Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Publication year range
1.
Radiother Oncol ; 106(1): 138-46, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23321494

ABSTRACT

BACKGROUND AND PURPOSE: Despite appropriate radiotherapy, high-risk prostate cancer patients often experience local relapse and progression to metastatic disease. Radioresistance may be due to tumor-hypoxia but also due to the PTEN mutation/deletion present in 70% prostate cancers. We investigated whether the novel PI3K/mTOR inhibitor BEZ235 might sensitize prostate cancer cells to radiation and reduce hypoxia-induced radioresistance. MATERIALS AND METHODS: The potential radiosensitizing properties of BEZ235 were investigated in vitro and in vivo using two prostate cancer cell lines, PC3 (PTEN(-/-)) and DU145 (PTEN(+/+)), under normoxic (21% O(2)) and hypoxic (0.5% O(2)) conditions. RESULTS: BEZ235 rapidly inhibited PI3K and mTOR signaling in a dose dependent manner and limited tumor cell proliferation and clonogenic survival in both cell lines independently of PTEN status. In vivo, BEZ235 pretreatment enhanced the efficacy of radiation therapy on PC3 xenograft tumors in mice without inducing intestinal radiotoxicity. In culture, BEZ235 radiosensitized both cell lines in a comparable manner. Moreover, BEZ235 inhibited PI3K/mTOR activation and radiosensitized both cell lines under normoxia and hypoxia. BEZ235 radiosensitizing effects correlated with a decrease in γH2AX foci repair and increased G2/M cell cycle arrest. CONCLUSIONS: BEZ235 is a potent radiosensitizer of normoxic and hypoxic prostate cancer cells.


Subject(s)
Imidazoles/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Prostatic Neoplasms/radiotherapy , Quinolines/pharmacology , Radiation-Sensitizing Agents/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Cell Cycle Checkpoints , Cell Hypoxia , Cell Line, Tumor , DNA Breaks , Humans , Male , Mice , Prostatic Neoplasms/pathology
2.
PLoS One ; 8(12): e84076, 2013.
Article in English | MEDLINE | ID: mdl-24391887

ABSTRACT

Although endothelial cell apoptosis participates in the tumor shrinkage after single high-dose radiotherapy, little is known regarding the vascular response after conventionally fractionated radiation therapy. Therefore, we evaluated hypoxia, perfusion and vascular microenvironment changes in an orthotopic prostate cancer model of conventionally fractionated radiation therapy at clinically relevant doses (2 Gy fractions, 5 fractions/week). First, conventionally fractionated radiation therapy decreased tumor cell proliferation and increased cell death with kinetics comparable to human prostate cancer radiotherapy. Secondly, the injection of Hoechst 33342 or fluorescent-dextrans showed an increased tumor perfusion within 14 days in irradiated tumors, which was correlated with a clear reduction of hypoxia. Improved perfusion and decreased hypoxia were not explained by increased blood vessel density, size or network morphology. However, a tumor vascular maturation defined by perivascular desmin+/SMA+ cells coverage was clearly observed along with an increase in endothelial, zonula occludens (ZO)-1 positive, intercellular junctions. Our results show that, in addition to tumor cell killing, vascular maturation plays an uncovered role in tumor reoxygenation during fractionated radiation therapy.


Subject(s)
Cell Hypoxia/radiation effects , Dose Fractionation, Radiation , Neovascularization, Pathologic/radiotherapy , Prostatic Neoplasms/blood supply , Animals , Apoptosis/radiation effects , Biomarkers, Tumor/metabolism , Cell Proliferation/radiation effects , Humans , Image Processing, Computer-Assisted , Immunoenzyme Techniques , Male , Mice, Nude , Prostatic Neoplasms/pathology , Prostatic Neoplasms/radiotherapy
3.
PLoS One ; 7(5): e35672, 2012.
Article in English | MEDLINE | ID: mdl-22567107

ABSTRACT

Despite advances in radiation delivery protocols, exposure of normal tissues during the course of radiation therapy remains a limiting factor of cancer treatment. If the canonical TGF-ß/Smad pathway has been extensively studied and implicated in the development of radiation damage in various organs, the precise modalities of its activation following radiation exposure remain elusive. In the present study, we hypothesized that TGF-ß1 signaling and target genes expression may depend on radiation-induced modifications in Smad transcriptional co-repressors/inhibitors expressions (TGIF1, SnoN, Ski and Smad7). In endothelial cells (HUVECs) and in a model of experimental radiation enteropathy in mice, radiation exposure increases expression of TGF-ß/Smad pathway and of its target gene PAI-1, together with the overexpression of Smad co-repressor TGIF1. In mice, TGIF1 deficiency is not associated with changes in the expression of radiation-induced TGF-ß pathway-related transcripts following localized small intestinal irradiation. In HUVECs, TGIF1 overexpression or silencing has no influence either on the radiation-induced Smad activation or the Smad3-dependent PAI-1 overexpression. However, TGIF1 genetic deficiency sensitizes mice to radiation-induced intestinal damage after total body or localized small intestinal radiation exposure, demonstrating that TGIF1 plays a role in radiation-induced intestinal injury. In conclusion, the TGF-ß/Smad co-repressor TGIF1 plays a role in radiation-induced normal tissue damage by a Smad-independent mechanism.


Subject(s)
DNA-Binding Proteins/metabolism , Intestinal Mucosa/metabolism , Intestines/radiation effects , Proto-Oncogene Proteins/metabolism , Smad Proteins/metabolism , Smad7 Protein/metabolism , Animals , Blotting, Western , DNA-Binding Proteins/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mice , Mice, Inbred C57BL , Plasminogen Activator Inhibitor 1/genetics , Plasminogen Activator Inhibitor 1/metabolism , Proto-Oncogene Proteins/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism , Smad Proteins/genetics , Smad7 Protein/genetics , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism
4.
PLoS One ; 7(4): e35740, 2012.
Article in English | MEDLINE | ID: mdl-22563394

ABSTRACT

Normal tissue toxicity still remains a dose-limiting factor in clinical radiation therapy. Recently, plasminogen activator inhibitor type 1 (SERPINE1/PAI-1) was reported as an essential mediator of late radiation-induced intestinal injury. However, it is not clear whether PAI-1 plays a role in acute radiation-induced intestinal damage and we hypothesized that PAI-1 may play a role in the endothelium radiosensitivity. In vivo, in a model of radiation enteropathy in PAI-1 -/- mice, apoptosis of radiosensitive compartments, epithelial and microvascular endothelium was quantified. In vitro, the role of PAI-1 in the radiation-induced endothelial cells (ECs) death was investigated. The level of apoptotic ECs is lower in PAI-1 -/- compared with Wt mice after irradiation. This is associated with a conserved microvascular density and consequently with a better mucosal integrity in PAI-1 -/- mice. In vitro, irradiation rapidly stimulates PAI-1 expression in ECs and radiation sensitivity is increased in ECs that stably overexpress PAI-1, whereas PAI-1 knockdown increases EC survival after irradiation. Moreover, ECs prepared from PAI-1 -/- mice are more resistant to radiation-induced cell death than Wt ECs and this is associated with activation of the Akt pathway. This study demonstrates that PAI-1 plays a key role in radiation-induced EC death in the intestine and suggests that this contributes strongly to the progression of radiation-induced intestinal injury.


Subject(s)
Apoptosis/radiation effects , Endothelial Cells/metabolism , Intestines/injuries , Plasminogen Activator Inhibitor 1/metabolism , Animals , Cells, Cultured , Endothelial Cells/radiation effects , Human Umbilical Vein Endothelial Cells , Humans , Intestines/radiation effects , Mice , Mice, Inbred C57BL , Mice, Knockout , PTEN Phosphohydrolase/antagonists & inhibitors , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Plasminogen Activator Inhibitor 1/deficiency , Plasminogen Activator Inhibitor 1/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction/radiation effects
5.
Int J Radiat Oncol Biol Phys ; 74(3): 942-8, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19480973

ABSTRACT

PURPOSE: To investigate effects of plasminogen activator inhibitor 1 (PAI-1) genetic deficiency and pharmacological PAI-1 inhibition with PAI-039 in a mouse model of radiation-induced enteropathy. METHODS AND MATERIALS: Wild-type (Wt) and PAI-1(-/-) knockout mice received a single dose of 19 Gy to an exteriorized localized intestinal segment. Sham and irradiated Wt mice were treated orally with 1 mg/g of PAI-039. Histological modifications were quantified using a radiation injury score. Moreover, intestinal gene expression was monitored by real-time PCR. RESULTS: At 3 days after irradiation, PAI-039 abolished the radiation-induced increase in the plasma active form of PAI-1 and limited the radiation-induced gene expression of transforming growth factor beta1 (TGF-beta1), CTGF, PAI-1, and COL1A2. Moreover, PAI-039 conferred temporary protection against early lethality. PAI-039 treatment limited the radiation-induced increase of CTGF and PAI-1 at 2 weeks after irradiation but had no effect at 6 weeks. Radiation injuries were less severe in PAI-1(-/-) mice than in Wt mice, and despite the beneficial effect, 3 days after irradiation, PAI-039 had no effects on microscopic radiation injuries compared to untreated Wt mice. CONCLUSIONS: A genetic deficiency of PAI-1 is associated with amelioration of late radiation enteropathy. Pharmacological inhibition of PAI-1 by PAI-039 positively impacts the early, acute phase increase in plasma PAI-1 and the associated radiation-induced gene expression of inflammatory/extracellular matrix proteins. Since PAI-039 has been shown to inhibit the active form of PAI-1, as opposed to the complete loss of PAI-1 in the knockout animals, these data suggest that a PAI-1 inhibitor could be beneficial in treating radiation-induced tissue injury in acute settings where PAI-1 is elevated.


Subject(s)
Indoleacetic Acids/pharmacology , Intestines/radiation effects , Plasminogen Activator Inhibitor 1/deficiency , Radiation Injuries/prevention & control , Administration, Oral , Animals , Antigens, Differentiation/genetics , Antigens, Differentiation/metabolism , Collagen/genetics , Collagen/metabolism , Collagen Type I , Collagen Type III/genetics , Collagen Type III/metabolism , Connective Tissue Growth Factor/genetics , Connective Tissue Growth Factor/metabolism , Gene Expression/drug effects , Intestinal Mucosa/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Plasminogen Activator Inhibitor 1/blood , Plasminogen Activator Inhibitor 1/genetics , Radiation Injuries/metabolism , Radiation Injuries/mortality , Time Factors , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism
6.
Med Sci (Paris) ; 25(3): 267-72, 2009 Mar.
Article in French | MEDLINE | ID: mdl-19361390

ABSTRACT

Despite constant progress in radiotherapy techniques such as tumour imaging and cartography, techniques of radiation delivery or fractionation schedules, damage to normal gastro-intestinal tissues is inevitably associated with radiation therapy of pelvic tumours. Acute radiation enteritis concerns 80% of patients. It is related to stem cell loss, default in epithelial regenerating capacity and inflammation-induced mucosal dystrophy and ulceration. Chronic injury may develop in 5 to 10% of patients and is characterized by intestinal wall fibrosis resulting from an exaggerated scarring process, chronic inflammation and tissue necrosis. Research in mechanistic processes of normal tissue damage paved the way for new therapeutic approaches to emerge. These new targets include mucosal regeneration, reduction of vascular activation, inflammation and thrombosis, and fight against mesenchymal cells sustained activation. Effective strategies are multiple on preclinical models, but numerous efforts have to be made to achieve the complicated goal of protection of normal tissues from the side effects of radiation therapy.


Subject(s)
Gastrointestinal Diseases/etiology , Neoplasms/radiotherapy , Pelvic Neoplasms/radiotherapy , Radiation Injuries/epidemiology , Radiotherapy/adverse effects , Animals , Enteritis/epidemiology , Humans , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...