Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
Clin Nucl Med ; 46(9): 717-722, 2021 Sep 01.
Article in English | MEDLINE | ID: mdl-34034333

ABSTRACT

PURPOSE: The aim of the present study was to obtain information about distribution, radiation dosimetry, toxicity, and pharmacokinetics of O-[18F]fluoromethyl-d-tyrosine (d-18F-FMT), an amino acid PET tracer, in patients with brain tumors. PATIENTS AND METHODS: A total of 6 healthy controls (age = 19-25 years, 3 males and 3 females) with brain PET images and radiation dosimetry and 12 patients (median age = 60 years, 6 males and 6 females) with primary (n = 5) or metastatic brain tumor (n = 7) were enrolled. We acquired 60-minute dynamic brain PET images after injecting 370 MBq of d-18F-FMT. Time-activity curves of d-18F-FMT uptake in normal brain versus brain tumors and tumor-to-background ratio were analyzed for each PET data set. RESULTS: Normal cerebral uptake of d-18F-FMT decreased from 0 to 5 minutes after injection, but gradually increased from 10 to 60 minutes. Tumoral uptake of d-18F-FMT reached a peak before 30 minutes. Tumor-to-background ratio peaked at less than 15 minutes for 8 patients and more than 15 minutes for 4 patients. The mean effective dose was calculated to be 13.2 µSv/MBq. CONCLUSIONS: Using d-18F-FMT as a PET radiotracer is safe. It can distinguish brain tumor from surrounding normal brain tissues with a high contrast. Early-time PET images of brain tumors should be acquired because the tumor-to-background ratio tended to reach a peak within 15 minutes after injection.


Subject(s)
Brain Neoplasms , Positron Emission Tomography Computed Tomography , Adult , Brain/diagnostic imaging , Brain Neoplasms/diagnostic imaging , Female , Humans , Male , Middle Aged , Positron-Emission Tomography , Radiopharmaceuticals , Tyrosine , Young Adult
2.
J Neurosurg ; 121(6): 1483-91, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25303017

ABSTRACT

OBJECT: Local invasiveness of malignant glioma is a major reason for the failure of current treatments including surgery and radiation therapy. Tetraarsenic oxide (As4O6 [TAO]) is a trivalent arsenic compound that has potential anticancer and antiangiogenic effects in selected cancer cell lines at a lower concentration than arsenic trioxide (As2O3 [ATO]), which has been more widely tested in vitro and in vivo. The authors tried to determine the cytotoxic concentration of TAO in malignant glioma cell lines and whether TAO would show anti-invasive effects under conditions independent of cell death or apoptosis. METHODS: The human phosphatase and tensin homolog (PTEN)-deficient malignant glioma cell lines U87MG, U251MG, and U373MG together with PTEN-functional LN428 were cultured with a range of micromolar concentrations of TAO. The invasiveness of the glioma cell lines was analyzed. The effect of TAO on matrix metalloproteinase (MMP) secretion and membrane type 1 (MT1)-MMP expression was measured using gelatin zymography and Western blot, respectively. Akt, or protein kinase B, activity, which is a downstream effector of PTEN, was assessed with a kinase assay using glycogen synthesis kinase-3ß (GSK-3ß) as a substrate and Western blotting of phosphorylated Akt. RESULTS: Tetraarsenic oxide inhibited 50% of glioma cell proliferation at 6.3-12.2 µM. Subsequent experiments were performed under the same TAO concentrations and exposure times, avoiding the direct tumoricidal effect of TAO, which was confirmed with apoptosis markers. An invasion assay revealed a dose-dependent decrease in invasiveness under the influence of TAO. Both the gelatinolytic activity of MMP-2 and MT1-MMP expression decreased in a dose-dependent manner in all cell lines, which was in accordance with the invasion assay results. The TAO decreased kinase activity of Akt on GSK-3ß assay and inhibited Akt phosphorylation in a dose-dependent manner in all cell lines regardless of their PTEN status. CONCLUSIONS: These results showed that TAO effectively inhibits proliferation of glioblastoma cell lines and also exerts an anti-invasive effect via decreased MMP-2 secretion, decreased MT1-MMP expression, and the inhibition of Akt phosphorylation under conditions devoid of cytotoxicity. Further investigations using an in vivo model are needed to evaluate the potential role of TAO as an anti-invasive agent.


Subject(s)
Arsenicals/pharmacology , Brain Neoplasms/pathology , Glioblastoma/pathology , Matrix Metalloproteinase 14/metabolism , Matrix Metalloproteinase 2/metabolism , Oxides/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Arsenic Trioxide , Brain Neoplasms/enzymology , Cell Line, Tumor , Cell Proliferation/drug effects , Chromones/pharmacology , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Glioblastoma/enzymology , Humans , Morpholines/pharmacology , Neoplasm Invasiveness , Phosphorylation/drug effects
3.
J Korean Neurosurg Soc ; 54(3): 175-82, 2013 Sep.
Article in English | MEDLINE | ID: mdl-24278644

ABSTRACT

OBJECTIVE: Intracavitary injection of beta-emitting radiation source for control of cystic tumors has been tried with a benefit of localized internal radiation. The authors treated cystic brain tumor patients with Holmium-166-chitosan complex (Ho-166-chico), composed of a beta-emitting radionuclide Holmium-166 and biodegradable chit polymer, and evaluated the safety and effective measurement for response. METHODS: Twenty-two patients with recurrent cystic brain tumor and/or located in a deep or eloquent area were enrolled in this pilot study. The cyst volume and wall thickness were determined on CT or MRI to assess radiological response. The activity of Ho-166-chico injected via Ommaya reservoir was prescribed to be 10-25 Gy to the cyst wall in a depth of 4 mm. RESULTS: There was neither complications related to systemic absorption nor leakage of Ho-166-chico in all 22 patients. But, two cases of oculomotor paresis were observed in patients with recurrent craniopharyngioma. Radiological response was seen in 14 of 20 available follow-up images (70%). Seven patients of 'evident' radiological response experienced more than 25% decrease of both cyst volume and wall thickness. Another 7 patients with 'suggestive' response showed decrease of cyst volume without definitive change of the wall thickness or vice versa. All patients with benign tumors or low grade gliomas experienced symptomatic improvement. CONCLUSION: Ho-166-chico intracavitary radiation therapy for cystic tumor is a safe method of palliation without serious complications. The determination of both minimal effective dosage and time interval of repeated injection through phase 1 trial could improve the results in the future.

4.
Acta Oncol ; 51(5): 589-95, 2012 May.
Article in English | MEDLINE | ID: mdl-22414095

ABSTRACT

BACKGROUND: The aim of this study is to evaluate local control rates after stereotactic body radiotherapy (SBRT) in recurrent spinal metastasis after external beam radiotherapy (EBRT) and new spinal metastatic lesions. MATERIAL AND METHODS: Retrospective review of medical records and radiological data was performed on 54 retreatment and 131 initial SBRT patients. To compare various fractionation schedules, the biologically effective dose (BED) was applied. SBRT dose was calculated with linear-quadratic model and normalized to a 2-Gy equivalent dose (nBED, α/ß =2 Gy for spinal cord, α/ß =10 Gy for tumor). Doses to a point within the spinal cord that received the maximum dose (Pmax) were checked. Local control failure was defined as progression by imaging study. Overall survival, progression free survival, delivered radiation dose to tumor and spinal cord, and spinal cord Pmax nBED were compared in two groups. RESULTS: The mean delivered radiation doses to tumor margin during SBRT were 51.1 Gy2/10 (retreatment) and 50.7 Gy2/10 (initial treatment). Mean survival was 29.6 months (overall)/20.7 months (retreatment)/ 32.4 months (initial treatment). Mean progression free period was 23.9 months (overall)/18.0 months (retreatment)/ 26.0 months (initial treatment). Radiological control rates of retreatment and initial treatment group were 96%/95% at six months, 81%/89% at 12 months and 79%/90% at 24 months. Among 54 retreatment lesions, 13 lesions showed local control failure during follow-up. With regard to spinal cord radiation dose during SBRT, Spinal cord Pmax nBED was 46.2 Gy2/2 (retreatment) and 48.7 Gy2/2 (initial treatment). In retreatment group, total nBED to spinal cord was a mean of 83.4 Gy2/2. There was no case of radiation myelopathy detected. CONCLUSIONS: Retreatment of spinal metastases using SBRT provided effective local control without neurological complications.


Subject(s)
Breast Neoplasms/surgery , Prostatic Neoplasms/surgery , Radiosurgery , Spinal Neoplasms/secondary , Spinal Neoplasms/surgery , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Female , Follow-Up Studies , Humans , Male , Medical Records , Middle Aged , Prognosis , Prostatic Neoplasms/mortality , Prostatic Neoplasms/pathology , Radiation Injuries , Radiation Tolerance , Relative Biological Effectiveness , Retreatment , Retrospective Studies , Spinal Neoplasms/mortality , Survival Rate
5.
J Neurooncol ; 101(1): 91-9, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20508971

ABSTRACT

This study evaluated clinical outcome and safety of radiosurgery using the Cyberknife for treatment of benign spinal tumors. The authors treated 30 benign spinal tumors in 20 patients with the Cyberknife (Accuray, Inc., Sunnyvale, CA, USA) from 2002 to 2008. Among these there were 20 neurogenic tumors, eight hemangioblastomas, and two meningiomas. Four patients with neurofibromatosis (NF) type 2 and four patients with Von Hippel Lindau disease were also included. Radiosurgery was done as primary treatment for 22 lesions, for postoperative residual tumor control for four lesions, and for the remaining four lesions with image-based progression after initial subtotal resection. The distribution of lesions was cervical (18 tumors), thoracic (six), and cauda equina level (six). Follow-up data included imaging studies, clinical findings, and radiotherapy data. Tumor volume ranged from 0.04 to 33.65 cm³ (mean, 4.52 cm³). A 14-33 Gy marginal dose was delivered in 1-5 fractions. The mean follow-up period was 35.6 months (range, 12-84 months). On follow-up, most lesions decreased in size (57%) or remained unchanged (33%). Two lesions initially decreased, then increased later. One lesion increased without response. With regard to clinical aspects, radicular pain and myelopathic pain improved after radiosurgery in most cases (94%). Motor weakness recovered in two out of five patients and recovery of sensory change occurred in four out of ten patients. In two patients, symptoms were aggravated by tumor enlargement and the occurrence of new lesion. Mean spinal cord volumes receiving more than 10 and 8 Gy were 0.40 ± 0.4 and 0.81 ± 0.7 cm³, respectively. Stereotactic radiosurgery (SRS) using the Cyberknife showed the ability to control benign spinal tumors without complication in most cases.


Subject(s)
Radiosurgery , Spinal Cord Neoplasms/surgery , Adolescent , Adult , Aged , Female , Hemangioblastoma/pathology , Hemangioblastoma/surgery , Humans , Korea , Male , Middle Aged , Neurilemmoma/pathology , Neurilemmoma/surgery , Neurofibroma/pathology , Neurofibroma/surgery , Recovery of Function , Spinal Cord Neoplasms/pathology , Treatment Outcome , Young Adult
6.
J Neurooncol ; 103(3): 595-602, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21052775

ABSTRACT

A prospective randomized controlled multicenter phase III trial was conducted to evaluate the effects of neoadjuvant chemotherapy with nimustine (ACNU)-cisplatin (CDDP) when used in conjunction with radiotherapy plus adjuvant temozolomide in patients with newly diagnosed glioblastoma. The study population was randomly assigned into one treatment and one control group. Both groups received radiotherapy followed by six cycles of adjuvant oral temozolomide (150-200 mg/m(2)) for 5 days every 28 days after surgery. Prior to radiotherapy, the treatment group also received two cycles, 6 weeks apart, of neoadjuvant chemotherapy with ACNU (40 mg/m(2)/day) and CDDP (40 mg/m(2)/day) infused continuously for 72 h. The primary end-point was median survival time. The study has closed after interim analysis with a total of 82 patients (48.8% of target number) due to unacceptable high frequency of toxicity profiles in spite of the promising actuarial survival outcome. Median survival time was 28.4 months [90% confidence interval (CI), 21.1 months to not available] in the treatment group and 18.9 months (90% CI, 17.1-27.4 months) in the control group (P = 0.2). The 2-year survival rate and progression-free survival time were 50.9% and 6.6 months (90% CI, 3.5-9.5 months) in the treatment group and 27.8% and 5.1 months (90% CI, 3.8-8.8 months) in the control group. Grade 3 or 4 toxicity was documented in 26 (68.4%) patients in the treatment group, including three neutropenic fever and one death from sepsis, while grade 3 or 4 toxicity occurred in 6 patients (15.8%) in the control group. The high frequency of serious hematological toxicity with ACNU-CDDP neoadjuvant chemotherapy followed by radiotherapy and adjuvant temozolomide limits its usage as primary treatment for glioblastoma. Future studies should aim to identify a subpopulation at reduced risk for ACNU-CDDP toxicity so that the potential of this protocol can be realized.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Combined Modality Therapy , Dacarbazine/analogs & derivatives , Glioblastoma/drug therapy , Glioblastoma/radiotherapy , Adolescent , Adult , Aged , Chemotherapy, Adjuvant/methods , Dacarbazine/therapeutic use , Disease Progression , Female , Follow-Up Studies , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Prospective Studies , Temozolomide , Treatment Outcome , Young Adult
7.
J Korean Neurosurg Soc ; 50(5): 426-33, 2011 Nov.
Article in English | MEDLINE | ID: mdl-22259689

ABSTRACT

OBJECTIVE: The Histoculture Drug Response Assay (HDRA), which measures chemosensitivity using minced tumor tissue on drug-soaked gelfoam, has been expected to overcome the limitations of in vitro chemosensitivity test in part. We analyzed interim results of HDRA in malignant gliomas to see if the test can deserve further clinical trials. METHODS: Thirty-three patients with malignant gliomas were operated and their tumor samples were examined for the chemosensitivity to 10 chosen drugs by HDRA. The most sensitive chemotherapy regimen among those pre-established was chosen based on the number of sensitive drugs or total inhibition rate (IR) of the regimen. The response was evaluated by 3 month magnetic resonance image. RESULTS: Among 13 patients who underwent total resection of the tumor, 12 showed no evidence of disease and one patient revealed progression. The response rate in 20 patients with residual tumors was 55% (3 complete and 8 partial responses). HDRA sensitivity at the cut-off value of more than one sensitive drug in the applied regimen showed a sensitivity of 100%, specificity of 60% and predictability of 70%. Another cut-off value of >80% of total IR revealed a sensitivity of 100%, specificity of 69%, and predictability of 80%. For 12 newly diagnosed glioblastoma patients, median progression-free survival of the HDRA sensitive group was 21 months, while that of the non-sensitive group was 6 months (p=0.07). CONCLUSION: HDRA for malignant glioma was inferred as a feasible method to predict the chemotherapy response. We are encouraged to launch phase 2 clinical trial with chemosensitivity on HDRA.

8.
Nucl Med Mol Imaging ; 44(1): 45-54, 2010 Apr.
Article in English | MEDLINE | ID: mdl-24899937

ABSTRACT

PURPOSE: The purpose of this study was to compare the uptakes and diagnostic accuracies between 3'-deoxy-3'-[(18)F]fluorothymidine (FLT) and O-(2-[(18)F]fluoroethyl)-L-tyrosine (FET) PET in patients with a clinical suspicion of having a recurrence of glioma after multimodality treatment. METHODS: Thirty-two patients who underwent FLT and FET PET due to abnormal enhancement on magnetic resonance (MR) images were included. According to surgical confirmation or follow-up results, patients were divided into those with therapy-related benign changes (TRBCs) and those with recurrence. Recurrences were divided again into initial low-grade glioma (LGG) and high-grade glioma (HGG). The uptakes of FLT and FET were compared with the maximum standardized uptake value (SUVmax) and lesion-to-normal ratio (LNR). The diagnostic accuracies were compared via a receiver-operating-characteristic (ROC) curve analysis. RESULTS: The LNRs of FLT in recurrences with initial HGG (8.26 ± 5.02) were significantly higher than those in recurrences with initial LGG (3.43 ± 2.14) and TRBC (1.81 ± 0.60). The LNRs of FET in recurrence with initial HGG (2.70 ± 0.48) and LGG (3.03 ± 1.32) were significantly higher than those in the TRBC (1.60 ± 0.47). The areas under the ROC curve (AUCs) of FLT and FET for initial LGG were 0.768 and 0.893, respectively. The AUCs of FLT and FET for initial HGG were 1.000 and 0.964. However, there were no statistical significances. The results for comparing with SUVmax were the same as those with LNR. CONCLUSIONS: Uptakes of FLT were different according to initial grade in patients with recurrent glioma, but those of FET were not. However, there were no statistical significances in the diagnostic accuracies according to initial grade between the two tracers in this study.

9.
J Biol Chem ; 284(45): 30871-80, 2009 Nov 06.
Article in English | MEDLINE | ID: mdl-19740745

ABSTRACT

The Hsp90-associated protein p23 modulates Hsp90 activity during the final stages of the chaperone pathway to facilitate maturation of client proteins. Previous reports indicate that p23 cleavage induced by caspases during cell death triggers destabilization of client proteins. However, the specific role of truncated p23 (Delta p23) in this process and the underlying mechanisms remain to be determined. One such client protein, hTERT, is a telomerase catalytic subunit regulated by several chaperone proteins, including Hsp90 and p23. In the present study, we examined the effects of p23 cleavage on hTERT stability and telomerase activity. Our data showed that overexpression of Delta p23 resulted in a decrease in hTERT levels, and a down-regulation in telomerase activity. Serine phosphorylation of Hsp90 was significantly reduced in cells expressing high levels of Delta p23 compared with those expressing full-length p23. Mutation analyses revealed that two serine residues (Ser-231 and Ser-263) in Hsp90 are important for activation of telomerase, and down-regulation of telomerase activity by Delta p23 was associated with inhibition of cell growth and sensitization of cells to cisplatin. Our data aid in determining the mechanism underlying the regulation of telomerase activity by the chaperone complex during caspase-dependent cell death.


Subject(s)
DNA-Binding Proteins/metabolism , Down-Regulation , HSP90 Heat-Shock Proteins/metabolism , Telomerase/metabolism , Cell Line , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , HSP90 Heat-Shock Proteins/genetics , Humans , Phosphorylation , Protein Stability , Telomerase/genetics
10.
J Korean Neurosurg Soc ; 45(5): 275-83, 2009 May.
Article in English | MEDLINE | ID: mdl-19516944

ABSTRACT

OBJECTIVE: We retrospectively analyzed survival, local control rate, and incidence of radiation toxicities after radiosurgery for recurrent metastatic brain lesions whose initial metastases were treated with whole-brain radiotherapy. Various radiotherapeutical indices were examined to suggest predictors of radiation-related neurological dysfunction. METHODS: In 46 patients, total 100 of recurrent metastases (mean 2.2, ranged 1-10) were treated by CyberKnife radiosurgery at average dose of 23.1 Gy in 1 to 3 fractions. The median prior radiation dose was 32.7 Gy, the median time since radiation was 5.0 months, and the mean tumor volume was 12.4 cm(3). Side effects were expressed in terms of radiation therapy oncology group (RTOG) neurotoxicity criteria. RESULTS: Mass reduction was observed in 30 patients (65%) on MRI. After the salvage treatment, one-year progression-free survival rate was 57% and median survival was 10 months. Age (<60 years) and tumor volume affected survival rate (p=0.03, each). Acute (6 months) toxicity occurred in 21%, respectively. Less acute toxicity was observed with small tumors (<10 cm(3), p=0.03), and less chronic toxicity occurred at lower cumulative doses (<100 Gy, p=0.004). "Radiation toxicity factor" (cumulative dose times tumor volume of <1,000 Gyxcm(3)) was a significant predictor of both acute and chronic CNS toxicities. CONCLUSION: Salvage CyberKnife radiosurgery is effective for recurrent brain metastases in previously irradiated patients, but careful evaluation is advised in patients with large tumors and high cumulative radiation doses to avoid toxicity.

11.
Free Radic Biol Med ; 46(8): 1158-67, 2009 Apr 15.
Article in English | MEDLINE | ID: mdl-19439225

ABSTRACT

Regulation of mRNA translation in mammalian cells involves the coordinated control of mammalian target of rapamycin (mTOR) signaling. At present, limited information is available on the potential relevance of mTOR regulation, although translation inhibition during oxidative and endoplasmic reticulum (ER) stress is clearly important. In this study, we show that activating transcription factor 4 (ATF4) and CCAAT/enhancer-binding protein-beta (C/EBP-beta) negatively regulate mTOR via Redd1 expression in response to oxidative and ER stress. Oxidative and ER stress conditions induce rapid and significant activation of ATF4 downstream of eIF2alpha phosphorylation, which is responsible for Redd1 expression. In our experiment, overexpression of ATF4 was associated with reduced mTOR activity via Redd1 expression, whereas suppression of ATF4 levels with small interfering RNA led to the recovery of decreased mTOR activity mediated by downregulation of Redd1 during oxidative and ER stress. We additionally identified Redd1 as a downstream effector of C/EBP-beta stimulated by ATF4 activated under the stress conditions examined. RNA interference studies provided further evidence of the requirement of C/EBP-beta for Redd1 expression. We conclude that the Redd1 gene is transactivated by the ATF4 and C/EBP family of transcription factors, leading to mTOR inhibition in response to oxidative and ER stress.


Subject(s)
Activating Transcription Factor 4/physiology , CCAAT-Enhancer-Binding Protein-beta/physiology , Transcription Factors/metabolism , Transcriptional Activation , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Endoplasmic Reticulum , Feedback, Physiological , HeLa Cells , Humans , Hydrogen Peroxide/metabolism , Oxidative Stress , Protein Kinases/genetics , Protein Kinases/metabolism , RNA, Small Interfering/genetics , Signal Transduction , Sirolimus/metabolism , TOR Serine-Threonine Kinases , Transcription Factors/genetics
12.
J Korean Neurosurg Soc ; 46(6): 538-44, 2009 Dec.
Article in English | MEDLINE | ID: mdl-20062569

ABSTRACT

OBJECTIVE: Primary treatment of spinal metastasis has been external beam radiotherapy. Recent advance of technology enables radiosurgery to be extended to extracranial lesions. The purpose of this study was to determine the clinical effectiveness and safety of stereotactic radiosurgery using Cyberknife in spinal metastasis. METHODS: From June, 2002 to December, 2007, 129 patients with 167 spinal metastases were treated with Cyberknife. Most of the patients (94%) presented with pain and nine patients suffered from motor deficits. Twelve patients were asymptomatic. Fifty-three patients (32%) had previous radiation therapy. Using Cyberknife, 16-39 Gy in 1-5 fractions were delivered to spinal metastatic lesions. Radiation dose was not different regarding the tumor pathology or tumor volume. RESULTS: After six months follow-up, patient evaluation was possible in 108 lesions. Among them, significant pain relief was seen in 98 lesions (91%). Radiological data were obtained in 83 lesions. The mass size was decreased or stable in 75 lesions and increased in eight lesions. Radiological control failure cases were hepatocellular carcinoma (5 cases), lung cancer (1 case), breast cancer (1 case) and renal cell carcinoma (1 case). Treatment-related radiation injury was not detected. CONCLUSION: Cyberknife radiosurgery is clinically effective and safe for spinal metastases. It is true even in previously irradiated patients. Compared to conventional radiation therapy, Cyberknife shows higher pain control rate and its treatment process is more convenient for patients. Thus, it can be regarded as a primary treatment modality for spinal metastases.

13.
Mol Cancer Res ; 6(6): 996-1002, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18567803

ABSTRACT

Epidermal growth factor receptor (EGFR) is activated by ionizing radiation (IR), but the molecular mechanism for this effect is unknown. We have found that intracellular generation of nitric oxide (NO) by NO synthase (NOS) is required for the rapid activation of EGFR phosphorylation by IR. Treatment of A549 lung cancer cells with IR increased NOS activity within minutes, accompanied by an increase of NO. 2-Phenyl-4,4,5,5,-tetramethylimidazolline-1-oxyl-3-oxide, an NO scavenger, and NG-monomethyl-l-arginine, an NOS inhibitor, abolished the increase in intracellular NO and activation of EGFR by IR. In addition, an NO donor alone induced EGFR phosphorylation. Transient transfection with small interfering RNA for endothelial NOS reduced IR-induced NO production and suppressed IR-induced EGFR activation. Overexpression of endothelial NOS increased IR-induced NO generation and EGFR activation. These results indicate a novel molecular mechanism for EGFR activation by IR-induced NO production via NOS.


Subject(s)
ErbB Receptors/metabolism , Nitric Oxide/biosynthesis , Radiation, Ionizing , Cell Line, Tumor , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/metabolism , Nitric Oxide Synthase Type III/metabolism , Signal Transduction
14.
Lung Cancer ; 61(3): 317-27, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18281123

ABSTRACT

SUMMARY: In the present study, we show that a combination of sulindac and arsenic trioxide (ATO) induces more extensive apoptosis than either drug alone in H1299 human non-small cell lung carcinoma (NSCLC) cells. Treatment with sulindac/ATO triggered three major apoptotic signaling events, namely, collapse of the mitochondrial membrane potential, release of cytochrome c, and activation of caspases. Furthermore, the sulindac/ATO combination induced reactive oxygen species (ROS) generation, and the antioxidant, N-acetyl-L-cysteine, blocked this apoptotic signaling. The c-Jun NH(2)-terminal kinase (JNK) was activated downstream of ROS production in H1299 cells. Blockage of JNK by pretreatment with SP600125, a pharmacological inhibitor, or transfection with dominant-negative (DN) JNK1 vectors abrogated sulindac/ATO-induced apoptosis, as evident from the disruption of caspase activation. Interestingly, a slower migrating Bcl-xL band was observed on immunoblots after treatment of cells with sulindac/ATO. The band was absent upon the treatment of cell lysates with lambda protein phosphatase. Moreover, confocal microscopy findings disclose that active JNK translocates to mitochondria. Treatment with SP600125 and transfection with DN-JNK blocked Bcl-xL phosphorylation, suggesting that JNK plays an important role in sulindac/ATO-induced Bcl-xL phosphorylation. In conclusion, in H1299 human NSCLC cells, sulindac and ATO synergistically induce a high degree of apoptosis, which is mediated by the ROS-dependent JNK activation pathway via Bcl-xL phosphorylation.


Subject(s)
Apoptosis/drug effects , Arsenicals/pharmacology , Carcinoma, Non-Small-Cell Lung/pathology , JNK Mitogen-Activated Protein Kinases/metabolism , Lung Neoplasms/pathology , Oxides/pharmacology , Sulindac/pharmacology , bcl-X Protein/metabolism , Acetylcysteine/pharmacology , Arsenic Trioxide , Arsenicals/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Caspases/metabolism , Cell Line, Tumor/metabolism , Drug Synergism , Humans , JNK Mitogen-Activated Protein Kinases/physiology , Lung Neoplasms/metabolism , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Oxides/metabolism , Phosphorylation , Reactive Oxygen Species/metabolism , Signal Transduction , Sulindac/metabolism , bcl-X Protein/physiology
15.
Mol Pharmacol ; 73(3): 1005-12, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18156316

ABSTRACT

Histone deacetylase (HDAC) inhibitors represent a promising group of anticancer agents. Treatment of cancer cells with HDAC blockers, such as suberoylanilide hydroxamic acid (SAHA), leads to the activation of apoptosis-promoting genes. To enhance proapoptotic efficiency, SAHA has been used in conjunction with radiation, kinase inhibitors, and cytotoxic drugs. In the present study, we show that at the suboptimal dose of 250 muM, sulindac [2-[6-fluoro-2-methyl-3-[(4-methylsulfinylphenyl)methylidene]inden-1-yl]-acetic acid] significantly enhances SAHA-induced growth suppression and apoptosis of A549 human non-small cell lung cancer cells, primarily via enhanced collapse of the mitochondrial membrane potential, release of cytochrome c, and caspase activation. Furthermore, sulindac/SAHA cotreatment induced marked down-regulation of survivin at both the mRNA and protein levels and stimulated the production of reactive oxygen species (ROS), which were blocked by the antioxidant N-acetyl-l-cysteine. Overexpression of survivin was associated with reduced sulindac/SAHA-induced apoptosis of A549 cells, whereas suppression of survivin levels with antisense oligonucleotides or small interfering RNA further sensitized cells to sulindac/SAHA-induced cell death. Our results collectively demonstrate that sulindac/SAHA-induced apoptosis is mediated by ROS-dependent down-regulation of survivin in lung cancer cells.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Apoptosis/drug effects , Hydroxamic Acids/pharmacology , Lung Neoplasms/drug therapy , Sulindac/pharmacology , Annexin A5/metabolism , Carcinoma, Non-Small-Cell Lung/drug therapy , Caspases/analysis , Cell Survival/drug effects , Drug Synergism , Enzyme Inhibitors/pharmacology , Fluorescein-5-isothiocyanate/metabolism , Fluorescent Dyes/metabolism , Histone Deacetylase Inhibitors , Humans , Indicators and Reagents/metabolism , Membrane Potential, Mitochondrial/drug effects , Membrane Potential, Mitochondrial/physiology , Propidium/metabolism , Tumor Cells, Cultured/drug effects , Vorinostat
16.
J Biol Chem ; 282(42): 30485-96, 2007 Oct 19.
Article in English | MEDLINE | ID: mdl-17666398

ABSTRACT

Nerve growth factor (NGF) is a well characterized neurotrophic agonist in the nervous system that triggers angiogenesis. In this study, we investigated the signaling mechanisms involved in NGF-induced angiogenesis. NGF stimulated endothelial cell invasion and cord formation on Matrigel in vitro but had marginal effect on proliferation and migration of these cells. NGF stimulated matrix metalloproteinase (MMP)-2 mRNA expression and protein secretion in human umbilical vein endothelial cells. Using synthetic and endogenous inhibitors of MMP-2 and MMP-2 small interfering RNA suppressed NGF-induced invasion and cord formation. We demonstrated that NGF-induced MMP-2 secretion, invasion, and cord formation are regulated via activation of the NGF receptor, TrkA, phosphatidylinositol 3-kinase (PI3K), and Akt using various pharmacological inhibitors. Specifically, NGF enhanced TrkA phosphorylation, PI3K activity, and Akt phosphorylation. Introduction of NGF-neutralizing antibodies, dominant-negative Akt, or wild-type PTEN effectively inhibited NGF-induced MMP-2 secretion and cord formation. Deletion and site-directed mutagenesis analysis of the MMP-2 promoter demonstrated that the AP-2-binding site is critical for NGF-induced MMP-2 promoter activity. NGF increased the DNA binding activity of AP-2, which was suppressed by inhibitors of TrkA and PI3K. Furthermore, transfection of AP-2 small interfering RNA effectively blocked NGF-induced MMP-2 secretion and cord formation. Finally, NGF promoted neovessel formation in Matrigel plugs in vivo, which was significantly inhibited by K252a and LY294002, but it failed to promote angiogenesis using MMP-2 knock-out mice. Our data collectively suggest that NGF stimulates endothelial cell invasion and cord formation by augmenting MMP-2 via the PI3K/Akt signaling pathway and AP-2 transcription factor, which may be responsible for triggering angiogenesis.


Subject(s)
Cell Movement/drug effects , Endothelial Cells/enzymology , Matrix Metalloproteinase 2/metabolism , Nerve Growth Factor/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Transcription Factor AP-2/metabolism , Umbilical Veins/enzymology , Animals , Cell Movement/physiology , Cells, Cultured , Chromones/pharmacology , Enzyme Activation/drug effects , Enzyme Activation/physiology , Enzyme Inhibitors/pharmacology , Gene Deletion , Humans , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase Inhibitors , Mice , Mice, Knockout , Morpholines/pharmacology , Mutagenesis, Site-Directed , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/physiology , Nitriles/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Protein Binding/drug effects , Protein Binding/physiology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Pyrrolidines/pharmacology , RNA, Small Interfering/pharmacology , Receptor, trkA/antagonists & inhibitors , Receptor, trkA/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Umbilical Veins/cytology
17.
Cell Signal ; 19(7): 1393-403, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17307335

ABSTRACT

Redd1, a recently discovered stress-response gene, is regulated by hypoxia via hypoxia-inducible factor 1 (HIF-1) and by DNA damage via p53/p63; however, the signaling pathway by which its expression is induced by hypoxia has not been elucidated. In the present study, we demonstrated that the expression of Redd1 in response to hypoxia (1% O(2)), hypoxia-mimetic agent, cobalt chloride (CoCl(2)) and high cell density (HCD) requires coactivation of HIF-1alpha and Sp1. CoCl(2) and HCD induced the activation of HIF-1alpha and Sp1 in HeLa cells, and siRNAs targeting HIF-1alpha and Sp1 abrogated Redd1 expression. Inhibition of phosphatidylinositol 3-kinase (PI3K) by LY294002 and by a dominant-negative PI3K mutant reduced the expression of Redd1 and activation of HIF-1alpha and Sp1 by CoCl(2) and HCD. Also, suppression of Akt activation blocked the expression of Redd1 and the activation of HIF-1alpha and Sp1 by CoCl(2) and HCD. Furthermore, we found that the induction of Redd1 expression by CoCl(2) can be mediated by activation of Sp1 in HIF-1alpha-deficient cells but that a higher level of Redd1 expression is achieved when these cells are transfected with HIF-1alpha. These results demonstrate that hypoxic condition-and HCD-induced expression of Redd1 is mediated by coactivation of Sp1 and HIF-1alpha downstream of the PI3K/Akt signaling pathway.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Sp1 Transcription Factor/metabolism , Transcription Factors/metabolism , Binding Sites , Cell Count , Cell Hypoxia , Consensus Sequence , Enzyme Activation , Gene Expression Regulation , HeLa Cells , Humans , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription Factors/genetics
18.
Apoptosis ; 12(1): 195-209, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17136320

ABSTRACT

Non-steroidal anti-inflammatory drugs are well known to induce apoptosis of cancer cells independent of their ability to inhibit cyclooxygenase-2, but the molecular mechanism for this effect has not yet been fully elucidated. The purpose of this study was to elucidate the potential signaling components underlying sulindac-induced apoptosis in human multiple myeloma (MM) cells. We found that sulindac induces apoptosis by promoting ROS generation, accompanied by opening of mitochondrial permeability transition pores, release of cytochrome c and apoptosis inducing factor from mitochondria, followed by caspase activation. Bcl-2 cleavage and down-regulation of the inhibitor of apoptosis proteins (IAPs) family including cIAP-1/2, XIAP, and survivin, occurred downstream of ROS production during sulindac-induced apoptosis. Forced expression of survivin and Bcl-2 blocked sulindac-induced apoptosis. Most importantly, sulindac-derived ROS activated p38 mitogen-activated protein kinase and p53. SB203580, a p38 mitogen-activated protein kinase inhibitor, and RNA inhibition of p53 inhibited the sulindac-induced apoptosis. Furthermore, p53, Bax, and Bak accumulated in mitochondria during sulindac-induced apoptosis. All of these events were significantly suppressed by SB203580. Our results demonstrate a novel mechanism of sulindac-induced apoptosis in human MM cells, namely, accumulation of p53, Bax, and Bak in mitochondria mediated by p38 MAPK activation downstream of ROS production.


Subject(s)
Apoptosis/drug effects , Apoptosis/physiology , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Reactive Oxygen Species/metabolism , Sulindac/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism , Antineoplastic Agents/pharmacology , Caspases/metabolism , Cell Line, Tumor , Enzyme Activation/drug effects , Humans , Inhibitor of Apoptosis Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Multiple Myeloma/pathology , Neoplasm Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Survivin , Tumor Suppressor Protein p53/metabolism , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/metabolism
19.
Cancer Res ; 66(17): 8511-9, 2006 Sep 01.
Article in English | MEDLINE | ID: mdl-16951163

ABSTRACT

Glioblastoma is a severe type of primary brain tumor, and its highly invasive character is considered to be a major therapeutic obstacle. Several recent studies have reported that ionizing radiation (IR) enhances the invasion of tumor cells, but the mechanisms for this effect are not well understood. In this study, we investigated the possible signaling mechanisms involved in IR-induced invasion of glioma cells. IR increased the matrix metalloproteinase (MMP)-2 promoter activity, mRNA transcription, and protein secretion along with the invasiveness of glioma cells lacking functional PTEN (U87, U251, U373, and C6) but not those harboring wild-type (WT)-PTEN (LN18 and LN428). IR activated phosphatidylinositol 3-kinase (PI3K), Akt, and mammalian target of rapamycin, and blockade of these kinases by specific inhibitors (LY294002, Akt inhibitor IV, and rapamycin, respectively) and transfection of dominant-negative (DN) mutants (DN-p85 and DN-Akt) or WT-PTEN suppressed the IR-induced MMP-2 secretion in U251 and U373 cells. In addition, inhibitors of epidermal growth factor receptor (EGFR; AG490 and AG1478), Src (PP2), and p38 (SB203580), EGFR neutralizing antibody, and transfection of DN-Src and DN-p38 significantly blocked IR-induced Akt phosphorylation and MMP-2 secretion. IR-induced activation of EGFR was suppressed by PP2, whereas LY294002 and SB203580 did not affect the activations of p38 and PI3K, respectively. Finally, these kinase inhibitors significantly reduced the IR-induced invasiveness of these cells on Matrigel. Taken together, our findings suggest that IR induces Src-dependent EGFR activation, which triggers the p38/Akt and PI3K/Akt signaling pathways, leading to increased MMP-2 expression and heightened invasiveness of PTEN mutant glioma cells.


Subject(s)
ErbB Receptors/physiology , Glioma/pathology , Matrix Metalloproteinase 2/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Cell Line, Tumor , DNA Primers , ErbB Receptors/radiation effects , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Genes, Reporter , Glioblastoma/pathology , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/radiation effects , Neoplasm Invasiveness , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/radiation effects , Phosphorylation , Proto-Oncogene Proteins c-akt/radiation effects , Radiation, Ionizing , Rats , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/radiation effects , Transfection , p38 Mitogen-Activated Protein Kinases/radiation effects
20.
Biochem Pharmacol ; 72(10): 1228-36, 2006 Nov 15.
Article in English | MEDLINE | ID: mdl-16950207

ABSTRACT

Survivin, a member of the inhibitor of apoptosis protein (IAP) family, may be a good target for cancer therapy because it is expressed in a variety of human tumors but not in differentiated adult tissues. In the present study, we show that a combination of sulindac and arsenic trioxide (ATO) induces more extensive apoptosis than either drug alone in A549 human non-small cell lung carcinoma (NSCLC) cells. Treatment with sulindac/ATO reduced the expression of survivin and promoted major apoptotic signaling events, namely, collapse of the mitochondrial membrane potential, release of cytochrome c, and activation of caspases. Combined sulindac/ATO treatment did not significantly affect the levels of other members of the IAP family (XIAP, cIAP1 and cIAP2), indicating that the effects were specific to survivin. In addition, sulindac/ATO treatment induced the production of reactive oxygen species and the antioxidant N-acetyl-l-cysteine blocked the down-regulation of survivin and induction of apoptotic signaling by the combination of sulindac and ATO. Combined sulindac/ATO treatment also activated p53 expression, and inhibition of p53 expression by small interfering RNA (siRNA) prevented sulindac/ATO-induced down-regulation of survivin, suggesting that survivin expression is negatively regulated by p53. Overexpression of survivin reduced sulindac/ATO-induced apoptosis in A549 cells and reduction of survivin levels by siRNA sensitized the cells to sulindac/ATO-induced cell death. These results demonstrate that, in A549 human NSCLC cells, sulindac/ATO-induced apoptosis is mediated by the reactive oxygen species-dependent down-regulation of survivin.


Subject(s)
Apoptosis/drug effects , Arsenicals/pharmacology , Microtubule-Associated Proteins/biosynthesis , Neoplasm Proteins/biosynthesis , Oxides/pharmacology , Reactive Oxygen Species/metabolism , Sulindac/pharmacology , Arsenic Trioxide , Arsenicals/administration & dosage , Cell Line, Tumor , Cell Survival/drug effects , Down-Regulation , Drug Synergism , Humans , Inhibitor of Apoptosis Proteins , Lung Neoplasms/pathology , Oxides/administration & dosage , Sulindac/administration & dosage , Survivin
SELECTION OF CITATIONS
SEARCH DETAIL
...