Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Sci Rep ; 14(1): 12363, 2024 05 29.
Article in English | MEDLINE | ID: mdl-38811596

ABSTRACT

Radiotherapy is the standard treatment for glioblastoma (GBM), but the overall survival rate for radiotherapy treated GBM patients is poor. The use of adjuvant and concomitant temozolomide (TMZ) improves the outcome; however, the effectiveness of this treatment varies according to MGMT levels. Herein, we evaluated whether MGMT expression affected the radioresponse of human GBM, GBM stem-like cells (GSCs), and melanoma. Our results indicated a correlation between MGMT promoter methylation status and MGMT expression. MGMT-producing cell lines ACPK1, GBMJ1, A375, and MM415 displayed enhanced radiosensitivity when MGMT was silenced using siRNA or when inhibited by lomeguatrib, whereas the OSU61, NSC11, WM852, and WM266-4 cell lines, which do not normally produce MGMT, displayed reduced radiosensitivity when MGMT was overexpressed. Mechanistically lomeguatrib prolonged radiation-induced γH2AX retention in MGMT-producing cells without specific cell cycle changes, suggesting that lomeguatrib-induced radiosensitization in these cells is due to radiation-induced DNA double-stranded break (DSB) repair inhibition. The DNA-DSB repair inhibition resulted in cell death via mitotic catastrophe in MGMT-producing cells. Overall, our results demonstrate that MGMT expression regulates radioresponse in GBM, GSC, and melanoma, implying a role for MGMT as a target for radiosensitization.


Subject(s)
DNA Modification Methylases , DNA Repair Enzymes , Glioblastoma , Melanoma , Radiation Tolerance , Tumor Suppressor Proteins , Humans , Glioblastoma/genetics , Glioblastoma/radiotherapy , Glioblastoma/metabolism , Glioblastoma/pathology , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , Melanoma/genetics , Melanoma/metabolism , Melanoma/pathology , Melanoma/radiotherapy , DNA Modification Methylases/metabolism , DNA Modification Methylases/genetics , Cell Line, Tumor , Radiation Tolerance/genetics , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/radiation effects , Neoplastic Stem Cells/pathology , Promoter Regions, Genetic , DNA Methylation , DNA Repair , DNA Breaks, Double-Stranded/radiation effects , Gene Expression Regulation, Neoplastic , Temozolomide/pharmacology , Brain Neoplasms/genetics , Brain Neoplasms/radiotherapy , Brain Neoplasms/pathology , Brain Neoplasms/metabolism , Purines
2.
Cancer Sci ; 113(1): 145-155, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34714604

ABSTRACT

To improve the poor survival rate of lung cancer patients, we investigated the role of HDGF-related protein 3 (HRP-3) as a potential biomarker for lung cancer. The expression of endogenous HRP-3 in human lung cancer tissues and xenograft tumor models is indicative of its clinical relevance in lung cancer. Additionally, we demonstrated that HRP-3 directly binds to the E2F1 promoter on chromatin. Interestingly, HRP-3 depletion in A549 cells impedes the binding of HRP-3 to the E2F1 promoter; this in turn hampers the interaction between Histone H3/H4 and HDAC1/2 on the E2F1 promoter, while concomitantly inducing Histone H3/H4 acetylation around the E2F1 promoter. The enhanced Histone H3/H4 acetylation on the E2F1 promoter through HRP-3 depletion increases the transcription level of E2F1. Furthermore, the increased E2F1 transcription levels lead to the enhanced transcription of Cyclin E, known as the E2F1-responsive gene, thus inducing S-phase accumulation. Therefore, our study provides evidence for the utility of HRP-3 as a biomarker for the prognosis and treatment of lung cancer. Furthermore, we delineated the capacity of HRP-3 to regulate the E2F1 transcription level via histone deacetylation.


Subject(s)
Biomarkers, Tumor/metabolism , Cyclin E/metabolism , E2F1 Transcription Factor/genetics , Histone Deacetylases/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Lung Neoplasms/pathology , A549 Cells , Animals , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mice , Neoplasm Transplantation , Promoter Regions, Genetic , Signal Transduction
3.
Mol Cancer Ther ; 20(9): 1672-1679, 2021 09.
Article in English | MEDLINE | ID: mdl-34158343

ABSTRACT

AZD0530, a potent small-molecule inhibitor of the Src kinase family, is an anticancer drug used in the treatment of various cancers. In the case of glioblastoma (GBM), where resistance to radiotherapy frequently occurs, Src kinase is known as one of the molecules responsible for imparting radioresistance to GBM. Thus, we evaluated the effect of AZD0530 on the radiosensitivity of human GBM cells and human glioblastoma stem-like cells (GSCs). We show that Src activity of GBM and GSC is increased by radiation and inhibited by AZD0530, and using clonogenic assays, AZD0530 enhances the radiosensitivity of GBM and GSCs. Also, AZD0530 induced a prolongation of radiation-induced γH2AX without specific cell cycle and mitotic index changes, suggesting that AZD0530-induced radiosensitization in GBM cells and GSCs results from the inhibition of DNA repair. In addition, AZD0530 was shown to inhibit the radiation-induced EGFR/PI3K/AKT pathway, which is known to promote and regulate radioresistance and survival of GBM cells by radiation. Finally, mice bearing orthotopic xenografts initiated from GBM cells were then used to evaluate the in vivo response to AZD0530 and radiation. The combination of AZD0530 and radiation showed the longest median survival compared with any single modality. Thus, these results show that AZD0530 enhances the radiosensitivity of GBM cells and GSCs and suggest the possibility of AZD0530 as a clinical radiosensitizer for treatment of GBM.


Subject(s)
Benzodioxoles/pharmacology , Gene Expression Regulation, Neoplastic , Glioblastoma/radiotherapy , Neoplastic Stem Cells/radiation effects , Quinazolines/pharmacology , Radiation Tolerance/drug effects , Radiation-Sensitizing Agents/pharmacology , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Cell Cycle , Cell Proliferation , Female , Glioblastoma/drug therapy , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Mice , Mice, Nude , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
Cell Death Dis ; 11(5): 398, 2020 05 26.
Article in English | MEDLINE | ID: mdl-32457423

ABSTRACT

The poor therapeutic efficacy of non-small cell lung cancer (NSCLC) is partly attributed to the acquisition of chemoresistance. To investigate the mechanism underlying this resistance, we examined the potential link between kinesin light chain 4 (KLC4), which we have previously reported to be associated with radioresistance in NSCLC, and sensitivity to chemotherapy in human lung cancer cell lines. KLC4 protein levels in lung cancer cells correlated with the degree of chemoresistance to cisplatin treatment. Furthermore, KLC4 silencing enhanced the cytotoxic effect of cisplatin by promoting DNA double-strand breaks and apoptosis. These effects were mediated by interaction with the checkpoint kinase CHK2, as KLC4 knockdown increased CHK2 activation, which was further enhanced in combination with cisplatin treatment. In addition, KLC4 and CHEK2 expression levels showed negative correlation in lung tumor samples from patients, and KLC4 overexpression correlated negatively with survival. Our results indicate a novel link between the KLC4 and CHK2 pathways regulating DNA damage response in chemoresistance, and highlight KLC4 as a candidate for developing lung cancer-specific drugs and customized targeted molecular therapy.


Subject(s)
Checkpoint Kinase 1/antagonists & inhibitors , Checkpoint Kinase 2/antagonists & inhibitors , DNA Repair , Drug Resistance, Neoplasm , Kinesins/metabolism , Lung Neoplasms/drug therapy , Microtubule-Associated Proteins/metabolism , Molecular Targeted Therapy , Apoptosis/drug effects , Cell Line, Tumor , Checkpoint Kinase 1/metabolism , Checkpoint Kinase 2/metabolism , Cisplatin/pharmacology , Colorectal Neoplasms/pathology , DNA Damage , DNA Repair/drug effects , DNA-Activated Protein Kinase/metabolism , Down-Regulation/drug effects , Down-Regulation/genetics , Drug Resistance, Neoplasm/drug effects , Etoposide/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Gene Silencing/drug effects , Humans , Kinesins/genetics , Lung Neoplasms/enzymology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Signal Transduction/drug effects
5.
Cell Death Dis ; 10(3): 156, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30770789

ABSTRACT

Current lung cancer treatments are far from satisfactory; thus, finding novel treatment targets is crucial. We recently identified procollagen-lysine, 2-oxoglutarate 5-dioxygenase 3 (PLOD3), which is involved in fibrosis and tissue remodeling as a radioresistance-related protein in lung cancer cells; however, its mechanism is unclear. In this study, we designed human PLOD3-specific short interfering (si)RNAs and tested their effects on tumor growth inhibition in vitro and in vivo. PLOD3 knockdown overcame chemoresistance and decreased radioresistance by inducing caspase-3-dependent apoptosis in lung cancer cells. Furthermore, PLOD3 interacted with PKCδ to activate caspase-2,4-dependent apoptosis through ER-stress-induced IRE1α activation and the downstream unfolded-protein response pathway. In a mouse xenograft model, PLOD3 knockdown promoted radiation-induced tumor growth inhibition, without side effects. Moreover, lung cancer patients with high PLOD3 expression showed poorer prognosis than those with low PLOD3 expression upon radiotherapy, suggesting that PLOD3 promotes tumor growth. Therefore, PLOD3 siRNA suppresses radioresistance and chemoresistance by inducing apoptosis and renders PLOD3 as a candidate lung cancer biomarker. PLOD3 gene therapy might enhance the efficacy of radiotherapy or chemotherapy in lung cancer patients.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/genetics , Lung Neoplasms/metabolism , Procollagen-Lysine, 2-Oxoglutarate 5-Dioxygenase/genetics , Protein Kinase C-delta/metabolism , RNA, Small Interfering/pharmacology , Signal Transduction/genetics , A549 Cells , Animals , Apoptosis/genetics , Biomarkers, Tumor/genetics , Carcinogenesis/genetics , Cell Proliferation/genetics , DNA Damage/genetics , Endoplasmic Reticulum Stress/genetics , Gene Knockdown Techniques , Humans , Lung Neoplasms/pathology , Mice , Transfection , Tumor Burden/drug effects , Tumor Burden/radiation effects , Unfolded Protein Response , Xenograft Model Antitumor Assays
6.
Cell Death Dis ; 9(12): 1138, 2018 11 15.
Article in English | MEDLINE | ID: mdl-30442941

ABSTRACT

Procollagen-lysine, 2-oxoglutarate 5-dioxygenase (PLOD3), a membrane-bound homodimeric enzyme, hydroxylates lysyl residues in collagen-like peptides; however, its role in lung cancer is unknown. This study aimed to investigate the role of PLOD3 as a pro-metastatic factor and to elucidate the underlying mechanism. First, we experimentally confirmed the release of PLOD3 in circulation in animal models, rendering it a potential serum biomarker for lung cancer in humans. Thereafter, we investigated the effects of PLOD3 overexpression and downregulation on cancer cell invasion and migration in vitro and in vivo, using human lung cancer cell lines and a mouse tumor xenograft model, respectively. Further, PLOD3 levels were determined in lung tissue samples from lung cancer patients. Functional analyses revealed that PLOD3 interacts with STAT3, thereby expressing matrix metalloproteinases (MMP-2 and MMP-9) and with urokinase plasminogen activator (uPA) to enhance tumor metastasis. PLOD3 and the STAT3 pathway were significantly correlated in the metastatic foci of lung cancer patients; PLOD3-STAT3 levels were highly correlated with a poor prognosis. These results indicate that PLOD3 promotes lung cancer metastasis in a RAS-MAP kinase pathway-independent manner. Therefore, secreted PLOD3 serves as a potent inducer of lung cancer metastasis and a potential therapeutic target to enhance survival in lung cancer.


Subject(s)
Cell Proliferation/genetics , Lung Neoplasms/genetics , Procollagen-Lysine, 2-Oxoglutarate 5-Dioxygenase/genetics , STAT3 Transcription Factor/genetics , Animals , Cell Line, Tumor , Cell Movement/genetics , Gene Expression Regulation, Neoplastic , Heterografts , Humans , Kaplan-Meier Estimate , Lung Neoplasms/pathology , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 9/genetics , Mice , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasm Metastasis , Protein Binding/genetics , Urokinase-Type Plasminogen Activator/genetics
7.
Cell Death Dis ; 9(5): 496, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29717133

ABSTRACT

Kinesins act as molecular microtubule-dependent motor proteins and have various important cellular functions related to cell division, intracellular transport, and membrane trafficking. However, the function of kinesin light chain 4 (KLC4) in cancer, especially radioresistance, has not been previously described. Thus, we investigated KLC4 function in lung cancer cells and radioresistant R-H460 cells by analyzing alterations in radiosensitivity after gene knockdown with siRNA and by evaluating cellular phenotypes and xenograft tumor growth. KLC4 was upregulated in human lung cancer cell lines. Moreover, in paired clinical specimens of lung cancer patients, KLC4 expression was significantly higher in tumor tissues than in paired adjacent normal tissues. Fluorescence-activated cell sorting (FACS) analysis showed that apoptosis rates and cleaved poly (ADP-ribose) polymerase (PARP) and cleaved caspase-3 levels in KLC4-knockdown lung cancer cells were significantly increased compared with those in control cells. Colony formation decreased as the radiation dose increased in KLC4-knockdown lung cancer cells, demonstrating an essential role for KLC4 in radioresistance. Importantly, KLC4 silencing suppressed tumor growth in an in vivo xenograft model, accompanied by increased apoptosis. Finally, KLC4-knockdown cells exhibited impaired mitochondrial respiration, increased mitochondrial reactive oxygen species production, and enhanced mitochondrial calcium uptake, resulting in mitochondrial dysfunction. Thus, KLC4 as a kinesin superfamily-targeted therapy may represent a novel, effective anticancer strategy, particularly for patients showing radioresistance.


Subject(s)
Apoptosis/radiation effects , Calcium Signaling/radiation effects , Lung Neoplasms/radiotherapy , Microtubule-Associated Proteins/metabolism , Mitochondria/radiation effects , Radiation Tolerance , Uterine Cervical Neoplasms/radiotherapy , A549 Cells , Animals , Caspase 3/metabolism , Female , Gene Expression Regulation, Neoplastic , HeLa Cells , Humans , Kinesins , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Mice, Inbred BALB C , Mice, Nude , Microtubule-Associated Proteins/genetics , Mitochondria/metabolism , Mitochondria/pathology , Poly(ADP-ribose) Polymerases/metabolism , Reactive Oxygen Species/metabolism , Tumor Burden/radiation effects , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/pathology , Xenograft Model Antitumor Assays
8.
Cancer Biol Ther ; 17(2): 208-18, 2016.
Article in English | MEDLINE | ID: mdl-26901847

ABSTRACT

Tumor cell radioresistance is a major contributor to radiotherapy failure, highlighting the importance of identifying predictive biomarkers for radioresistance. In this work, we established a radioresistant H460 (RR-H460) cell line from parental radiosensitive H460 lung cancer cells by exposure to fractionated radiation. The radiation-resistant, anti-apoptotic phenotype of RR-H460 cell lines was confirmed by their enhanced clonogenic survival and increased expression of the radioresistance genes Hsp90 and Her-3. RR-H460 cells displayed characteristics of cancer stem-like cells (CSCs), including induction of the surface marker CD44 and stem cell markers Nanog, Oct4, and Sox2. RR-H460 cells also exhibited sphere formation and malignant behavior, further supporting a CSC phenotype. Using proteomic analyses, we identified 8 proteins that were up-regulated in RR-H460 CSC lines and therefore potentially involved in radioresistance and CSC-related biological processes. Notably, 4 of these-PAI-2, NOMO2, KLC4, and PLOD3-have not been previously linked to radioresistance. Depletion of these individual genes sensitized RR-H460 cells to radiotoxicity and additively enhancing radiation-induced apoptosis. Our findings suggest the possibility of integrating molecular targeted therapy with radiotherapy as a strategy for resolving the radioresistance of lung tumors.


Subject(s)
Carcinoma, Non-Small-Cell Lung/radiotherapy , Lung Neoplasms/radiotherapy , Proteomics , Radiation Tolerance/genetics , Apoptosis/radiation effects , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/radiation effects , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Neoplasm Proteins/biosynthesis , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/radiation effects
9.
Biochem Biophys Res Commun ; 469(1): 94-100, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26592665

ABSTRACT

The function of PSMC5 (proteasome 26S subunit, ATPase 5) in tumors, particularly with respect to cancer radioresistance, is not known. Here, we identified PSMC5 as a novel radiosensitivity biomarker, demonstrating that radiosensitive H460 cells were converted to a radioresistance phenotype by PSMC5 depletion. Exposure of H460 cells to radiation induced a marked accumulation of cell death-promoting reactive oxygen species, but this effect was blocked in radiation-treated H460 PSMC5-knockdown cells through downregulation of the p53-p21 pathway. Interestingly, PSMC5 depletion in H460 cells enhanced both AKT activation and MDM2 transcription, thereby promoting the degradation of p53 and p21 proteins. Furthermore, specific inhibition of AKT with triciribine or knockdown of MDM2 with small interfering RNA largely restored p21 expression in PSMC5-knockdown H460 cells. Our data suggest that PSMC5 facilitates the damaging effects of radiation in radiation-responsive H460 cancer cells and therefore may serve as a prognostic indicator for radiotherapy and molecular targeted therapy in lung cancer patients.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Apoptosis/radiation effects , LIM Domain Proteins/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/radiotherapy , Radiation Tolerance , Transcription Factors/metabolism , ATPases Associated with Diverse Cellular Activities , Cell Line, Tumor , Dose-Response Relationship, Radiation , Humans , Lung Neoplasms/pathology , Proteasome Endopeptidase Complex , Radiotherapy Dosage , Treatment Outcome
10.
Biochem Biophys Res Commun ; 449(4): 471-6, 2014 Jul 11.
Article in English | MEDLINE | ID: mdl-24857986

ABSTRACT

We previously identified hepatoma-derived growth factor-related protein-3 (HRP-3) as a radioresistant biomarker in p53 wild-type A549 cells and found that p53-dependent induction of the PUMA pathway was a critical event in regulating the radioresistant phenotype. Here, we found that HRP-3 knockdown regulates the radioresistance of p53-null H1299 cells through a distinctly different molecular mechanism. HRP-3 depletion was sufficient to cause apoptosis of H1299 cells by generating substantial levels of reactive oxygen species (ROS) through inhibition of the Nrf2/HO-1 antioxidant pathway. Subsequent, ROS-dependent and p53-independent NF-κB activation stimulated expression of c-Myc and Noxa proteins, thereby inducing the apoptotic machinery. Our results thus extend the range of targets for the development of new drugs to treat both p53 wild-type or p53-null radioresistant lung cancer cells.


Subject(s)
Apoptosis/drug effects , Lung Neoplasms/pathology , NF-kappa B/metabolism , Nuclear Proteins/genetics , Reactive Oxygen Species/metabolism , Cell Line, Tumor , Cytoskeletal Proteins , Gene Knockdown Techniques , Humans , Intracellular Signaling Peptides and Proteins , Lung Neoplasms/radiotherapy , Tumor Suppressor Protein p53/metabolism
11.
Biochem Biophys Res Commun ; 439(3): 333-9, 2013 Sep 27.
Article in English | MEDLINE | ID: mdl-24012673

ABSTRACT

Biomarkers based on functional signaling have the potential to provide greater insight into the pathogenesis of cancer and may offer additional targets for anticancer therapeutics. Here, we identified hepatoma-derived growth factor-related protein-3 (HRP-3) as a radioresistance-related gene and characterized the molecular mechanism by which its encoded protein regulates the radio- and chemoresistant phenotype of lung cancer-derived A549 cells. Knockdown of HRP-3 promoted apoptosis of A549 cells and potentiated the apoptosis-inducing action of radio- and chemotherapy. This increase in apoptosis was associated with a substantial generation of reactive oxygen species (ROS) that was attributable to inhibition of the Nrf2/HO-1 antioxidant pathway and resulted in enhanced ROS-dependent p53 activation and p53-dependent expression of PUMA (p53 upregulated modulator of apoptosis). Therefore, the HRP-3/Nrf2/HO-1/ROS/p53/PUMA cascade is an essential feature of the A549 cell phenotype and a potential radiotherapy target, extending the range of targets in multimodal therapies against lung cancer.


Subject(s)
Anticarcinogenic Agents/pharmacology , Lung Neoplasms/drug therapy , Lung Neoplasms/radiotherapy , Nuclear Proteins/genetics , Reactive Oxygen Species/metabolism , Tumor Suppressor Protein p53/metabolism , Apoptosis/drug effects , Apoptosis/radiation effects , Cell Line, Tumor , Cytoskeletal Proteins , Drug Resistance, Neoplasm , Heme Oxygenase-1/metabolism , Humans , Intracellular Signaling Peptides and Proteins , Lung/drug effects , Lung/metabolism , Lung/pathology , Lung/radiation effects , Lung Neoplasms/genetics , Lung Neoplasms/pathology , NF-E2-Related Factor 2/metabolism , Nuclear Proteins/metabolism , RNA Interference
12.
Cancer Lett ; 339(1): 15-24, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23900080

ABSTRACT

Although end-binding protein 1 (EB1) is well known to regulate microtubule dynamics, the role of EB1 in apoptosis of non-small cell lung cancer (NSCLC) is poorly understood. Here, we investigated the molecular mechanism by which EB1 regulates apoptosis in H460, A549, and H1299 cells. Depletion of EB1 in A549 and H1299 cells, which express high levels of EB1, induced cell death in a p53-independent manner through over-production of reactive oxygen species (ROS) and Bax induction. This phenomenon was potentiated in radiation-treated EB1-knockdown cells and was largely blocked by N-acetyl-L-cysteine, a scavenger of ROS. ROS accelerated the activation of nuclear factor-kappa B (NF-κB) to promote transcriptional activity of Bax, an action that was accompanied by cytochrome c translocation and apoptosis-inducing factor (AIF) release. The NF-κB inhibitor, BAY 11-7082, potently inhibited the apoptosis induced by EB1 knockdown and radiation treatment, in association with diminished activity of the mitochondrial death pathway. Conversely, ectopic overexpression of EB1 in H460 cells, which express low levels of EB1, remarkably abrogated radiation-induced apoptosis and NF-κB-mediated mitochondrial dysfunction. Our data provide the first demonstration that down-regulation of EB1 promotes NSCLC cell death by inducing ROS-mediated, NF-κB-dependent Bax signaling cascades, a process in which cytochrome c and AIF play important roles, indicating a potential therapeutic benefit of EB1 in lung cancer.


Subject(s)
Apoptosis , Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/metabolism , Microtubule-Associated Proteins/metabolism , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , bcl-2-Associated X Protein/metabolism , Apoptosis/genetics , Apoptosis/radiation effects , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , Cell Proliferation , Gene Expression , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/genetics , Microtubule-Associated Proteins/genetics , RNA Interference , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
13.
Biochem Biophys Res Commun ; 436(3): 436-42, 2013 Jul 05.
Article in English | MEDLINE | ID: mdl-23747726

ABSTRACT

The mechanisms by which activated Ras accelerates malignant transformation of normal cells are not fully understood. Here, we characterized the role and molecular mechanism of γ-catenin in regulating the malignant phenotype of Rat2 cells induced by codon 12-mutant K-Ras (K-Ras12V). Suppression of γ-catenin signaling by K-Ras12V was an early event and played a crucial role in promoting the acquisition of a highly metastatic phenotype of Rat2 cells. Notably, the gene encoding histone deacetylase 4 (HDAC4) was identified as a target of γ-catenin during this process. The transcription factor, lymphoid enhancer-binding factor-1 (Lef1), was involved in the modulation of HDAC4 transcription, and disruption of this pathway was a key event in promoting the invasion and migration of K-Ras12V-transduced Rat2 cells. Thus, our findings extend the range of targets for the development of new drugs for the therapy of oncogenic K-Ras-driven cancer.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Gene Expression Regulation, Neoplastic , Genes, ras , Histone Deacetylases/metabolism , gamma Catenin/metabolism , Animals , Cell Line , Cell Movement , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/pathology , Histone Deacetylases/genetics , Histone Deacetylases/pharmacology , Lymphoid Enhancer-Binding Factor 1/genetics , Lymphoid Enhancer-Binding Factor 1/metabolism , Neoplasm Invasiveness/pathology , Phenotype , Protein Interaction Mapping , Protein Transport , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Signal Transduction , Transcription, Genetic , beta Catenin/genetics , beta Catenin/metabolism , gamma Catenin/genetics
14.
Cancer Biol Ther ; 13(13): 1307-18, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22895072

ABSTRACT

Although tumor resistance remains a significant impediment to successful radiotherapy, associated regulatory markers and detailed molecular mechanisms underlying this phenomenon are not well defined. In this study, we identified inositol polyphosphate 4-phosphatase type II (INPP4B) as a novel marker of radioresistance by systematically analyzing Unigene libraries of laryngeal cancer. INPP4B was highly expressed in radioresistant laryngeal cancer cells and was induced by treatment with either radiation or anticancer drugs in various types of cancer cells. Ectopic INPP4B overexpression increased radioresistance and anticancer drug resistance by suppressing apoptosis in HEp-2 cells. Conversely, INPP4B depletion with small interfering RNA resensitized HEp-2 as well as A549 and H1299 cells to radiation- and anticancer drug-induced apoptosis. Furthermore, radiation-induced INPP4B expression was blocked by inhibition of extracellular signal-regulated kinase (ERK). INPP4B depletion significantly attenuated radiation-induced increases in Akt phosphorylation, indicating an association of INPP4B-mediated radioresistance with Akt survival signaling. Taken together, our data suggest that ERK-dependent induction of INPP4B triggers the development of a tumor-resistance phenotype via Akt signaling and identify INPP4B as a potentially important target molecule for resolving the radioresistance of cancer cells.


Subject(s)
Biomarkers, Tumor/biosynthesis , Laryngeal Neoplasms/enzymology , Phosphoric Monoester Hydrolases/biosynthesis , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis/radiation effects , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Drug Resistance, Neoplasm , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , HCT116 Cells , Humans , Laryngeal Neoplasms/drug therapy , Laryngeal Neoplasms/genetics , Laryngeal Neoplasms/radiotherapy , MCF-7 Cells , Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/metabolism , Phosphorylation/drug effects , Phosphorylation/radiation effects , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Radiation Tolerance
15.
J Biol Chem ; 286(32): 28619-31, 2011 Aug 12.
Article in English | MEDLINE | ID: mdl-21697093

ABSTRACT

Although much is known about interleukin (IL)-1ß and its role as a key mediator of cartilage destruction in osteoarthritis, only limited information is available on IL-1ß signaling in chondrocyte dedifferentiation. Here, we have characterized the molecular mechanisms leading to the dedifferentiation of primary cultured articular chondrocytes by IL-1ß treatment. IL-1ß or lipopolysaccharide, but not phorbol 12-myristate 13-acetate, retinoic acid, or epidermal growth factor, induced nicotinamide phosphoribosyltransferase (NAMPT) expression, showing the association of inflammatory cytokines with NAMPT regulation. SIRT1, in turn, was activated NAMPT-dependently, without any alteration in the expression level. Activation or inhibition of SIRT1 oppositevely regulates IL-1ß-mediated chondrocyte dedifferentiation, suggesting this protein as a key regulator of chondrocytes phenotype. SIRT1 activation promotes induction of ERK and p38 kinase activities, but not JNK, in response to IL-1ß. Subsequently, ERK and p38 kinase activated by SIRT1 also induce SIRT1 activation, forming a positive feedback loop to sustain downstream signaling of these kinases. Moreover, we found that the SIRT1-ERK complex, but not SIRT1-p38, is engaged in IL-1ß-induced chondrocyte dedifferentiation via a Sox-9-mediated mechanism. JNK is activated by IL-1ß and modulates dedifferentiation of chondrocytes, but this pathway is independent on NAMPT-SIRT1 signaling. Based on these findings, we propose that IL-1ß induces dedifferentiation of articular chondrocytes by up-regulation of SIRT1 activity enhanced by both NAMPT and ERK signaling.


Subject(s)
Cartilage, Articular/metabolism , Cell Dedifferentiation/physiology , Chondrocytes/metabolism , Cytokines/metabolism , Interleukin-1beta/metabolism , MAP Kinase Signaling System/physiology , Multienzyme Complexes/metabolism , Nicotinamide Phosphoribosyltransferase/metabolism , Sirtuin 1/metabolism , Animals , Carcinogens/pharmacology , Cartilage, Articular/cytology , Cell Dedifferentiation/drug effects , Cells, Cultured , Chondrocytes/cytology , Cytokines/genetics , Humans , Interleukin-1beta/genetics , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/metabolism , Multienzyme Complexes/genetics , Nicotinamide Phosphoribosyltransferase/genetics , Rabbits , Sirtuin 1/genetics , Tetradecanoylphorbol Acetate/pharmacology , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
16.
Proteomics ; 10(14): 2589-604, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20461716

ABSTRACT

The nature of the molecules underlying the radioresistance phenotype of laryngeal cancer cells remains to be established. We initially generated radioresistant laryngeal cancer cell lines from human HEp-2 cells with fractionated radiation. These RR-HEp-2 cells and isolated clones displayed more radioresistant and anti-apoptotic phenotypes than parental HEp-2 cells after radiation. Characteristics of RR-Hep-2 cell lines were confirmed by upregulation of radioresistance-related genes, such as epidermal growth factor receptor, Hsp90, and Bcl-xl. Subsequently, we examined proteome changes between HEp-2 and RR-HEp-2 cells and identified 16 proteins showing significantly altered expression levels. Interestingly, protein expression of chloride intracellular channel 1 (CLIC1) was markedly suppressed in RR-HEp-2 cells, compared with non-irradiated control cells. Suppression of CLIC1 with an indanyloxyacetic acid-94 or small interfering RNA led to radioresistance in HEp-2 cells by suppressing the radiation-induced cellular ROS level. However, ectopic overexpression of CLIC1 induced radiosensitivity in RR-HEp-2 cells via induction of ROS level after radiation, suggesting that the protein acts as a positive regulator of ROS production. Our results collectively indicate that suppression of CLIC1 contributes to acquisition of the radioresistance phenotype of laryngeal cancer cells via inhibition of ROS production, implying that this protein is an important candidate molecule for radiotherapy in radioresistant laryngeal cancer cells.


Subject(s)
Chloride Channels/metabolism , Proteomics/methods , Radiation Tolerance , Reactive Oxygen Species/metabolism , Cell Death/radiation effects , Cell Line, Tumor , Chloride Channels/antagonists & inhibitors , Humans , Neoplasm Proteins/metabolism , Radiation Tolerance/radiation effects , Radiation, Ionizing , Reproducibility of Results
SELECTION OF CITATIONS
SEARCH DETAIL