Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Int J Mol Sci ; 24(19)2023 Sep 28.
Article in English | MEDLINE | ID: mdl-37834109

ABSTRACT

Melanogenesis, the intricate process of melanin synthesis, is central to skin pigmentation and photoprotection and is regulated by various signaling pathways and transcription factors. To develop potential skin-whitening agents, we used B16F1 melanoma cells to investigate the inhibitory effects of anhydrous alum on melanogenesis and its underlying molecular mechanisms. Anhydrous alum (KAl(SO4)2) with high purity (>99%), which is generated through the heat-treatment of hydrated alum (KAl(SO4)2·12H2O) at 400 °C, potentiates a significant reduction in melanin content without cytotoxicity. Anhydrous alum downregulates the master regulator of melanogenesis, microphthalmia-associated transcription factor (MITF), which targets key genes involved in melanogenesis, thereby inhibiting α-melanocyte-stimulating hormone (α-MSH)-induced melanogenesis. Phosphorylation of the cAMP response element-binding protein, which acts as a co-activator of MITF gene expression, is attenuated by anhydrous alum, resulting in compromised MITF transcription. Notably, anhydrous alum promoted extracellular signal-regulated kinase phosphorylation, leading to the impaired nuclear localization of MITF. Overall, these results demonstrated the generation and mode of action of anhydrous alum in B16F1 cells, which constitutes a promising option for cosmetic or therapeutic use.


Subject(s)
Melanins , alpha-MSH , Melanins/metabolism , alpha-MSH/metabolism , Monophenol Monooxygenase/metabolism , Microphthalmia-Associated Transcription Factor/genetics , Microphthalmia-Associated Transcription Factor/metabolism , Cell Line, Tumor
2.
Hum Exp Toxicol ; 41: 9603271221089257, 2022.
Article in English | MEDLINE | ID: mdl-35417658

ABSTRACT

BACKGROUND: Remdesivir is an anti-viral drug that inhibits RNA polymerase. In 2020, remdesivir was recognized as the most promising therapeutic agents against coronavirus disease 2019 (COVID-19). However, the effects of remdesivir on cancers have hardly been studied. PURPOSE: Here, we reported that the anti-carcinogenic effect of remdesivir on SKOV3 cells, one of human ovarian cancer cell lines. RESEARCH DESIGN: We anlalyzed the anti-carcarcinogenic effect of remdesivir in SKOV3 cells by performing in vitro cell assay and western blotting. RESULTS: WST-1 showed that remdesivir decreased cell viability in SKOV3 cells. Experiments conducted by Muse Cell Analyzer showed that remdesivir-induced apoptosis in SKOV3 cells. We found that the expression level of FOXO3, Bax, and Bim increased, whereas Bcl-2, caspase-3, and caspase-7 decreased by remdesivir in SKOV3 cells. Furthermore, we observed that intracellular reactive oxygen species (ROS) level increased after treatment of remdesivir in SKOV3 cells. Interestingly, cytotoxicity of remdesivir decreased after treatment of N-Acetylcysteine. CONCLUSION: Taken together, our results demonstrated that remdesivir has an anti-carcinogenic effect on SKOV3 cells vis up-regulation of reactive oxygen species, which suggests that remdesivir could be a promising reagent for treatment of ovarian cancer.


Subject(s)
Anticarcinogenic Agents , COVID-19 Drug Treatment , Ovarian Neoplasms , Adenosine Monophosphate/analogs & derivatives , Alanine/analogs & derivatives , Anticarcinogenic Agents/pharmacology , Apoptosis , Cell Line, Tumor , Cell Proliferation , Female , Humans , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Reactive Oxygen Species/metabolism
3.
J Pers Med ; 12(2)2022 Feb 09.
Article in English | MEDLINE | ID: mdl-35207737

ABSTRACT

BACKGROUND: Gallbladder cancer is commonly associated with inflammation, which indicates that inflammation-related cytokines and cytokine receptors are related to the progression of gallbladder cancers. Interleukin 4 (IL4) is a well-known cytokine that promotes the differentiation of naive helper T cells (Th0) to T helper type 2 cells (Th2). IL13 is a cytokine that is secreted by Th2 cells. IL4 and IL13 are closely related in immune responses. However, the role of IL4Rα and IL13Rα1 signaling pathway has not been fully understood in the development of gallbladder cancer. METHODS: In human gallbladder carcinomas, the expression of IL4Rα and IL13Rα1 were evaluated with immunohistochemical staining in tissue microarray tissue sections. After knockdown of IL4Rα or IL13Rα1, cell assays to measure the proliferation and apoptosis and Western blotting analysis were conducted in SNU308 human gallbladder cancer cells. Since Janus kinases2 (JAK2) was considered as one of the down-stream kinases under IL4Rα and IL13Rα1 complex, the same kinds of experiments were performed in SNU308 cells treated with AZD1480, Janus-associated kinases2 (JAK2) inhibitor, to demonstrate the cytotoxic effect of AZD1480 in SNU308 cells. RESULTS: Immunohistochemical expression of IL4Rα was significantly associated with the expression of IL13Rα1 in human carcinoma tissue. In univariate analysis, nuclear expression of IL4Rα, cytoplasmic expression of IL4Rα, nuclear expression of IL13Rα1, and cytoplasmic expression of IL13Rα1 were significantly associated with shorter overall survival and shorter relapse-free survival. Multivariate analysis revealed nuclear expression of IL4Rα as an independent poor prognostic indicator of overall survival and relapse-free survival. Then, we found that knockdown of IL4Rα or IL13Rα1 decreased viability and induced apoptosis in SNU308 cells via activation of FOXO3 and similarly, AZD1480 decreased viability and induced apoptosis in SNU308 cells with dose dependent manner. CONCLUSIONS: Taken together, our results suggest that IL4Rα and IL13Rα1 might be involved in the development of human gallbladder cancer cells and IL4Rα and IL13Rα1 complex/JAK2 signaling pathway could be efficient therapeutic targets for gallbladder cancer treatment.

4.
J Pers Med ; 11(4)2021 Apr 08.
Article in English | MEDLINE | ID: mdl-33917914

ABSTRACT

Previously, we reported a close relationship between type II IL4Rα and IL13Rα1 complex and poor outcomes in renal cell carcinoma (RCC). In this study, we investigated the clinicopathologically significant oncogenic role of IL13Rα2, a kind of the independent receptor for IL13, in 229 RCC patients. The high expression of IL13Rα2 was closely related to relapse-free survival in specific cancers in univariate and multivariate analysis. Then, the oncogenic role of IL13Rα2 was evaluated by performing in vitro assays for cell proliferation, cell cycle arrest, and apoptosis in A498, ACHN, Caki1, and Caki2, four kinds of RCC cells after transfection of siRNA against IL13Rα2. Cell proliferation was suppressed, and apoptosis was induced in A498, ACHN, Caki1, and Caki2 cells by knockdown of IL13Rα2. Interestingly, the knockdown of IL13Rα2 decreased the phosphorylation of JAK2 and increased the expression of FOXO3. Furthermore, the knockdown of IL13Rα2 reduced the protein interaction among IL13Rα2, phosphorylated JAK2, and FOXO3. Since phosphorylation of JAK2 was regulated by IL13Rα2, we tried to screen a novel JAK2 inhibitor from the FDA-approved drug library and selected telmisartan, a clinically used medicine against hypertension, as one of the strongest candidates. Telmisartan treatment decreased the cell proliferation rate and increased apoptosis in A498, ACHN, Caki1, and Caki2 cells. Mechanistically, telmisartan treatment decreased the phosphorylation of JAK2 and increased the expression of FOXO3. Taken together, these results suggest that IL13Rα2 regulates the progression of RCC via the JAK2/FOXO3-signaling path pathway, which might be targeted as the novel therapeutic option for RCC patients.

5.
Biomolecules ; 11(3)2021 02 27.
Article in English | MEDLINE | ID: mdl-33673494

ABSTRACT

Best known as a transcriptional factor, CCCTC-binding factor (CTCF) is a highly conserved multifunctional DNA-binding protein with 11 zinc fingers. It functions in diverse genomic processes, including transcriptional activation/repression, insulation, genome imprinting and three-dimensional genome organization. A big surprise has recently emerged with the identification of CTCF engaging in the repair of DNA double-strand breaks (DSBs) and in the maintenance of genome fidelity. This discovery now adds a new dimension to the multifaceted attributes of this protein. CTCF facilitates the most accurate DSB repair via homologous recombination (HR) that occurs through an elaborate pathway, which entails a chain of timely assembly/disassembly of various HR-repair complexes and chromatin modifications and coordinates multistep HR processes to faithfully restore the original DNA sequences of broken DNA sites. Understanding the functional crosstalks between CTCF and other HR factors will illuminate the molecular basis of various human diseases that range from developmental disorders to cancer and arise from impaired repair. Such knowledge will also help understand the molecular mechanisms underlying the diverse functions of CTCF in genome biology. In this review, we discuss the recent advances regarding this newly assigned versatile role of CTCF and the mechanism whereby CTCF functions in DSB repair.


Subject(s)
CCCTC-Binding Factor/metabolism , DNA Breaks, Double-Stranded , Animals , CCCTC-Binding Factor/chemistry , DNA Repair/genetics , Homologous Recombination/genetics , Humans , Models, Biological , Promoter Regions, Genetic/genetics
6.
J Food Biochem ; : e13373, 2020 Aug 03.
Article in English | MEDLINE | ID: mdl-32743825

ABSTRACT

Auricularia auricula (A. auricula) has been reported to have positive health effects. Therefore, this study was conducted to explore possible mechanisms of A. auricula-induced anticancer activity in hepatocellular carcinoma (HCC) cells. First, using proliferative assay including MTT assay and real-time cell electronic sensing technique, we founded that A. auricula has an antiproliferative effect on various cancer cell lines. Among five cancer cell lines, we focused on huh-7 cell line, HCC cell line, due to that A. auricula has most dramatic antiproliferative effects on huh-7 cell line. Following experiments, we founded that its antiproliferative effects was related with apoptosis-inducing activities. For more investigation, a two-dimensional electrophoresis based-proteomic analysis (2DE-GE) was employed for identification of possible target-related proteins of A. auricula-induced apoptosis. Among seven identified proteins, we focused on peroxiredoxin1 (PRDX1), which has been known as an anti-oxidative enzyme. We confirmed downregulation of expression of PRDX1 following A. auricula treatment in mRNA and protein level. In order to obtain a more validation of the correlation of A. auricula-induced cell death and anti-oxidative enzyme. We investigated the level of anti-oxidative enzymes, total glutathione (GSSG/GSH), and superoxide dismutase (SOD) levels in treated cells and PRDX1 gene-silenced cells. GSH and SOD levels were decreased in the treated cells and PRDX1 gene-silenced cells. Our findings suggest that A. auricula is a potent inducer of apoptosis in HCC cells via PRDX1-inhibition pathways. PRACTICAL APPLICATIONS: Hepatocellular carcinoma (HCC) is among the leading causes of cancer-related mortality. The principal treatment is surgical resection or liver transplantation. However, in most patients with HCC the diagnosis is often late, thereby excluding the patients from definitive surgical resection. Chemotherapy and radiotherapy are generally ineffective. Newer treatments are needed with several being in development. In this research article, we provide regulation mechanism of PRDX1 in HCC. PRDX1 has a proliferative effect and play a role in cancer development or progression. Overexpression of PRDX1 in cancer cells implies the role of PRDX1 in the cancer therapy. PRDX1 is currently being investigated as a new target for gene therapy in cancer. A. auricula is an apoptotic inducer of HCC cells through PRDX1 pathway. Regulation of PRDX1 in HCC may contribute to cancer treatment. Therefore, the potentials of targeting apoptosis would be a viable therapeutic strategy to improve the outcome of HCC patients.

7.
Environ Toxicol ; 35(12): 1318-1325, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32656944

ABSTRACT

In this study, we report the potential of cannabidiol, one of the major cannabis constituents, for enhancing osteoblastic differentiation in U2OS and MG-63 cells. Cannabidiol increased the expression of Angiopoietin1 and the enzyme activity of alkaline phosphatase in U2OS and MG-63. Invasion and migration assay results indicated that the cell mobility was activated by cannabidiol in U2OS and MG-63. Western blotting analysis showed that the expression of tight junction related proteins such as Claudin1, Claudin4, Occuludin1, and ZO1 was increased by cannabidiol in U2OS and MG-63. Alizarin Red S staining analysis showed that calcium deposition and mineralization was enhanced by cannabidiol in U2OS and MG-63. Western blotting analysis indicated that the expression of osteoblast differentiation related proteins such as distal-less homeobox 5, bone sialoprotein, osteocalcin, type I collagen, Runt-related transcription factor 2 (RUNX2), osterix (OSX), and alkaline phosphatase was time dependently upregulated by cannabidiol in U2OS and MG-63. Mechanistically, cannabidiol-regulated osteoblastic differentiation in U2OS and MG-63 by strengthen the protein-protein interaction among RUNX2, OSX, or the phosphorylated p38 mitogen-activated protein kinase (MAPK). In conclusion, cannabidiol increased Angiopoietin1 expression and p38 MAPK activation for osteoblastic differentiation in U2OS and MG-63 suggesting that cannabidiol might provide a novel therapeutic option for the bone regeneration.


Subject(s)
Angiopoietin-1/metabolism , Cannabidiol/pharmacology , Cell Differentiation/drug effects , Osteoblasts/drug effects , Osteogenesis/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism , Alkaline Phosphatase/metabolism , Cell Line , Cell Movement/drug effects , Core Binding Factor Alpha 1 Subunit/metabolism , Humans , Osteoblasts/cytology , Osteoblasts/metabolism , Phosphorylation
8.
Int J Mol Sci ; 21(15)2020 Jul 26.
Article in English | MEDLINE | ID: mdl-32722598

ABSTRACT

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) induces apoptosis in various tumor cells without affecting most normal cells. Despite being in clinical testing, novel strategies to induce TRAIL-mediated apoptosis are in need to overcome cancer cell unresponsiveness and resistance. Plasma-activated medium (PAM) markedly stimulates reactive oxygen/nitrogen species (ROS/RNS)-dependent apoptosis in cancer cells. We investigate the capability of PAM and TRAIL (PAM/TRAIL) combination therapy to overcome TRAIL resistance and improve the anticancer efficacy of TRAIL. The combinatorial treatment of PAM and TRAIL shows synergistic effects on growth inhibition in TRAIL-resistant cancer cells via augmented apoptosis by two attributes. DR5 (TRAIL-R2) transcription by CHOP is upregulated in a PAM-generated ROS/RNS-dependent manner, and PAM itself upregulates PTEN expression mediated by suppression of miR-425 which is involved in Akt inactivation, leading to increased apoptosis induction. Treatment of cancer cell lines with the antioxidant N-acetylcysteine reduces the extent of membrane dysfunction and the expression of both CHOP-DR5 and miR-425-PTEN axes, attenuating PAM/TRAIL-induced cancer cell apoptosis. These data suggest that PAM/TRAIL treatment is a novel approach to sensitizing cancer cells to TRAIL-induced apoptosis and overcoming TRAIL resistance. PAM is a promising candidate for further investigations as a chemotherapeutic sensitizer in the treatment of cancer.


Subject(s)
Gene Expression Regulation, Neoplastic/drug effects , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Oxidative Stress/drug effects , Plasma Gases/pharmacology , Receptors, TNF-Related Apoptosis-Inducing Ligand/biosynthesis , Signal Transduction/drug effects , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Up-Regulation/drug effects , A549 Cells , Apoptosis/drug effects , HeLa Cells , Hep G2 Cells , Humans , MicroRNAs/metabolism , Neoplasms/drug therapy , Neoplasms/pathology , RNA, Neoplasm/metabolism
9.
Aging (Albany NY) ; 12(6): 4727-4741, 2020 02 11.
Article in English | MEDLINE | ID: mdl-32045366

ABSTRACT

In this study, we aimed to identify a candidate drug that can activate endogenous Angiopoietin 1 (Ang1) expression via drug repositioning as a pharmacological treatment for avascular osteonecrosis. After incubation with 821 drugs from the Food and Drug Administration (FDA)-approved drug library, Ang1 expression in U2OS cell culture media was examined by ELISA. Metformin, the first-line medication for treatment of type 2 diabetes, was selected as a candidate for in vitro and in vivo experimental evaluation. Ang1 was induced, and alkaline phosphatase activity was increased by metformin treatment in U2OS and MG63 cells. Wound healing and migration assay showed increased osteoblastic cell mobility by metformin treatment in U2OS and MG63 cells. Metformin upregulated expression of protein markers for osteoblastic differentiation in U2OS and MG63 cells but inhibited osteoclastic differentiation in Raw264.7 cells. Metformin (25 mg/kg) protected against ischemic necrosis in the epiphysis of the rat femoral head by maintaining osteoblast/osteocyte function and vascular density but inhibiting osteoclast activity in the necrotic femoral head. These findings provide novel insight into the specific biomarkers that are targeted and regulated by metformin in osteoblast differentiation and contribute to understanding the effects of these FDA-approved small-molecule drugs as novel therapeutics for ischemic osteonecrosis.


Subject(s)
Angiogenesis Inducing Agents/administration & dosage , Cell Differentiation/drug effects , Ischemia/physiopathology , Metformin/administration & dosage , Osteoblasts/drug effects , Osteoclasts/drug effects , Osteonecrosis/physiopathology , Angiopoietin-1/metabolism , Angiopoietin-1/physiology , Animals , Cell Line, Tumor , Femur Head/blood supply , Femur Head/physiopathology , Humans , Ischemia/complications , Male , Osteonecrosis/complications , Rats, Sprague-Dawley
10.
Sci Rep ; 9(1): 15042, 2019 10 21.
Article in English | MEDLINE | ID: mdl-31636298

ABSTRACT

Fibrosarcoma is a skin tumor that is frequently observed in humans, dogs, and cats. Despite unsightly appearance, studies on fibrosarcoma have not significantly progressed, due to a relatively mild tumor severity and a lower incidence than that of other epithelial tumors. Here, we focused on the role of a recently-found dermis zinc transporter, ZIP13, in fibrosarcoma progression. We generated two transformed cell lines from wild-type and ZIP13-KO mice-derived dermal fibroblasts by stably expressing the Simian Virus (SV) 40-T antigen. The ZIP13-/- cell line exhibited an impairment in autophagy, followed by hypersensitivity to nutrient deficiency. The autophagy impairment in the ZIP13-/- cell line was due to the low expression of LC3 gene and protein, and was restored by the DNA demethylating agent, 5-aza-2'-deoxycytidine (5-aza) treatment. Moreover, the DNA methyltransferase activity was significantly increased in the ZIP13-/- cell line, indicating the disturbance of epigenetic regulations. Autophagy inhibitors effectively inhibited the growth of fibrosarcoma with relatively minor damages to normal cells in xenograft assay. Our data show that proper control over autophagy and zinc homeostasis could allow for the development of a new therapeutic strategy to treat fibrosarcoma.


Subject(s)
Autophagy , Cation Transport Proteins/deficiency , Dermis/metabolism , Fibrosarcoma/pathology , Animals , Autophagy/drug effects , Azacitidine/pharmacology , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Cell Death/drug effects , Cell Line , DNA (Cytosine-5-)-Methyltransferases/metabolism , Disease Models, Animal , Down-Regulation/drug effects , Down-Regulation/genetics , Ethylenediamines/pharmacology , Fibrosarcoma/genetics , Humans , Mice, Knockout , Microtubule-Associated Proteins/metabolism , Zinc/metabolism
11.
Cancers (Basel) ; 11(9)2019 Sep 18.
Article in English | MEDLINE | ID: mdl-31540495

ABSTRACT

Specific kinds of interleukin (IL) receptors are known to mediate lymphocyte proliferation and survival. However, recent reports have suggested that the high expression of IL4Rα and IL13Rα1 in tumor tissue might be associated with tumorigenesis in several kinds of tumor. We found that a significant association between mRNA level of IL4Rα or IL13Rα1 and the poor prognosis of renal cell carcinoma (RCC) from the public database (http://www.oncolnc.org/). Then, we evaluated the clinicopathological significance of the immunohistochemical expression of IL4Rα and IL13Rα1 in 199 clear cell RCC (CCRCC) patients. The individual and co-expression patterns of IL4Rα and IL13Rα1 were significantly associated with cancer-specific survival (CSS) and relapse-free survival (RFS) in univariate analysis. Multivariate analysis indicated IL4Rα-positivity and co-expression of IL4Rα and IL13Rα1 as the independent indicators of shorter CSS and RFS of CCRCC patients. For the in vitro evaluation of the oncogenic role of IL4Rα and IL13Rα1 in RCC, we knock-downed IL4Rα or IL13Rα1 and observed that the cell proliferation rate was decreased, and the apoptosis rate was increased in A498 and ACHN cells. Furthermore, we examined the possible role of Janus kinase 2 (JAK2), well-known down-stream tyrosine kinase under the heterodimeric receptor complex of IL4Rα and IL13Rα1. Interestingly, JAK2 interacted with Forkhead box O3 (FOXO3) to cause tyrosine-phosphorylation of FOXO3. Silencing IL4Rα or JAK2 in A498 and ACHN cells reduced the interaction between JAK2 and FOXO3. Moreover, pharmacological inhibition of JAK2 induced the nuclear localization of FOXO3, leading to increase apoptosis and decrease cell proliferation rate in A498 and ACHN cells. Taken together, these results suggest that IL4Rα and IL13Rα1 might be involved in the progression of RCC through JAK2/FOXO3 pathway, and their expression might be used as the novel prognostic factor and therapeutic target for RCC patients.

12.
Nucleic Acids Res ; 47(17): 9160-9179, 2019 09 26.
Article in English | MEDLINE | ID: mdl-31340001

ABSTRACT

The pleiotropic CCCTC-binding factor (CTCF) plays a role in homologous recombination (HR) repair of DNA double-strand breaks (DSBs). However, the precise mechanistic role of CTCF in HR remains largely unclear. Here, we show that CTCF engages in DNA end resection, which is the initial, crucial step in HR, through its interactions with MRE11 and CtIP. Depletion of CTCF profoundly impairs HR and attenuates CtIP recruitment at DSBs. CTCF physically interacts with MRE11 and CtIP and promotes CtIP recruitment to sites of DNA damage. Subsequently, CTCF facilitates DNA end resection to allow HR, in conjunction with MRE11-CtIP. Notably, the zinc finger domain of CTCF binds to both MRE11 and CtIP and enables proficient CtIP recruitment, DNA end resection and HR. The N-terminus of CTCF is able to bind to only MRE11 and its C-terminus is incapable of binding to MRE11 and CtIP, thereby resulting in compromised CtIP recruitment, DSB resection and HR. Overall, this suggests an important function of CTCF in DNA end resection through the recruitment of CtIP at DSBs. Collectively, our findings identify a critical role of CTCF at the first control point in selecting the HR repair pathway.


Subject(s)
CCCTC-Binding Factor/genetics , Carrier Proteins/genetics , Homologous Recombination/genetics , MRE11 Homologue Protein/genetics , Nuclear Proteins/genetics , DNA Breaks, Double-Stranded , DNA Repair/genetics , Endodeoxyribonucleases , HeLa Cells , Humans , Protein Binding/genetics , Recombinational DNA Repair/genetics , Zinc Fingers/genetics
13.
J Exp Clin Cancer Res ; 38(1): 267, 2019 Jun 18.
Article in English | MEDLINE | ID: mdl-31215499

ABSTRACT

BACKGROUND: FAM83H was initially identified as a protein essential for dental enamel formation. Recent reports have shown that FAM83H is also involved in the progression of human cancers in conjunction with tumor-associated molecules, such as MYC and ß-catenin. However, the role of FAM83H in sarcoma has not yet been investigated. METHODS: The expression and roles of FAM83H and ß-catenin were evaluated in human osteosarcomas from 34 patients and osteosarcoma cells. RESULTS: The expression of nuclear FAM83H, cytoplasmic FAM83H, and ß-catenin were significantly associated with each other and significantly associated with shorter survival of osteosarcoma patients by univariate analysis. In multivariate analysis, cytoplasmic expression of FAM83H was an independent indicator of shorter survival of osteosarcoma patients (overall survival; P <  0.001, relapse-free survival; P <  0.001). In U2OS, MG63, and KHOS/NP osteosarcoma cells, the knock-down of FAM83H decreased proliferation and invasion activity and overexpression of FAM83H increased proliferation and invasion activity. In KHOS/NP cells, knock-down of FAM83H significantly inhibited, and overexpression of FAM83H significantly increased in vivo growth of cells. In addition, the knock-down of FAM83H decreased protein expression of ß-catenin, active ß-catenin, cyclin D1, vimentin, and snail. Overexpression of FAM83H increased protein expression of ß-catenin, active ß-catenin, cyclin D1, vimentin, and snail. However, the expression of ß-catenin mRNA was not significantly altered with knock-down or overexpression of FAM83H. In addition, FAM83H and ß-catenin shown to directly interact via immunoprecipitation and nuclear and cytoplasmic localization of ß-catenin was decreased with knock-down of FAM83H. Moreover, the ubiquitination and proteasomal degradation of ß-catenin was increased with knock-down of FAM83H. CONCLUSIONS: This study suggests that FAM83H is involved in the progression of osteosarcomas via a mechanism involving the stabilization of ß-catenin and the promotion of proliferation and invasiveness of osteosarcomas.


Subject(s)
Bone Neoplasms/pathology , Osteosarcoma/pathology , Proteins/metabolism , beta Catenin/chemistry , beta Catenin/metabolism , Adult , Animals , Bone Neoplasms/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Proliferation , Cytoplasm/metabolism , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Neoplasm Invasiveness , Neoplasm Transplantation , Osteosarcoma/metabolism , Protein Stability , Survival Analysis , Tissue Array Analysis , Up-Regulation
14.
Oncol Lett ; 15(6): 9687-9696, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29928344

ABSTRACT

The outcome of chemotherapy for osteosarcoma have improved during the past decade and more patients have access to combination chemotherapy, but there has been no significant clinical progress in the patient survival rate. Recently, forkhead-box O3 (FOXO3) was identified as a pivotal transcription factor responsible for the transcriptional regulation of genes associated with suppression of cancer. The purpose of the present study was to screen small chemicals activating FOXO3 and elucidate their underlying mechanism. Using a drug discovery platform based on the phosphorylation status of FOXO3 in osteosarcoma cells, mitoxantrone (MTZ), a type of DNA-damaging agent, was selected as a possible FOXO3 activator from the food and drug administration-approved drug library. MTZ treatments significantly inhibited the phosphorylation level of Akt-pS473 and caused nuclear localization of FOXO3 in osteosarcoma cells. MTZ treatment inhibited proliferation in osteosarcoma cells in vitro, whereas silencing FOXO3 potently attenuates MTZ-mediated apoptosis in osteosarcoma cells. Taken together, the results indicated that MTZ induces apoptosis in osteosarcoma cells through an Akt/FOXO3-dependent mechanism.

15.
Int J Oncol ; 53(2): 703-712, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29901072

ABSTRACT

Cyclin-dependent kinases (CDK) are considered to be potential targets of anticancer drugs that can interrupt the uncontrolled division of cancer cells. In this study, we selected two selective CDK inhibitors, AT7519 and SNS­032, from current clinical trials and examined their anticancer and radiosensitizing effects in a cervical cancer model. SNS­032 was found to be more potent than AT7519, with a lower half maximal inhibitory concentration (IC50) value. Both AT7519 and SNS­032 induced the apoptosis, premature senescence and cytostasis of cervical cancer cells, which led to the attenuation of tumor growth in vivo. Moreover, using these CDK inhibitors together with radiation synergistically inhibited tumor growth in a human xenograft tumor model. The concomitant activation of the p53 tumor suppressor and the suppression of cell cycle checkpoint responses mediated by Chk1 led to the cytostasis of cervical cancer cells. Finally, AT7519 and SNS­032 inhibited cancer cell migration, invasion and angiogenesis in vitro, and suppressed lung metastases in a spontaneous metastasis model. On the whole, the findings of this study indicate that the utilization of AT7519 and SNS­032 as part of an adjuvant treatment may help control cervical cancer progression.


Subject(s)
Antineoplastic Agents/pharmacology , Cyclin-Dependent Kinases/antagonists & inhibitors , Lung Neoplasms/therapy , Oxazoles/pharmacology , Piperidines/pharmacology , Pyrazoles/pharmacology , Radiation-Sensitizing Agents/pharmacology , Thiazoles/pharmacology , Uterine Cervical Neoplasms/therapy , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Synergism , Female , Gene Expression Regulation, Neoplastic , HeLa Cells , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/secondary , Mice , Uterine Cervical Neoplasms/enzymology , Xenograft Model Antitumor Assays
16.
Mol Cells ; 41(4): 362-372, 2018 Apr 30.
Article in English | MEDLINE | ID: mdl-29487276

ABSTRACT

High mobility group box 2 (HMGB2) is an abundant, chromatin-associated, non-histone protein involved in transcription, chromatin remodeling, and recombination. Recently, the HMGB2 gene was found to be significantly downregulated during senescence and shown to regulate the expression of senescent-associated secretory proteins. Here, we demonstrate that HMGB2 transcription is repressed by p21 during radiation-induced senescence through the ATM-p53-p21 DNA damage signaling cascade. The loss of p21 abolished the downregulation of HMGB2 caused by ionizing radiation, and the conditional induction of p21 was sufficient to repress the transcription of HMGB2. We also showed that the p21 protein binds to the HMGB2 promoter region, leading to sequestration of RNA polymerase and transcription factors E2F1, Sp1, and p300. In contrast, NF-Y, a CCAAT box-binding protein complex, is required for the expression of HMGB2, but NF-Y binding to the HMGB2 promoter was unaffected by either radiation or p21 induction. A proximity ligation assay results confirmed that the chromosome binding of E2F1 and Sp1 was inhibited by p21 induction. As HMGB2 have been shown to regulate premature senescence by IR, targeting the p21-mediated repression of HMGB2 could be a strategy to overcome the detrimental effects of radiation-induced senescence.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/genetics , HMGB2 Protein/genetics , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adenocarcinoma/radiotherapy , Adenocarcinoma of Lung , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Line, Tumor , Cellular Senescence/physiology , Cellular Senescence/radiation effects , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Colorectal Neoplasms/radiotherapy , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA Damage , Down-Regulation , Fibroblasts/metabolism , Fibroblasts/pathology , Fibroblasts/radiation effects , HMGB2 Protein/metabolism , HT29 Cells , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/radiotherapy , Promoter Regions, Genetic , Radiotherapy , Transcription, Genetic , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
17.
Biochem Biophys Res Commun ; 478(3): 1409-15, 2016 09 23.
Article in English | MEDLINE | ID: mdl-27569283

ABSTRACT

Angiogenesis is closely associated with osteoblast differentiation. Previously, we demonstrated that bone formation can be accelerated by treatment with COMP-Angiopoietin1, a known angiogenic factor. Angiopoietin1 (Ang1) is a specific growth factor that generates stable and mature vasculature through the Tie2 receptor. In this study, we aimed to identify a novel drug that can activate endogenous Ang1 expression as a pharmacological treatment for bone formation. Therefore, Ang1 expression was examined in U2OS osteoblast-like cells treated with 770 drugs from a library of Food and Drug Administration (FDA)-approved drugs by using ELISA for Ang1. l-thyroxine was selected as a novel drug candidate. l-Thyroxine is a synthetic form of the hormone thyroxine, which is used to treat patients with hypothyroidism. Enzyme-linked immunosorbent assays (ELISAs) were performed to test whether Ang1 is induced in a dose-dependent manner in human osteoblast-like cell lines, U2OS and MG63. The effects of l-thyroxine on osteoblast differentiation and mineralization were evaluated by alkaline phosphatase (ALP) activity and Alizarin red s staining. To determine the molecular mechanism, the expression of proteins related to bone formation and differentiation, such as type I collagen (COL1A1), osteocalcin (OC), bone sialoprotein (BSP), distal-less homeobox 5 (Dlx5), Runt-related transcription factor 2 (Runx2), osterix (OSX), and ALP, was tested by Western blotting analysis. Consequently, l-thyroxine induced Ang1 expression in a dose-dependent manner in both U2OS and M63 cells, which was confirmed by ELISA and Western blotting. Also, l-thyroxine activated ALP activity in U2OS and MG63 cells as well as ALP expression. Furthermore, l-thyroxine enhanced the expression of COL1A1, Runx2, OC, BSP, Dlx5, and OSX mRNA and proteins. Taken together, we demonstrated that l-thyroxine increased Ang1 expression and induces bone formation, differentiation, and mineralization in U2OS and MG63 cell lines, which suggests that l-thyroxine could be a potential bone production agent.


Subject(s)
Angiopoietin-1/metabolism , Cell Differentiation/drug effects , Osteoblasts/cytology , Osteoblasts/metabolism , Thyroxine/pharmacology , Biomarkers/metabolism , Calcification, Physiologic/drug effects , Cell Line, Tumor , Enzyme-Linked Immunosorbent Assay , Humans , Osteoblasts/drug effects , Thyroxine/chemistry , Up-Regulation/drug effects
18.
Oncotarget ; 7(5): 6074-87, 2016 Feb 02.
Article in English | MEDLINE | ID: mdl-26756216

ABSTRACT

Cardiac glycosides are clinically used for cardiac arrhythmias. In this study, we investigated the mechanism responsible for anti-cancer and radiosensitizing effects of lanatoside C in colorectal cancer cells. Lanatoside C-treated cells showed classic patterns of autophagy, which may have been caused by lanatoside C-induced mitochondrial aggregation or degeneration. This mitochondrial dysfunction was due to disruption of K+ homeostasis, possibly through inhibition of Na+/K+-ATPase activity. In addition, lanatoside C sensitized HCT116 cells (but not HT-29 cells) to radiation in vitro. γ-H2AX, a representative marker of DNA damage, were sustained longer after combination of irradiation with lanatoside C, suggesting lanatoside C impaired DNA damage repair processes. Recruitment of 53BP1 to damaged DNA, a critical initiation step for DNA damage repair signaling, was significantly suppressed in lanatoside C-treated HCT116 cells. This may have been due to defects in the RNF8- and RNF168-dependent degradation of KDM4A/JMJD2A that increases 53BP1 recruitment to DNA damage sites. Although lanatoside C alone reduced tumor growth in the mouse xenograft tumor model, combination of lanatoside C and radiation inhibited tumor growth more than single treatments. Thus, lanatoside C could be a potential molecule for anti-cancer drugs and radiosensitizing agents.


Subject(s)
Colorectal Neoplasms/drug therapy , DNA Repair/drug effects , Lanatosides/pharmacology , Mitochondria/drug effects , Animals , Autophagy/drug effects , Cell Growth Processes/drug effects , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , DNA Damage , HCT116 Cells , HT29 Cells , Humans , Mice , Mitochondria/genetics , Radiation Tolerance/drug effects , Radiation-Sensitizing Agents/pharmacology , Random Allocation , Signal Transduction
19.
PLoS One ; 10(4): e0123133, 2015.
Article in English | MEDLINE | ID: mdl-25849377

ABSTRACT

Suppressor of cytokine signaling (SOCS) family is an important negative regulator of cytokine signaling and deregulation of SOCS has been involved in many types of cancer. All cervical cancer cell lines tested showed lower expression of SOCS1, SOCS3, and SOCS5 than normal tissue or cell lines. The immunohistochemistry result for SOCS proteins in human cervical tissue also confirmed that normal tissue expressed higher level of SOCS proteins than neighboring tumor. Similar to the regulation of SOCS in other types of cancer, DNA methylation contributed to SOCS1 downregulation in CaSki, ME-180, and HeLa cells. However, the expression of SOCS3 or SOCS5 was not recovered by the inhibition of DNA methylation. Histone deacetylation may be another regulatory mechanism involved in SOCS1 and SOCS3 expression, however, SOCS5 expression was neither affected by DNA methylation nor histone deacetylation. Ectopic expression of SOCS1 or SOCS3 conferred radioresistance to HeLa cells, which implied SOCS signaling regulates the response to radiation in cervical cancer. In this study, we have shown that SOCS expression repressed by, in part, epigenetically and altered SOCS1 and SOCS3 expression could contribute to the radiosensitive phenotype in cervical cancer.


Subject(s)
DNA Methylation , Gene Expression Regulation, Neoplastic , Histones/metabolism , RNA Interference , Radiation Tolerance/genetics , Suppressor of Cytokine Signaling Proteins/antagonists & inhibitors , Uterine Cervical Neoplasms/genetics , Acetylation , Blotting, Western , Cells, Cultured , Cervix Uteri/metabolism , Cytokines/metabolism , Down-Regulation , Female , Humans , Immunoenzyme Techniques , Promoter Regions, Genetic/genetics , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Radiotherapy , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/metabolism , Transcription Factors/metabolism , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/radiotherapy
20.
Int J Oncol ; 46(1): 342-50, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25338966

ABSTRACT

The tumor suppressor p53 plays a key role in regulation of the cell cycle, apoptosis and senescence in response to various stresses. We screened a library of 7920 chemical compounds for the p53 activator and identified N-[2-(dimethylamino)ethyl]-2,3-dimethyl-4-oxo-4H-pyrido[1,2-a]thieno[2,3-d]pyrimidine-9-carboxamide (PTP), which significantly increased p53-mediated reporter activity in colorectal cancer cells. PTP was found to induce p53 protein and activated transcription of downstream genes, such as p21 and PUMA, in HCT116 cells, leading to growth delay, G1-phase cell cycle arrest, cell senescence and cell death. Proximity ligation assay revealed that PTP weakened the interaction between p53 and murine double minute 2 (MDM2) in situ, thereby inhibiting MDM2-mediated p53 degradation. Although DNA damage has been known to promote phosphorylation of p53 and MDM2, thereby preventing their interaction and stabilizing p53, PTP did not cause DNA damage or activate any DNA damage response signaling. Instead, phosphorylation of p53 was mediated by Erk1/2 MAP kinase. In addition, PTP induced acetylation of p53 at Lys382 in a p300-dependent manner, but sirtuin (SIRT)1 and histone deacetylase (HDAC)1, a well-known p53-regulating deacetylase, were not involved. In the present study, the novel anticancer agent PTP was shown to cause the accumulation of p53 by inducing multiple post-translational modifications, as well as cell cycle arrest, senescence and cell death.


Subject(s)
Antineoplastic Agents/pharmacology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Pyrimidines/pharmacology , Pyrimidinones/pharmacology , Thiophenes/pharmacology , Tumor Suppressor Protein p53/agonists , Acetylation/drug effects , Colorectal Neoplasms/genetics , G1 Phase Cell Cycle Checkpoints/drug effects , G1 Phase Cell Cycle Checkpoints/genetics , Gene Expression Regulation, Neoplastic/drug effects , HCT116 Cells , HT29 Cells , Humans , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...