glucopyranosides of varying hydrophobicity on ultrasound (1.057 MHz)-induced cytolysis of HL-60 cells in vitro. n-Alkyl glucopyranosides with hexyl- (5 mM), heptyl- (3 mM), or octyl- (2 mM) n-alkyl chains protected 100% of the cell population from ultrasound-induced cytolysis under a range of conditions that resulted in 35 to 100% cytolysis in the absence of glucopyranosides. The protected cell populations also possessed long-term reproductive viability. However, the hydrophilic methyl-beta-D-glucopyranoside could not protect cells, even up to a concentration of 30 mM. Furthermore, none of the glucopyranosides could prevent cytolysis of cells from a mechanically induced shear stress. Spin trapping and electron spin resonance experiments confirmed the presence of inertial cavitation in cell suspensions both in the presence and in the absence of the surfactants. It is concluded that surface-active glucopyranosides efficiently quench cytotoxic radicals and/or their precursors at the gas/solution interface of collapsing cavitation bubbles.


Assuntos
Citoproteção/efeitos dos fármacos , Glucosídeos/farmacologia , Ultrassom , Animais , Derivados de Benzeno/farmacologia , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Cricetinae , Radicais Livres/química , Radicais Livres/efeitos da radiação , Raios gama , Células HL-60 , Humanos , Técnicas In Vitro , Tamanho da Partícula
12.
Clin Cancer Res ; 21(12): 2792-801, 2015 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-25724523

RESUMO

PURPOSE: Radiation remains a mainstay for the treatment of nonmetastatic head and neck squamous cell carcinoma (HNSCC), a malignancy characterized by a high rate of PI3K/mTOR signaling axis activation. We investigated the ATP-competitive dual PI3K/mTOR inhibitor, PF-05212384, as a radiosensitizer in preclinical HNSCC models. EXPERIMENTAL DESIGN: Extent of radiation enhancement of two HNSCC cell lines (UMSCC1-wtP53 and UMSCC46-mtP53) and normal human fibroblast (1522) was assessed by in vitro clonogenic assay with appropriate target inhibition verified by immunoblotting. Radiation-induced DNA damage repair was evaluated by γH2AX Western blots with the mechanism of DNA double-strand break repair abrogation investigated by cell cycle analysis, immunoblotting, and RT-PCR. PF-05212384 efficacy in vivo was assessed by UMSCC1 xenograft tumor regrowth delay, xenograft lysate immunoblotting, and tissue section immunohistochemistry. RESULTS: PF-05212384 effectively inhibited PI3K and mTOR, resulting in significant radiosensitization of exponentially growing and plateau-phase cells with 24-hour treatment following irradiation, and variable radiation enhancement with 24-hour treatment before irradiation. Tumor cells radiosensitized to a greater extent than normal human fibroblasts. Postirradiation PF-05212384 treatment delays γH2AX foci resolution. PF-05212384 24-hour exposure resulted in an evident G1-S phase block in p53-competent cells. Fractionated radiation plus i.v. PF-05212384 synergistically delayed nude mice bearing UMSCC1 xenograft regrowth, with potential drug efficacy biomarkers identified, including pS6, pAkt, p4EBP1, and Ki67. CONCLUSIONS: Taken together, our results of significant radiosensitization both in vitro and in vivo validate the PI3K/mTOR axis as a radiation modification target and PF-05212384 as a potential clinical radiation modifier of nonmetastatic HNSCC.


Assuntos
Carcinoma de Células Escamosas/metabolismo , Neoplasias de Cabeça e Pescoço/metabolismo , Morfolinas/farmacocinética , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Radiossensibilizantes/farmacologia , Serina-Treonina Quinases TOR/metabolismo , Triazinas/farmacocinética , Animais , Carcinoma de Células Escamosas/patologia , Carcinoma de Células Escamosas/terapia , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Quebras de DNA de Cadeia Dupla/efeitos da radiação , Reparo do DNA/efeitos dos fármacos , Reparo do DNA/efeitos da radiação , Modelos Animais de Doenças , Relação Dose-Resposta à Radiação , Técnicas de Silenciamento de Genes , Neoplasias de Cabeça e Pescoço/patologia , Neoplasias de Cabeça e Pescoço/terapia , Humanos , Camundongos , Inibidores de Fosfoinositídeo-3 Quinase , Tolerância a Radiação/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Carcinoma de Células Escamosas de Cabeça e Pescoço , Serina-Treonina Quinases TOR/antagonistas & inibidores , Carga Tumoral/efeitos dos fármacos , Carga Tumoral/efeitos da radiação , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Cancer Res ; 75(14): 2788-99, 2015 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-25990221

RESUMO

Nitric oxide synthases (NOS) are important mediators of progrowth signaling in tumor cells, as they regulate angiogenesis, immune response, and immune-mediated wound healing. Ionizing radiation (IR) is also an immune modulator and inducer of wound response. We hypothesized that radiation therapeutic efficacy could be improved by targeting NOS following tumor irradiation. Herein, we show enhanced radiation-induced (10 Gy) tumor growth delay in a syngeneic model (C3H) but not immunosuppressed (Nu/Nu) squamous cell carcinoma tumor-bearing mice treated post-IR with the constitutive NOS inhibitor N(G)-nitro-l-arginine methyl ester (L-NAME). These results suggest a requirement of T cells for improved radiation tumor response. In support of this observation, tumor irradiation induced a rapid increase in the immunosuppressive Th2 cytokine IL10, which was abated by post-IR administration of L-NAME. In vivo suppression of IL10 using an antisense IL10 morpholino also extended the tumor growth delay induced by radiation in a manner similar to L-NAME. Further examination of this mechanism in cultured Jurkat T cells revealed L-NAME suppression of IR-induced IL10 expression, which reaccumulated in the presence of exogenous NO donor. In addition to L-NAME, the guanylyl cyclase inhibitors ODQ and thrombospondin-1 also abated IR-induced IL10 expression in Jurkat T cells and ANA-1 macrophages, which further suggests that the immunosuppressive effects involve eNOS. Moreover, cytotoxic Th1 cytokines, including IL2, IL12p40, and IFNγ, as well as activated CD8(+) T cells were elevated in tumors receiving post-IR L-NAME. Together, these results suggest that post-IR NOS inhibition improves radiation tumor response via Th1 immune polarization within the tumor microenvironment.


Assuntos
Quimiotaxia de Leucócito/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Ativação Linfocitária/efeitos dos fármacos , Neoplasias/patologia , Óxido Nítrico Sintase/antagonistas & inibidores , Tolerância a Radiação/efeitos dos fármacos , Radiossensibilizantes/farmacologia , Animais , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/efeitos da radiação , Inibidores Enzimáticos/uso terapêutico , Feminino , Humanos , Células Jurkat , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Nus , Neoplasias/tratamento farmacológico , Neoplasias/radioterapia
14.
Free Radic Biol Med ; 37(10): 1648-55, 2004 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-15477016

RESUMO

This study was designed to determine if radiation-mediated activation of the apoptotic pathways would be influenced by antioxidants and if a correlation would be found between radioprotection and changes in transduction pathways. Human lymphoblastoid TK6 cells, known to undergo apoptosis as a result of radiation, were irradiated (6 Gy) with and without antioxidants, and then whole-cell lysates were collected. Parallel studies were conducted to assess the survival (clonogenic assay) and apoptotic index. The impacts of two nitroxide antioxidants, tempol and CAT-1, differing in cell permeability, as well as the sulfhydryl antioxidant N-acetyl-L-cysteine (L-NAC), were estimated. Changes in apoptotic pathway proteins and p53 were assessed by Western blotting. Fraction of apoptotic cells was determined by flow cytometry. Tempol (10 mM), which readily enters cells, partially radioprotected TK6 cells against clonogenic killing, but had no effect on radiation-induced apoptotic parameters such as cleaved caspase 3 or cleaved PARP. Tempol alone did not induce cytotoxicity, yet did increase cleaved PARP levels. The radiation-induced increase in p53 protein was partly inhibited by tempol, but was unaffected by CAT-1 and L-NAC. Both CAT-1 (10 mM), which does not enter cells, and L-NAC (10 mM) had no radioprotective effect on cell survival. Although L-NAC did not protect against radiation-induced cytotoxicity, it completely inhibited radiation-induced increase in cleaved caspase 3 and cleaved PARP. Collectively, the results question the validity of using selected apoptosis pathway members as sole indicators of cytotoxicity.


Assuntos
Acetilcisteína/farmacologia , Antioxidantes/farmacologia , Apoptose/fisiologia , Sobrevivência Celular/fisiologia , Óxidos N-Cíclicos/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Caspase 3 , Caspases/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Colágeno Tipo XI/metabolismo , Humanos , Sistema de Sinalização das MAP Quinases/fisiologia , Fosforilação/efeitos dos fármacos , Fosforilação/efeitos da radiação , Radiação , Marcadores de Spin , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/metabolismo
15.
Cancer Res ; 74(23): 6771-83, 2014 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-25297630

RESUMO

Although significant advances in radiotherapy have increased its effectiveness in many cancer settings, general strategies to widen the therapeutic window between normal tissue toxicity and malignant tumor destruction would still offer great value. CD47 blockade has been found to confer radioprotection to normal tissues while enhancing tumor radiosensitivity. Here, we report that CD47 blockade directly enhances tumor immunosurveillance by CD8(+) T cells. Combining CD47 blockade with irradiation did not affect fibrosarcoma growth in T cell-deficient mice, whereas adoptive transfer of tumor-specific CD8(+) T cells restored combinatorial efficacy. Furthermore, ablation of CD8(+) T cells abolished radiotherapeutic response in immunocompetent syngeneic hosts. CD47 blockade in either target cells or effector cells was sufficient to enhance antigen-dependent CD8(+) CTL-mediated tumor cell killing in vitro. In CD47-deficient syngeneic hosts, engrafted B16 melanomas were 50% more sensitive to irradiation, establishing that CD47 expression in the microenvironment was sufficient to limit tumor radiosensitivity. Mechanistic investigations revealed increased tumor infiltration by cytotoxic CD8(+) T cells in a CD47-deficient microenvironment, with an associated increase in T cell-dependent intratumoral expression of granzyme B. Correspondingly, an inverse correlation between CD8(+) T-cell infiltration and CD47 expression was observed in human melanomas. Our findings establish that blocking CD47 in the context of radiotherapy enhances antitumor immunity by directly stimulating CD8(+) cytotoxic T cells, with the potential to increase curative responses.


Assuntos
Antígeno CD47/imunologia , Linfócitos T CD8-Positivos/imunologia , Tolerância a Radiação/imunologia , Linfócitos T Citotóxicos/imunologia , Microambiente Tumoral/imunologia , Transferência Adotiva/métodos , Animais , Linhagem Celular Tumoral , Citotoxicidade Imunológica , Fibrossarcoma/imunologia , Granzimas/imunologia , Humanos , Melanoma Experimental/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Radioterapia/métodos
16.
PLoS One ; 9(9): e107995, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25254649

RESUMO

BACKGROUND: TH-302 is a hypoxia-activated prodrug (HAP) of bromo isophosphoramide mustard that is selectively activated within hypoxic regions in solid tumors. Our recent study showed that intravenously administered bolus pyruvate can transiently induce hypoxia in tumors. We investigated the mechanism underlying the induction of transient hypoxia and the combination use of pyruvate to potentiate the anti-tumor effect of TH-302. METHODOLOGY/RESULTS: The hypoxia-dependent cytotoxicity of TH-302 was evaluated by a viability assay in murine SCCVII and human HT29 cells. Modulation in cellular oxygen consumption and in vivo tumor oxygenation by the pyruvate treatment was monitored by extracellular flux analysis and electron paramagnetic resonance (EPR) oxygen imaging, respectively. The enhancement of the anti-tumor effect of TH-302 by pyruvate treatment was evaluated by monitoring the growth suppression of the tumor xenografts inoculated subcutaneously in mice. TH-302 preferentially inhibited the growth of both SCCVII and HT29 cells under hypoxic conditions (0.1% O2), with minimal effect under aerobic conditions (21% O2). Basal oxygen consumption rates increased after the pyruvate treatment in SCCVII cells in a concentration-dependent manner, suggesting that pyruvate enhances the mitochondrial respiration to consume excess cellular oxygen. In vivo EPR oxygen imaging showed that the intravenous administration of pyruvate globally induced the transient hypoxia 30 min after the injection in SCCVII and HT29 tumors at the size of 500-1500 mm(3). Pretreatment of SCCVII tumor bearing mice with pyruvate 30 min prior to TH-302 administration, initiated with small tumors (∼ 550 mm(3)), significantly delayed tumor growth. CONCLUSIONS/SIGNIFICANCE: Our in vitro and in vivo studies showed that pyruvate induces transient hypoxia by enhancing mitochondrial oxygen consumption in tumor cells. TH-302 therapy can be potentiated by pyruvate pretreatment if started at the appropriate tumor size and oxygen concentration.


Assuntos
Antineoplásicos/farmacologia , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Nitroimidazóis/farmacologia , Consumo de Oxigênio/efeitos dos fármacos , Mostardas de Fosforamida/farmacologia , Pró-Fármacos/farmacologia , Ácido Pirúvico/farmacologia , Animais , Hipóxia Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sinergismo Farmacológico , Feminino , Humanos , Camundongos , Oxigênio/metabolismo , Fatores de Tempo , Ensaios Antitumorais Modelo de Xenoenxerto
17.
Autophagy ; 8(11): 1628-42, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22874555

RESUMO

Accidental or therapeutic exposure to ionizing radiation has severe physiological consequences and can result in cell death. We previously demonstrated that deficiency or blockade of the ubiquitously expressed receptor CD47 results in remarkable cell and tissue protection against ischemic and radiation stress. Antagonists of CD47 or its ligand THBS1/thrombospondin 1 enhance cell survival and preserve their proliferative capacity. However the signaling pathways that mediate this cell-autonomous radioprotection are unclear. We now report a marked increase in autophagy in irradiated T-cells and endothelial cells lacking CD47. Irradiated T cells lacking CD47 exhibit significant increases in formation of autophagosomes comprising double-membrane vesicles visualized by electron microscopy and numbers of MAP1LC3A/B(+) puncta. Moreover, we observed significant increases in BECN1, ATG5, ATG7 and a reduction in SQSTM1/p62 expression relative to irradiated wild-type T cells. We observed similar increases in autophagy gene expression in mice resulting from blockade of CD47 in combination with total body radiation. Pharmacological or siRNA-mediated inhibition of autophagy selectively sensitized CD47-deficient cells to radiation, indicating that enhanced autophagy is necessary for the prosurvival response to CD47 blockade. Moreover, re-expression of CD47 in CD47-deficient T cells sensitized these cells to death by ionizing radiation and reversed the increase in autophagic flux associated with survival. This study indicates that CD47 deficiency confers cell survival through the activation of autophagic flux and identifies CD47 blockade as a pharmacological route to modulate autophagy for protecting tissue from radiation injury.


Assuntos
Autofagia , Antígeno CD47/genética , Antígeno CD47/metabolismo , Especificidade de Órgãos/efeitos da radiação , Proteção Radiológica , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Apoptose/efeitos da radiação , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Autofagia/genética , Proteína 5 Relacionada à Autofagia , Proteína 7 Relacionada à Autofagia , Proteína Beclina-1 , Regulação da Expressão Gênica/efeitos da radiação , Inativação Gênica/efeitos da radiação , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Células Endoteliais da Veia Umbilical Humana/efeitos da radiação , Humanos , Células Jurkat , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Fagossomos/metabolismo , Fagossomos/ultraestrutura , Radiação Ionizante , Proteína Sequestossoma-1 , Enzimas Ativadoras de Ubiquitina/genética , Enzimas Ativadoras de Ubiquitina/metabolismo , Regulação para Cima/efeitos da radiação , Irradiação Corporal Total
18.
Oncotarget ; 3(11): 1472-82, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23178531

RESUMO

The tumor suppressor gene, Von Hippel-Lindau (VHL), is frequently mutated in the most common form of kidney cancer, clear cell renal cell carcinoma (CCRCC). In hypoxic conditions, or when there is a VHL mutation, the hypoxia inducible factors, HIF1α and HIF2α, are stabilized and transcribe a panel of genes associated with cancer such as vascular endothelial growth factor receptor (VEGFR), platelet derived growth factor (PDGF), and glucose transporter 1 (GLUT1). Recent studies in clear cell kidney cancer have suggested that HIF2α, but not HIF1α, is the critical oncoprotein in the VHL pathway. Therefore, targeting HIF2α could provide a potential therapeutic approach for patients with advanced CCRCC. Since iron regulatory protein 1 (IRP1) is known to inhibit the translation of HIF2α, we investigated whether Tempol, a stable nitroxide that activates IRP1 towards IRE-binding, might have a therapeutic effect on a panel of human CCRCC cells expressing both HIF1α and HIF2α. We first evaluated the protein expression of HIF1α and HIF2α in 15 different clear cell renal carcinoma cell lines established from patient tumors in our laboratory. Tempol decreased the expression of HIF2α, and its downstream targets in all the cell lines of the panel. This effect was attributed to a dramatic increase of IRE-binding activity of IRP1. Several cell lines were found to have an increased IRP1 basal activity at 20% O2 compared to 5% O2, which may lower HIF2α expression in some of the cell lines in a VHL-independent manner. Taken together our data identify Tempol as an agent with potential therapeutic activity targeting expression of HIF2α in VHL-deficient clear cell kidney cancer and illustrate the importance of studying biochemical processes at relevant physiological O2 levels.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Carcinoma de Células Renais/tratamento farmacológico , Carcinoma de Células Renais/genética , Óxidos N-Cíclicos/farmacologia , Neoplasias Renais/tratamento farmacológico , Neoplasias Renais/genética , Proteína Supressora de Tumor Von Hippel-Lindau/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/biossíntese , Carcinoma de Células Renais/metabolismo , Carcinoma de Células Renais/patologia , Processos de Crescimento Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/biossíntese , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Proteína 1 Reguladora do Ferro/metabolismo , Neoplasias Renais/metabolismo , Neoplasias Renais/patologia , Mutação , Biossíntese de Proteínas/efeitos dos fármacos , Marcadores de Spin , Proteína Supressora de Tumor Von Hippel-Lindau/genética , Doença de von Hippel-Lindau/genética , Doença de von Hippel-Lindau/metabolismo
19.
Front Oncol ; 1: 19, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22649756

RESUMO

PURPOSE: To observe the effect of guggulsterone (GS) on the radiation response in human cancer cell lines. MATERIALS AND METHODS: The radiation response of cancer cells treated with GS was observed by cell survival studies, cell growth assay, NF-κB activity assay, western blotting of some key growth promoting receptors, the DNA repair protein γH2AX, and flow cytometry for DNA analyses. RESULTS: GS inhibited radiation induced NF-κB activation and enhanced radiosensitivity in the pancreatic cell line, PC-Sw. It reduced both cell cycle movement and cell growth. GS reduced ERα protein in MCF7 cells and IGF1-Rß protein in colon cancer cells and pancreatic cancer cells and inhibited DNA double strand break (DSB) repair following radiation. CONCLUSION: GS induced radiation sensitization may be due to several different mechanisms including the inhibition of NF-κB activation and reductions in IGF1-Rß. In addition, GS induced γH2AX formation, primarily in the S-phase, indicates that DNA DSB's in the S-phase may be another reason for GS induced radiosensitivity. ERα down-regulation in response to GS suggests that it can be of potential use in the treatment of estrogen positive tumors that are resistant to tamoxifen.

20.
Cancer Biol Ther ; 12(10): 915-23, 2011 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-22024758

RESUMO

Excess reactive oxygen species (ROS) generated from ionizing radiation (IR) or endogenous sources like cellular respiration and inflammation produce cytotoxic effects that can lead to carcinogenesis. Resveratrol (RSV), a polyphenol with antioxidant and anticarcinogenic capabilities, has shown promise as a potential radiation modifier. The present study focuses on examining the effects of RSV or RSV metabolites as a radiation modifier in normal tissue. RSV or a RSV metabolite, piceatannol (PIC) did not protect human lung fibroblasts (1522) from the radiation-induced cell killing. Likewise, neither RSV nor PIC afforded protection against lethal total body IR in C3H mice. Additional research has shown protection in cells against hydrogen peroxide when treated with RSV. Therefore, clonogenic survival was measured in 1522 cells with RSV and RSV metabolites. Only the RSV derivative, piceatannol (PIC), showed protection against hydrogen peroxide mediated cytotoxicity; whereas, RSV enhanced hydrogen peroxide sensitivity at a 50 µM concentration; the remaining metabolites evaluated had little to no effect on survival. PIC also showed enhancement to peroxide exposure at a higher concentration (150 µM). A potential mechanism for RSV-induced sensitivity to peroxides could be its ability to block 1522 cells in the S-phase, which is most sensitive to hydrogen peroxide treatment. In addition, both RSV and PIC can be oxidized to phenoxyl radicals and quinones, which may exert cytotoxic effects. These cytotoxic effects were abolished when HBED, a metal chelator, was added. Taken together RSV and many of its metabolic derivatives are not effective as chemical radioprotectors and should not be considered for clinical use.


Assuntos
Antioxidantes/farmacologia , Protetores contra Radiação/farmacologia , Estilbenos/farmacologia , Animais , Antioxidantes/metabolismo , Ciclo Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Relação Dose-Resposta a Droga , Ácido Edético/análogos & derivados , Ácido Edético/farmacologia , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibroblastos/efeitos da radiação , Humanos , Peróxido de Hidrogênio/toxicidade , Camundongos , Camundongos Endogâmicos C3H , Estresse Oxidativo/efeitos dos fármacos , Fenóis/metabolismo , Quinonas/metabolismo , Protetores contra Radiação/metabolismo , Resveratrol , Estilbenos/metabolismo , Irradiação Corporal Total , terc-Butil Hidroperóxido/toxicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA