Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
Cell Signal ; 115: 111029, 2024 03.
Article in English | MEDLINE | ID: mdl-38163576

ABSTRACT

Sirtuin 3 (SIRT3) regulates mitochondrial function as a mitochondrial deacetylase during oxidative stress. However, the specific regulatory mechanism and function of SIRT3 in radioresistant cancer cells are unclear. In this study, we aim to investigate how SIRT3 determines the susceptibility to glucose deprivation and its regulation in p53-based radioresistant head and neck cancer cells. We observed mitochondrial function using two established isogenic radioresistant subclones (HN3R-A [p53 null] and HN3R-B [p53 R282W]) with intratumoral p53 heterogeneity. Cell counting analysis was performed to evaluate cell proliferation and cell death. The correlation between the regulation of SIRT3 and enhancer of zeste homolog 2 (EZH2) was confirmed by immunoblotting and chromatin immunoprecipitation assay. p53-deficient radioresistant cells (HN3R-A) expression reduced SIRT3 levels and increased sensitivity to glucose deprivation due to mitochondrial dysfunction compared to other cells. In these cells, activation of SIRT3 significantly prevented glucose deprivation-induced cell death, whereas the loss of SIRT3 increased the susceptibility to glucose deficiency. We discovered that radiation-induced EZH2 directly binds to the SIRT3 promoter and represses the expression. Conversely, inhibiting EZH2 increased the expression of SIRT3 through epigenetic changes. Our findings indicate that p53-deficient radioresistant cells with enhanced EZH2 exhibit increased sensitivity to glucose deprivation due to SIRT3 suppression. The regulation of SIRT3 by EZH2 plays a critical role in determining the cell response to glucose deficiency in radioresistant cancer cells. Therefore, EZH2-dependent SIRT3 could be used as a predictive biomarker to select treatment options for patients with radiation-resistance.


Subject(s)
Head and Neck Neoplasms , Sirtuin 3 , Humans , Enhancer of Zeste Homolog 2 Protein/metabolism , Sirtuin 3/metabolism , Tumor Suppressor Protein p53/metabolism , Head and Neck Neoplasms/radiotherapy , Oxidative Stress
3.
Bioact Mater ; 25: 527-540, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37056267

ABSTRACT

Highly immunosuppressive tumor microenvironment containing various protumoral immune cells accelerates malignant transformation and treatment resistance. In particular, tumor-associated macrophages (TAMs), as the predominant infiltrated immune cells in a tumor, play a pivotal role in regulating the immunosuppressive tumor microenvironment. As a potential therapeutic strategy to counteract TAMs, here we explore an exosome-guided in situ direct reprogramming of tumor-supportive M2-polarized TAMs into tumor-attacking M1-type macrophages. Exosomes derived from M1-type macrophages (M1-Exo) promote a phenotypic switch from anti-inflammatory M2-like TAMs toward pro-inflammatory M1-type macrophages with high conversion efficiency. Reprogrammed M1 macrophages possessing protein-expression profiles similar to those of classically activated M1 macrophages display significantly increased phagocytic function and robust cross-presentation ability, potentiating antitumor immunity surrounding the tumor. Strikingly, these M1-Exo also lead to the conversion of human patient-derived TAMs into M1-like macrophages that highly express MHC class II, offering the clinical potential of autologous and allogeneic exosome-guided direct TAM reprogramming for arming macrophages to join the fight against cancer.

4.
Exp Ther Med ; 24(3): 541, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35978923

ABSTRACT

CD47 is expressed in all human cancer cells, including head and neck cancer, and initiates a signaling cascade to inhibit macrophage phagocytosis. However, the mechanism underlying CD47 overexpression has not been elucidated in radioresistant head and neck cancer. The present study demonstrated that decreased Tristetraprolin (TTP) expression induced a sustained overexpression of CD47 using reverse transcription-quantitative PCR and western blotting, and that CD47 overexpression prevented phagocytosis using a phagocytosis assay in a radioresistant HN31R cell line. Subsequently, using TTP transfection, RNA interference, duel-luciferase assay and EMSA, it was revealed that TTP transfection enhanced phagocytosis through degradation of CD47 mRNA by directly binding to CD47 AREs within the CD47 3'UTR. Based on our previous study, methylation-specific PCR and western blotting revealed that DNMT1 was overexpressed in radioresistant HN31R cell line and TTP expression was decreased epigenetically by DMNT1 associated DNA methylation. Overall, these findings provided novel insight into the role of TTP as a biomarker of CD47-positive head and neck cancer patients.

5.
J Control Release ; 346: 158-168, 2022 06.
Article in English | MEDLINE | ID: mdl-35452763

ABSTRACT

Therapy-induced senescence (TIS), a common outcome of current cancer therapy, is a known cause of late recurrence and metastasis and thus its eradication is crucial for therapy success. In this study, we introduced a conceptually novel strategy combining radiation-induced apoptosis-targeted chemotherapy (RIATC) with an effective glycolysis inhibitor, 2-deoxy-d-glucose (2DG) to target TIS. RIATC releases cytotoxic payload by amplification, continually increasing TIS, and this can be targeted by 2DG that stimulates an intrinsic apoptotic pathway in senescent cells, the senolysis; the senolytic 2DG also sensitizes cancer cells to chemo/radiation treatment. Anti-tumor efficacy of RIATC was investigated in numerous tumor models, and various cancer types were screened for TIS. Furthermore, in vitro evaluations of molecular markers of senescence, such as senescence-associated ß-galactosidase (SA-ß-Gal) assay, were performed to confirm that TIS was induced by RIATC therapy in MCF-7 cells. The combination therapy with 2DG proved to be effective in MCF-7 tumor-bearing mice that demonstrated feedback amplification of senolysis and successful inhibition of tumor growth. Our findings suggest that RIATC, when given together with 2DG, can overcome therapy-induced senescence and this combination is a promising strategy that enhances the therapeutic benefit of anti-cancer cytotoxic therapy.


Subject(s)
Antineoplastic Agents , Caspase 3 , Doxorubicin , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Caspase 3/metabolism , Deoxyglucose/therapeutic use , Doxorubicin/pharmacology , Humans , MCF-7 Cells , Mice , Peptides/pharmacology
6.
J Control Release ; 344: 26-38, 2022 04.
Article in English | MEDLINE | ID: mdl-35202743

ABSTRACT

Despite recent breakthroughs in the development of direct KRAS inhibitors and modulators, no drugs targeting pan-KRAS mutant cancers are clinically available. Here, we report a novel strategy to treat pan-KRAS cancers using a caspase-3 cleavable peptide-drug conjugate that exploits enhanced albumin metabolism in KRAS altered cancers to deliver a cytotoxic agent that can induce a widespread bystander killing effect in tumor cells. Increased albumin metabolism in KRAS mutant cancer cells induced apoptosis via the intracellular uptake of albumin-bound MPD1. This allowed caspase-3 upregulation activated MPD1 to release the payload and exert the non-selective killing of neighboring cancer cells. MPD1 exhibited potent and durable antitumor efficacy in mouse xenograft models with different KRAS genotypes. An augmentation of anti-cancer efficacy was achieved by the bystander killing effect derived from the caspase-3 mediated activation of MPD1. In summary, albumin metabolism-induced apoptosis, together with the bystander killing effect of MPD1 boosted by caspase-3 mediated activation, intensified the efficacy of MPD1 in KRAS mutant cancers. These findings suggest that this novel peptide-drug conjugate could be a promising breakthrough for the treatment in the targeting of pan-KRAS mutant cancers.


Subject(s)
Antineoplastic Agents , Neoplasms , Albumins , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Humans , Mice , Mutation , Neoplasms/drug therapy , Peptides , Proto-Oncogene Proteins p21(ras)/genetics , Xenograft Model Antitumor Assays
7.
Biomolecules ; 11(4)2021 04 18.
Article in English | MEDLINE | ID: mdl-33919657

ABSTRACT

EPHA3, a member of the EPH family, is overexpressed in various cancers. We demonstrated previously that EPHA3 is associated with radiation resistance in head and neck cancer via the PTEN/Akt/EMT pathway; the inhibition of EPHA3 significantly enhances the efficacy of radiotherapy in vitro and in vivo. In this study, we investigated the mechanisms of PTEN regulation through EPHA3-related signaling. Increased DNA methyltransferase 1 (DNMT1) and enhancer of zeste homolog 2 (EZH2) levels, along with increased histone H3 lysine 27 trimethylation (H3K27me3) levels, correlated with decreased levels of PTEN in radioresistant head and neck cancer cells. Furthermore, PTEN is regulated in two ways: DNMT1-mediated DNA methylation, and EZH2-mediated histone methylation through EPHA3/C-myc signaling. Our results suggest that EPHA3 could display a novel regulatory mechanism for the epigenetic regulation of PTEN in radioresistant head and neck cancer cells.


Subject(s)
Epigenetic Repression , Head and Neck Neoplasms/genetics , PTEN Phosphohydrolase/genetics , Radiation Tolerance , Receptor, EphA3/genetics , Cell Line, Tumor , DNA (Cytosine-5-)-Methyltransferase 1/genetics , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA Methylation , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/radiotherapy , Histone Code , Humans , PTEN Phosphohydrolase/metabolism , Receptor, EphA3/metabolism
8.
Exp Mol Med ; 53(3): 422-431, 2021 03.
Article in English | MEDLINE | ID: mdl-33727672

ABSTRACT

Sirt6 is involved in multiple biological processes, including aging, metabolism, and tumor suppression. Sirt1, another member of the sirtuin family, functionally overlaps with Sirt6, but its role in tumorigenesis is controversial. In this study, we focused on cell death in association with Sirt6/Sirt1 and reactive oxygen species (ROS) in head and neck squamous cell carcinomas (HNSCCs). Sirt6 induced cell death, as widely reported, but Sirt1 contributed to cell death only when it was suppressed by Sirt6 via regulation of MDM2. Sirt6 and Sirt6-mediated suppression of Sirt1 upregulated ROS, which further led to HNSCC cell death. These results provide insight into the molecular roles of Sirt6 and Sirt1 in tumorigenesis and could therefore contribute to the development of novel strategies to treat HNSCC.


Subject(s)
Biomarkers, Tumor/metabolism , Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms/pathology , Proto-Oncogene Proteins c-mdm2/metabolism , Sirtuin 1/metabolism , Sirtuins/metabolism , Animals , Apoptosis , Biomarkers, Tumor/genetics , Cell Proliferation , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/metabolism , Humans , Male , Mice , Mice, Nude , Prognosis , Proteolysis , Proto-Oncogene Proteins c-mdm2/genetics , Reactive Oxygen Species , Sirtuin 1/genetics , Sirtuins/genetics , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
9.
Cell Signal ; 77: 109820, 2021 01.
Article in English | MEDLINE | ID: mdl-33137455

ABSTRACT

The manner in which p53 maintains redox homeostasis and the means by which two key metabolic elements, glucose and glutamine, contribute to p53-dependent redox stability remain unclear. To elucidate the manner in which p53 deals with glucose-deprived, reactive oxygen species (ROS)-prone conditions in this regard, two isogenic cancer subclones (HN3R-A and HN3R-B) bearing distinct p53 mutations as an in vitro model of intratumoral p53 heterogeneity were identified. Following cumulative irradiation, the subclones showed a similar metabolic shift to aerobic glycolysis and increasing NADPH biogenesis for cellular defense against oxidative damage irrespective of p53 status. The radioresistant cancer cells became more sensitive to glycolysis-targeting drugs. However, in glucose-deprived and ROS-prone conditions, HN3R-B, the subclone with the original p53 increased the utilization of glutamine by GLS2, thereby maintaining redox homeostasis and ATP. Conversely, HN3R-A, the p53-deficient radioresistant subclone displayed an impairment in glutamine usage and high susceptibility to metabolic stresses as well as ROS-inducing agents despite the increased ROS scavenging system. Collectively, our findings suggest that p53 governs the alternative utilization of metabolic ingredients, such as glucose and glutamine, in ROS-prone conditions. Thus, p53 status may be an important biomarker for selecting cancer treatment strategies, including metabolic drugs and ROS-inducing agents, for recurrent cancers after radiotherapy.


Subject(s)
Glutamine/metabolism , Oxidative Stress/genetics , Tumor Suppressor Protein p53/metabolism , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Glucose/pharmacology , Glutaminase/metabolism , Glutathione/metabolism , Glycolysis , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Humans , NADP/metabolism , Oxidation-Reduction , Radiation Tolerance , Reactive Oxygen Species/metabolism , Signal Transduction/genetics , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics
10.
Cells ; 9(8)2020 08 07.
Article in English | MEDLINE | ID: mdl-32784606

ABSTRACT

Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) has attracted attention as a potential candidate for cancer therapy. However, many primary cancers are resistant to TRAIL, even when combined with standard chemotherapy. The mechanism of TRAIL resistance in cancer cells has not been fully elucidated. The TRAIL death receptor (DR) 3'-untranslated region (3'-UTR) is reported to contain AU-rich elements (AREs) that are important for regulating DR mRNA stability. However, the mechanisms by which DR mRNA stability is determined by its 3'-UTR are unknown. We demonstrate that tristetraprolin (TTP), an ARE-binding protein, has a critical function of regulating DR mRNA stability. DR4 mRNA contains three AREs and DR5 mRNA contains four AREs in 3'-UTR. TTP bound to all three AREs in DR4 and ARE3 in DR5 and enhanced decay of DR4/5 mRNA. TTP overexpression in colon cancer cells changed the TRAIL-sensitive cancer cells to TRAIL-resistant cells, and down-regulation of TTP increased TRAIL sensitivity via DR4/5 expression. Therefore, this study provides a molecular mechanism for enhanced levels of TRAIL DRs in cancer cells and a biological basis for posttranscriptional modification of TRAIL DRs. In addition, TTP status might be a biomarker for predicting TRAIL response when a TRAIL-based treatment is used for cancer.


Subject(s)
Neoplasms/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Tristetraprolin/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Neoplasms/drug therapy , RNA Processing, Post-Transcriptional
11.
Anticancer Res ; 40(5): 2627-2635, 2020 May.
Article in English | MEDLINE | ID: mdl-32366407

ABSTRACT

BACKGROUND/AIM: Head and neck squamous cell carcinoma (HNSCC) is an aggressive head and neck malignancy. The aim of this study was to elucidate the role of periostin (POSTN) in the epithelial-to-mesenchymal transition (EMT) process mediating the acquisition of radioresistance in HNSCC. MATERIALS AND METHODS: The expression levels of EMT hallmark genes including POSTN and Erk/Akt signaling pathways were compared between radiosensitive and radioresistant HNSCC cells. RESULTS: POSTN mRNA expression was higher in radioresistant HNSCC cells, and silencing POSTN significantly impaired their invasiveness under the effect of EMT process represented by up-regulation of mesenchymal markers and down-regulation of an epithelial marker. Expression levels of Erk and Akt were higher in radioresistant cells. CONCLUSION: POSTN in association with the Erk and Akt signaling pathways was up-regulated during the EMT process, leading to the conversion of radiosensitive to radioresistant HNSCC cells. POSTN may be a key marker for predicting the radioresistance and therapeutic target of HNSCC.


Subject(s)
Cell Adhesion Molecules/metabolism , Epithelial-Mesenchymal Transition , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Radiation Tolerance , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Down-Regulation , Epithelium/metabolism , Epithelium/pathology , Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms/genetics , Humans , Mesoderm/pathology , Neoplasm Invasiveness , Radiation Tolerance/genetics , Signal Transduction/genetics , Squamous Cell Carcinoma of Head and Neck/metabolism , Squamous Cell Carcinoma of Head and Neck/pathology
12.
Oncogene ; 38(19): 3729-3742, 2019 05.
Article in English | MEDLINE | ID: mdl-30664690

ABSTRACT

The role of p53 in genotoxic therapy-induced metabolic shift in cancers is not yet known. In this study, we investigated the role of p53 in the glycolytic shift in head and neck squamous cell carcinoma cell lines following irradiation. Isogenic p53-null radioresistant cancer cells established through cumulative irradiation showed decreased oxygen consumption and increased glycolysis with compromised mitochondria, corresponding with their enhanced sensitivity to drugs that target glycolysis. In contrast, radioresistant cancer cells with wild-type p53 preserved their primary metabolic profile with intact mitophagic processes and maintained their mitochondrial integrity. Moreover, we identified a previously unappreciated link between p53 and mitophagy, which limited the glycolytic shift through the BNIP3-dependent clearance of abnormal mitochondria. Thus, drugs targeting glycolysis could be used as an alternative strategy for overcoming radioresistant cancers, and the p53 status could be used as a biomarker for selecting participants for clinical trials.


Subject(s)
Head and Neck Neoplasms/metabolism , Membrane Proteins/metabolism , Mitophagy/physiology , Proto-Oncogene Proteins/metabolism , Squamous Cell Carcinoma of Head and Neck/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Cell Line, Tumor , Glycolysis/physiology , Head and Neck Neoplasms/radiotherapy , Humans , Male , Membrane Proteins/genetics , Mice, Inbred NOD , Mitochondria/metabolism , Mitochondria/pathology , Proto-Oncogene Proteins/genetics , Squamous Cell Carcinoma of Head and Neck/radiotherapy , Tumor Suppressor Protein p53/genetics , Xenograft Model Antitumor Assays
13.
Biomaterials ; 182: 35-43, 2018 11.
Article in English | MEDLINE | ID: mdl-30103170

ABSTRACT

Metronomic chemotherapy, which is defined as a low-dose and frequent administration of cytotoxic drugs without drug-free breaks, has been recently emerged as an alternative to traditional MTD therapy and has shown therapeutic benefit in breast cancer patients in numbers of clinical studies. Unlike MTD, metronomic chemotherapy acts by multiple mechanisms including antiangiogenic effect and immunomodulation, but the direct cytotoxic effect only playing a minor role due to the lowered dose. In this light, within the limits of p53-deficient breast cancer, we demonstrate the enhanced anticancer effect of metronomic chemotherapy using doxorubicin when combined with Chk1 inhibitor MK-8776 by specifically augmenting the direct cytotoxic effect on cancer cells. Since the oral drug is greatly favored in metronomic chemotherapy due to the frequent and potential long-term administration, we prepared an oral doxorubicin by producing an ionic complex with deoxycholic acid, which showed sufficient bioavailability and anticancer effect when administered orally. MK-8776 selectively enhanced the cytotoxic effect of low-concentration doxorubicin in p53-deficient breast cancer cells by abrogating the Chk1-dependent cell cycle arrest in vitro. Consistently, combining MK-8776 significantly improved the anticancer effect of the daily administered oral doxorubicin in p53-deficient breast cancer xenografts especially in a lower dose of doxorubicin without evident systemic toxicities. Combination therapy of MK-8776 and metronomic oral doxorubicin would be thus promising in the treatment of p53-deficient breast cancer benefited from the augmented direct cytotoxic effect and low risk of toxicities.


Subject(s)
Antibiotics, Antineoplastic/administration & dosage , Breast Neoplasms/drug therapy , Checkpoint Kinase 1/antagonists & inhibitors , Doxorubicin/administration & dosage , Protein Kinase Inhibitors/administration & dosage , Pyrazoles/administration & dosage , Pyrimidines/administration & dosage , Tumor Suppressor Protein p53/genetics , Administration, Metronomic , Administration, Oral , Animals , Antibiotics, Antineoplastic/therapeutic use , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Doxorubicin/therapeutic use , Female , Gene Deletion , Humans , Mice, Inbred BALB C , Mice, Nude , Protein Kinase Inhibitors/therapeutic use , Pyrazoles/therapeutic use , Pyrimidines/therapeutic use
14.
Cell Signal ; 47: 122-130, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29653204

ABSTRACT

Radiotherapy is a well-established therapeutic modality used in the treatment of many cancers. However, radioresistance remains a serious obstacle to successful treatment. Radioresistance can cause local recurrence and distant metastases in some patients after radiation treatment. Thus, many studies have attempted to identify effective radiosensitizers. Eph receptor functions contribute to tumor development, modulating cell-cell adhesion, invasion, neo-angiogenesis, tumor growth and metastasis. However, the role of EphA3 in radioresistance remains unclear. In the current study, we established a stable radioresistant head and neck cancer cell line (AMC HN3R cell line) and found that EphA3 was expressed predominantly in the radioresistant head and neck cancer cell line through DNA microarray, real time PCR and Western blotting. Additionally, we found that EphA3 was overexpressed in recurrent laryngeal cancer specimens after radiation therapy. EphA3 mediated the tumor invasiveness and migration in radioresistant head and neck cancer cell lines and epithelial mesenchymal transition- related protein expression. Inhibition of EphA3 enhanced radiosensitivity in the AMC HN 3R cell line in vitro and in vivo study. In conclusion, our results suggest that EphA3 is overexpressed in radioresistant head and neck cancer and plays a crucial role in the development of radioresistance in head and neck cancers by regulating the epithelial mesenchymal transition pathway.


Subject(s)
Epithelial-Mesenchymal Transition , Receptor Protein-Tyrosine Kinases/metabolism , Animals , Cell Line, Tumor , Cell Movement , Gamma Rays , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Head and Neck Neoplasms/radiotherapy , Humans , Male , Mice , Mice, Nude , Neoplasm Recurrence, Local , RNA Interference , RNA, Small Interfering/metabolism , Radiation Tolerance , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/genetics , Receptor, EphA3 , Transplantation, Heterologous
15.
Oral Oncol ; 78: 56-63, 2018 03.
Article in English | MEDLINE | ID: mdl-29496059

ABSTRACT

OBJECTIVES: PI3K/Akt/mTOR pathway is commonly activated in most cancers and is correlated with resistance to anticancer therapies such as radiotherapy. Therefore, PI3K is an attractive target for treating PI3K-associated cancers. MATERIAL AND METHODS: We investigated the basal expression and the expression after treatment of PI3K inhibitor or Src inhibitor of PI3K/Akt pathway-related proteins in AMC-HN3, AMC-HN3R, HN30 and HN31 cells by performing immunoblotting analysis. The sensitivity to PI3K inhibitors or Src inhibitor was analyzed by MTT assay and clonogenic assay. To determine the antitumoral activity of combination treatment with PI3K inhibitor and Src inhibitor, we used using xenograft mouse model. RESULTS: We found that PI3K regulatory subunit p85 was predominantly phosphorylated in radioresistant head and neck cancer cell line (HN31), which showed resistance to PI3K inhibitors. Next, we investigated mechanism through which PI3K p85 phosphorylation modulated response to PI3K inhibitors. Of note, constitutive activation of Src was found in HN31 cells and upon PI3K inhibitor treatment, restoration of p-Src was occurred. Src inhibitor improved the efficacy of PI3K inhibitor treatment and suppressed the reactivation of both Src and PI3K p85 in HN31 cells. Furthermore, downregulation of PI3K p85 expression by using a specific siRNA suppressed Src phosphorylation. CONCLUSIONS: Together, our results imply the novel role of the PI3K regulatory subunit p85 in the development of resistance to PI3K inhibitors and suggest the presence of a regulatory loop between PI3K p85 and Src in radioresistant head and neck cancers with constitutively active PI3K/Akt pathway.


Subject(s)
Drug Resistance, Neoplasm , Enzyme Inhibitors/therapeutic use , Head and Neck Neoplasms/enzymology , Phosphatidylinositol 3-Kinases/metabolism , Radiation Tolerance , Animals , Cell Line, Tumor , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/pathology , Head and Neck Neoplasms/radiotherapy , Humans , Male , Mice , Mice, Nude , Phosphatidylinositol 3-Kinases/chemistry , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Proto-Oncogene Proteins pp60(c-src)/metabolism , Xenograft Model Antitumor Assays
16.
Anticancer Res ; 36(10): 5117-5125, 2016 10.
Article in English | MEDLINE | ID: mdl-27798871

ABSTRACT

BACKGROUND/AIM: Recurrent laryngeal cancer often shows an aggressive phenotype after radiotherapy and does not respond to conventional therapeutic strategies. In this study, we investigated the contribution of furin to cellular invasiveness in radio-resistant laryngeal cancer. MATERIALS AND METHODS: Using previously established AMC-HN-3 and AMC-HN-8 cell lines from laryngeal carcinoma patients, recurrent laryngeal cancer models were generated by cumulative irradiation (AMC-HN-3-70Gy and AMC-HN-8-70Gy). Immunocytochemistry and western blotting were used to determine the epithelial-mesenchymal transition (EMT). Invasion capacity was assessed using an in vitro invasion assay. Zymography was used to assess metalloproteinase-2 (MMP-2) activity. Tumor xenografts were developed to compare growth rate and furin expression in vivo. Furin expression in 35 patients (45 samples) with salvage total laryngectomy after radiation-based treatment was assessed by laryngeal cancer tissue microarray. RESULTS: Both AMC-HN-3-70Gy and AMC-HN-8-70Gy cell lines underwent EMT following radiation. However, AMC-HN-3-70Gy cells showed increased cellular invasiveness, whereas AMC-HN-8-70Gy cells showed no difference. AMC-HN-3-70Gy cells also exhibited elevated furin expression with up-regulated expression of the active form of membrane type 1-matrix metalloproteinase (MT1-MMP)/MMP-2, whereas AMC-HN-8-70Gy cells did not show significant changes. After administration of a furin inhibitor (chloromethyl ketone (CMK)), AMC-HN-3-70Gy cells showed a significant decrease in MT1-MMP/MMP-2 expression and cellular invasiveness. Nine of 22 samples (40.9%) from salvage total laryngectomy and one of 13 pre-radiation samples (7.7%) had high furin expression. Post-radiation, furin expression increased in seven of 10 patients whose pre- and post-radiation samples were available; all-cancer mortality (three patients) was observed in this group. CONCLUSION: Together with EMT, furin activity may serve as an indicator of an aggressive cancer phenotype, suggesting that furin is a potentially useful target for recurrent laryngeal cancer.


Subject(s)
Epithelial-Mesenchymal Transition , Furin/metabolism , Laryngeal Neoplasms/metabolism , Laryngeal Neoplasms/radiotherapy , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/radiotherapy , Animals , Cell Line, Tumor , Humans , Male , Matrix Metalloproteinases/metabolism , Mice, Nude , Neoplasm Invasiveness
17.
Head Neck ; 38 Suppl 1: E1909-17, 2016 04.
Article in English | MEDLINE | ID: mdl-26713771

ABSTRACT

BACKGROUND: We attempted to elucidate the mechanism of cell death after radiation by studying how ß-catenin silencing controls the radiation sensitivity of radioresistant head and neck cancer cells. METHODS: The most radioresistant cancer cell line (AMC-HN-9) was selected for study. Targeted silencing of ß-catenin was used on siRNAs. Sensitivity to radiation was examined using clonogenic and methylthiazol tetrazolium (MTT) assays. RESULTS: A combination of irradiation plus ß-catenin silencing led to a significant reduction in the inherent radioresistance of AMC-HN-9 cells. Although expression of Ku70/80 was upregulated in AMC-HN-9 cells after irradiation, Ku70/80 was dramatically decreased in a combination of irradiation and ß-catenin silencing. Interestingly, irradiation-induced Ku70/80 was completely prevented by ß-catenin silencing-induced LKB1/AMP-activated protein kinase (LKB1/AMPK) signal. CONCLUSION: The LKB1/AMPK pathway might relay the signal between the Wnt/ß-catenin pathway and the Ku70/Ku80 DNA repair machinery, and play a decisive role in fine-tuning the responses of cancer cells to irradiation. © 2015 Wiley Periodicals, Inc. Head Neck 38: E1909-E1917, 2016.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Gene Silencing , Head and Neck Neoplasms/radiotherapy , Ku Autoantigen/metabolism , Radiation Tolerance , beta Catenin/genetics , AMP-Activated Protein Kinase Kinases , Cell Line, Tumor , Head and Neck Neoplasms/genetics , Humans , Protein Serine-Threonine Kinases/metabolism , RNA, Small Interfering , Wnt Signaling Pathway
18.
Hand Surg ; 20(2): 316-8, 2015.
Article in English | MEDLINE | ID: mdl-26051778

ABSTRACT

Ganglion is a common benign lesion that usually arises adjacent to the joints or tendons of the hand. However, an intratendinous ganglion is a rare condition. We report two cases of intratendinous ganglion of the extensor tendon of the hand which were treated with excision.


Subject(s)
Ganglion Cysts/diagnosis , Orthopedic Procedures/methods , Tendons/surgery , Female , Ganglion Cysts/surgery , Hand , Humans , Middle Aged , Tendons/pathology
19.
Arch Orthop Trauma Surg ; 134(9): 1227-35, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25027675

ABSTRACT

BACKGROUND: Although intramedullary nailing is an ideal treatment for subtrochanteric femoral fractures, it is technically challenging in fractures extending into the nail entry area and/or involving the lesser trochanter. Although the application of circumferential wire may facilitate reduction in these situations, its use remains controversial due to possible blood supply disturbances to underlying bone. In the present study, we evaluated complex subtrochanteric fractures treated by percutaneous cerclage wiring followed by intramedullary (IM) nailing for anatomical fracture reduction and union. METHODS: Twelve patients (mean age 48.3 years) with an unstable subtrochanteric fracture were prospectively treated. Indications of percutaneous cerclage wiring followed by IM nailing were a fracture extending proximally into the nail entry area deemed difficult to treat by anatomical reconstruction by IM nailing or a fracture with long oblique or spiral component. One or two cerclage wires were percutaneously applied for the temporary reduction of main fragments, and then, a cephalo-medullary or a reconstruction nail was fixed. We assessed radiologic results (union time, alignment), functional results, and complications. RESULTS: All 12 cases healed, without a bone graft, at an average of 19.1 weeks after surgery (range 16-24). In 11 cases, acceptable alignment was achieved (mean, valgus 0.3° extension 0.6°) with minimal leg-length discrepancy; the other exhibited 1 cm of shortening. All patients were able to return to pre-injury activity levels, and median Merle d'Aubigne score was 16.9 (15-18). No infection or implant-related complication was encountered to latest follow-up (minimum 12 months postoperatively). CONCLUSION: Temporary reduction by percutaneous wiring offers a means of satisfactory nailing in difficult subtrochanteric femoral fractures, and affords anatomical reconstruction and favorable bony union.


Subject(s)
Bone Wires , Fracture Fixation, Intramedullary/methods , Hip Fractures/surgery , Adult , Aged , Bone Nails , Female , Follow-Up Studies , Fracture Fixation, Intramedullary/instrumentation , Humans , Internal Fixators , Male , Middle Aged , Prospective Studies , Treatment Outcome , Young Adult
20.
Anticancer Res ; 34(3): 1449-55, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24596393

ABSTRACT

BACKGROUND/AIM: Autophagy is frequently activated in radioresistant cancer cells. In the present study, we evaluated the role of autophagy and transforming growth factor-activated kinase 1 (TAK1) in radioresistance. MATERIALS AND METHODS: TAK1 phosphorylation in MDA-MB231 breast cancer cells was evaluated by western blotting. The regulatory effects of the TAK1 inhibitor and autophagy inhibitor were assessed by cell morphology, cell survival and induction of apoptosis. RESULTS: Radiation induced the phosphorylation of TAK1, whereas the inhibition of TAK1 activity enhanced the cytotoxicity of radiation in MDA-MB231 cells. Autophagy inhibitors significantly enhanced radiation-induced apoptosis of MDA-MB231 cells. This augmentation in radiosensitivity seemed to result from the suppression of TAK1 activation. CONCLUSION: Inhibition of autophagy enhanced radiosensitivity through suppression of radiation-induced TAK1 activation, suggesting that the modulation of TAK1-induced autophagy may be a good therapeutic strategy to treat radioresistant breast cancer.


Subject(s)
Autophagy/drug effects , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Gene Expression Regulation, Neoplastic/drug effects , MAP Kinase Kinase Kinases/metabolism , Radiation Tolerance/drug effects , Signal Transduction/drug effects , Antifungal Agents/pharmacology , Antimalarials/pharmacology , Autophagy/radiation effects , Blotting, Western , Breast Neoplasms/drug therapy , Breast Neoplasms/radiotherapy , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Chloroquine/pharmacology , Female , Gamma Rays , Gene Expression Regulation, Neoplastic/radiation effects , Humans , Macrolides/pharmacology , Phosphorylation/drug effects , Signal Transduction/radiation effects , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...