Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Biochem Pharmacol ; 212: 115581, 2023 06.
Article in English | MEDLINE | ID: mdl-37146834

ABSTRACT

The therapeutic activity of paclitaxel against ovarian carcinoma is relatively low due to the frequent occurrence of chemoresistance and disease recurrence. We found earlier that a combination of curcumin and paclitaxel reduces cell viability and promotes apoptosis in paclitaxel-resistant (i.e., taxol-resistant, Txr) ovarian cancer cells. In the present study, we first used RNA sequencing (RNAseq) analysis to identify genes that are upregulated in Txr cell lines but downregulated by curcumin in ovarian cancer cells. The nuclear factor kappa B (NFκB) signaling pathway was shown to be upregulated in Txr cells. Furthermore, based on the protein interaction database BioGRID, we found that Smad nuclear interacting protein 1 (SNIP1) may be involved in regulating the activity of NFκB in Txr cells. Accordingly, curcumin upregulated SNIP1 expression, which in turn downregulated the pro-survival genes Bcl-2 and Mcl-1. Using shRNA-guided gene silencing, we found that SNIP1 depletion reversed the inhibitory effect of curcumin on NFκB activity. Moreover, we identified that SNIP1 enhanced NFκB protein degradation, thereby suppressing NFκB/p65 acetylation, which is involved in the inhibitory effect of curcumin on NFκB signaling. The transcription factor early growth response protein 1 (EGR1) was shown to represent an upstream transactivator of SNIP1. Consequently, we show that curcumin inhibits NFκB activity by modulating the EGR1/SNIP1 axis to attenuate p65 acetylation and protein stability in Txr cells. These findings provide a new mechanism to account for the effects of curcumin in inducing apoptosis and reducing paclitaxel resistance in ovarian cancer cells.


Subject(s)
Carcinoma , Curcumin , Ovarian Neoplasms , Female , Humans , Paclitaxel/pharmacology , Paclitaxel/therapeutic use , NF-kappa B/metabolism , Curcumin/pharmacology , Curcumin/therapeutic use , Cell Line, Tumor , Neoplasm Recurrence, Local , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Drug Resistance, Neoplasm , RNA-Binding Proteins
2.
Cancer Sci ; 113(8): 2616-2626, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35639349

ABSTRACT

More than 90% of ovarian cancer deaths are due to relapse following development of chemoresistance. Our main objective is to better understand the molecular mechanism underlying paclitaxel resistance (taxol resistance, Txr) in ovarian cancer. Here, we observed that the linker histone H1.0 is upregulated in paclitaxel-resistant ovarian cancer cells. Knockdown of H1.0 significantly downregulates the androgen receptor (AR) and sensitizes paclitaxel-resistant SKOV3/Txr and 2774/Txr cell lines to paclitaxel. Conversely, ectopic expression of H1.0 upregulates AR and increases Txr in parental SKOV3 and MDAH2774 cells. Notably, H1.0 upregulation is associated with disease recurrence and poor survival in a subset of ovarian cancer subjects. Inhibition of PI3K significantly reduces H1.0 mRNA and protein levels in paclitaxel-resistant cells, suggesting the involvement of the PI3K/AKT signaling pathway. Knockdown of H1.0 and AR also downregulates the Txr genes ABCB1 and ABCG2 in paclitaxel-resistant cells. Our data show that H1.0 induces GCN5 expression and histone acetylation, thereby enhancing Txr gene transactivation. These findings suggest that Txr in ovarian cancer involves the PI3K/AKT pathway and leads to upregulation of histone H1.0, recruitment of GCN5 and AR, followed by upregulation of a subgroup of Txr genes that include ABCB1 and ABCG2. This study is the first report describing the relationship between histone H1.0 and GCN5 that cooperate to induce AR-dependent Txr in ovarian cancer cells.


Subject(s)
Drug Resistance, Neoplasm , Ovarian Neoplasms , Paclitaxel , Receptors, Androgen , p300-CBP Transcription Factors , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Regulation, Neoplastic , Histones/metabolism , Humans , Neoplasm Recurrence, Local/genetics , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Paclitaxel/pharmacology , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , p300-CBP Transcription Factors/genetics , p300-CBP Transcription Factors/metabolism
3.
Biochem Pharmacol ; 186: 114456, 2021 04.
Article in English | MEDLINE | ID: mdl-33556340

ABSTRACT

Ovarian cancer is poorly treatable due, at least in part, to induced drug resistance to taxol- and cisplatin-based chemotherapy. Recent studies showed that ectopic overexpression of toll-like receptor 4 (TLR4) in ovarian cancer cells leads to upregulation of the androgen receptor (AR) and transactivation of taxol resistance genes, thereby causing chemoresistance. In the present study, we examined the signaling pathways involving TLR4 and interleukin 6 (IL-6) that enhance AR expression. Based on transcriptomic analysis, we show that IL-6 functions as a hub gene among the upregulated genes in taxol-treated TLR4-overexpressing ovarian cancer cells. Both the TLR4 activator taxol and IL-6 can induce AKT phosphorylation, whereas TLR4 knockdown or inhibition of the IL-6 signal transducer GP130 abrogates AKT activation. Furthermore, expression of AR and IL-6 is downregulated in TLR4-knockdown, taxol-resistant cells. In addition, TLR4 knockdown inhibits GP130 and IL-6 receptor alpha (IL6Rα) activities, indicating that TLR4 plays a critical role in IL-6 signaling. On the other hand, nuclear translocation of AR is induced by IL-6 treatment, whereas knockdown of endogenous IL-6 reduces AR and TLR4 expression in taxol-resistant ovarian cancer cells. These results indicate that TLR4 and IL-6 play a crucial role in AR gene regulation and function. We also identify interferon regulatory factor 1 (IRF1) as a downstream target of IL-6 signaling and as a regulator of AR expression. Moreover, analysis of clinical samples indicates that high IL-6 expression correlates with poor progression-free survival in ovarian cancer patients treated with taxol. Overall, our findings indicate that the TLR4/IL-6/IRF1 signaling axis represents a potential therapeutic target to overcome AR-based taxol resistance in ovarian cancer.


Subject(s)
Interferon Regulatory Factor-1/biosynthesis , Interleukin-6/biosynthesis , Ovarian Neoplasms/metabolism , Paclitaxel/administration & dosage , Receptors, Androgen/biosynthesis , Toll-Like Receptor 4/biosynthesis , Antineoplastic Agents, Phytogenic/administration & dosage , Biomarkers, Tumor/biosynthesis , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/physiology , Female , Gene Expression Regulation, Neoplastic , Humans , Interferon Regulatory Factor-1/genetics , Interleukin-6/genetics , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Receptors, Androgen/genetics , Toll-Like Receptor 4/genetics
4.
Biochem Pharmacol ; 177: 113965, 2020 07.
Article in English | MEDLINE | ID: mdl-32278794

ABSTRACT

Toll-like receptor 4 (TLR4) is often overexpressed in taxol-resistant cancer cells. Here we used whole-genome transcriptomic analysis to identify 787 upregulated genes in SKOV3 ovarian carcinoma cells that ectopically express TLR4. Using chromatin immunoprecipitation enrichment analysis, we observed that 27.8% of the TLR4-upregulated genes identified were androgen receptor (AR)-regulated genes. Accordingly, AR expression was induced in taxol-resistant SKOV3 cells overexpressing TLR4, whereas depletion of TLR4 by shRNA repressed AR expression. Activation of AR by androgens or silencing of AR using shRNA also regulated expression of AR-related genes. We found that expression of DCDC2, ANKRD18B, ALDH1A1, c14orf105, ITGBL1 and NEB was overexpressed in taxol-resistant cells, suggesting the involvement of these AR-related genes in taxol resistance. Pathway enrichment analysis confirmed that the expression of several upregulated genes enriched in steroid biosynthesis pathways was inducible by androgens, supporting the results of previous studies. We also observed that genistein inhibits AR activation, leading to suppression of AR-driven genes and reduced taxol resistance in ovarian cancer cells. Overall, we identified six TLR4- and AR-regulated genes involved in taxol resistance. Our results reveal that the TLR4/AR axis plays a critical role in taxol resistance and that genistein is a candidate compound to limit chemoresistance and improve cancer treatment in ovarian cancer.


Subject(s)
Drug Resistance, Neoplasm/drug effects , Genistein/pharmacology , Ovarian Neoplasms/genetics , Paclitaxel/pharmacology , Receptors, Androgen/genetics , Signal Transduction/genetics , Toll-Like Receptor 4/genetics , Anticarcinogenic Agents/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic/drug effects , Gene Ontology , Humans , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , RNA Interference , Receptors, Androgen/metabolism , Toll-Like Receptor 4/metabolism
5.
Mol Cancer Res ; 18(3): 375-389, 2020 03.
Article in English | MEDLINE | ID: mdl-31792079

ABSTRACT

Chronic hepatitis B virus (HBV) infection is a major risk factor for the development of hepatocellular carcinoma (HCC), which represents one of the most common cancers worldwide. Recent studies suggest that HBV's protein X (HBx) plays a crucial role in HCC development and progression. Earlier, genome-wide analysis identified that the receptor for hyaluronan-mediated motility (RHAMM) represents a putative oncogene and is overexpressed in many human cancers, including HCC. However, the mechanism underlying RHAMM upregulation and its role in tumorigenesis remain unclear. Here, we show that ectopic expression of HBx activates the PI3K/Akt/Oct-1 pathway and upregulates RHAMM expression in HCC cells. HBx overexpression leads to dissociation of C/EBPß from the RHAMM gene promoter, thereby inducing RHAMM upregulation. RHAMM knockdown attenuates HBx-induced cell migration and invasion in vitro. In mice, HBx promotes cancer cell colonization via RHAMM upregulation, resulting in enhanced metastasis. Analysis of gene expression datasets reveals that RHAMM mRNA level is upregulated in patients with HCC with poor prognosis. IMPLICATIONS: These results indicate that RHAMM expression is upregulated by HBx, a process that depends on the inhibition of C/EBPß activity and activation of the PI3K/Akt/Oct-1 pathway. These results have several implications for the treatment of HBV-positive HCC involving upregulation of RHAMM and cancer metastasis. VISUAL OVERVIEW: http://mcr.aacrjournals.org/content/molcanres/18/3/375/F1.large.jpg.


Subject(s)
Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Trans-Activators/genetics , Viral Regulatory and Accessory Proteins/genetics , Animals , Cell Movement , Humans , Male , Mice , Signal Transduction , Xenograft Model Antitumor Assays
6.
J Cell Physiol ; 234(6): 8760-8775, 2019 06.
Article in English | MEDLINE | ID: mdl-30317630

ABSTRACT

We report here that the androgen receptor (AR) and ABCB1 are upregulated in a model of acquired taxol resistance (txr) in ovarian carcinoma cells. AR silencing sensitizes txr cells to taxol threefold, whereas ectopic AR expression in AR-null HEK293 cells induces resistance to taxol by 1.7-fold. AR activation using the agonist dihydrotestosterone (DHT) or sublethal taxol treatment upregulates ABCB1 expression in both txr cells and AR-expressing HEK293 cells. In contrast, AR inactivation using the antagonist bicalutamide downregulates ABCB1 expression and enhances cytotoxicity to taxol. A functional ABCB1 promoter containing five predicted androgen-response elements (AREs) is cloned. Deletion assays reveal a taxol-responsive promoter segment which harbors ARE4. Notably, DHT- or taxol-activated AR potentiates binding of the AR to ARE4 as revealed by the chromatin immunoprecipitation. On the other hand, txr cells display an increase in chromatin remodeling. AR/H3K9ac and AR/H3K14ac complexes bind specifically to ARE4 in response to taxol. Furthermore, acetyltransferase protein levels (p300 and GCN5) are upregulated in txr cells. Silencing of p300 or GCN5 reduces chromatin modification and enhances cytotoxicity in both parental and txr SKOV3 cells. While the phosphatidylinositol 3-kinase (PI3K)/serine/threonine protein kinase (AKT) pathway is significantly activated by taxol, taxol-induced ABCB1 expression, histone posttranslational modifications, and p300 binding to ARE4 are suppressed following inhibition of the PI3K/AKT cellular pathway. These results demonstrate that the AKT/p300/AR axis can be activated to target ABCB1 gene expression in response to taxol, thus revealing a new treatment target to counter taxol resistance.


Subject(s)
Chromatin/metabolism , Genes, MDR/genetics , Ovarian Neoplasms/drug therapy , Paclitaxel/pharmacology , Receptors, Androgen/metabolism , Transcription, Genetic/drug effects , ATP Binding Cassette Transporter, Subfamily B , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Chromatin/genetics , Drug Resistance, Neoplasm , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Receptors, Androgen/genetics
7.
J Cell Physiol ; 233(3): 2489-2501, 2018 Mar.
Article in English | MEDLINE | ID: mdl-28771725

ABSTRACT

We report here that toll-like receptor 4 (TLR4) and ABCB1 are upregulated in SKOV3 ovarian carcinoma cells that acquired resistance to the anticancer drug taxol. Silencing of TLR4 using short-hairpin RNA sensitized taxol-resistant SKOV3 cells to taxol (4.6 fold), whereas ectopic expression of TLR4 in parental, taxol-sensitive SKOV3 cells or TLR4-null HEK293 cells induced taxol resistance (∼2 fold). A sub-lethal dose of taxol induced ABCB1 protein expression in taxol-resistant SKOV3 cells. Inactivation of TLR4 using chemical inhibitors (CLI-095 and AO-I) downregulated ABCB1 protein expression and enhanced the cytotoxic activity of taxol in taxol-resistant SKOV3 cells. While the sensitization effect of TLR4 inactivation was also detected in TOV21G ovarian cancer cells, which express moderate level of TLR4, ectopic expression of ABCB1 prevented the sensitization effect in these cells. Notably, the NFκB pathway was significantly activated by taxol, and inhibition of this pathway suppressed TLR4-regulated ABCB1 expression. Furthermore, taxol-induced NFκB signaling was reduced following TLR4 silencing in taxol-resistant SKOV3 cells. Consistent with these results, ectopic expression of TLR4 in taxol-sensitive SKOV3 cells enhanced ABCB1 expression and conferred resistance to taxol. The protective effect of exogenous TLR4 expression against taxol was reduced by treatment with NFκB inhibitor in these cells. These results demonstrate that taxol activates the TLR4-NFκB pathway which in turn induces ABCB1 gene expression. This cellular pathway thus represents a novel target to limit resistance to taxol in ovarian cancer cells.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Carcinoma/drug therapy , Drug Resistance, Neoplasm , NF-kappa B/metabolism , Ovarian Neoplasms/drug therapy , Paclitaxel/pharmacology , Signal Transduction/drug effects , Toll-Like Receptor 4/metabolism , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Binding Sites , Carcinoma/genetics , Carcinoma/metabolism , Carcinoma/pathology , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/drug effects , Female , Gene Expression Regulation, Neoplastic , Humans , Inhibitory Concentration 50 , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Promoter Regions, Genetic , RNA Interference , Sulfonamides/pharmacology , Toll-Like Receptor 4/antagonists & inhibitors , Toll-Like Receptor 4/genetics , Transfection , Up-Regulation
8.
Oncotarget ; 6(29): 27065-82, 2015 Sep 29.
Article in English | MEDLINE | ID: mdl-26318424

ABSTRACT

A systematic analysis of the genes involved in taxol resistance (txr) has never been performed. In the present study, we created txr ovarian carcinoma cell lines to identify the genes involved in chemoresistance. Transcriptome analysis revealed 1,194 overexpressed genes in txr cells. Among the upregulated genes, more than 12 cryptic transcription factors were identified using MetaCore analysis (including AR, C/EBPß, ERα, HNF4α, c-Jun/AP-1, c-Myc, and SP-1). Notably, individual silencing of these transcription factors (except HNF4`)sensitized txr cells to taxol. The androgen receptor (AR) and its target genes were selected for further analysis. Silencing AR using RNA interference produced a 3-fold sensitization to taxol in txr cells, a response similar to that produced by silencing abcb1. AR silencing also downregulated the expression of prominent txr gene candidates (including abcb1, abcb6, abcg2, bmp5, fat3, fgfr2, h1f0, srcrb4d, and tmprss15). In contrast, AR activation using the agonist DHT upregulated expression of the target genes. Individually silencing seven out of nine (78%) AR-regulated txr genes sensitized txr cells to taxol. Inhibition of AKT and JNK cellular kinases using chemical inhibitors caused a dramatic suppression of AR expression. These results indicate that the AR represents a critical driver of gene expression involved in txr.


Subject(s)
Carcinoma/metabolism , Drug Resistance, Neoplasm , Ovarian Neoplasms/metabolism , Paclitaxel/chemistry , Receptors, Androgen/metabolism , Antineoplastic Agents, Phytogenic/pharmacology , Cell Line, Tumor , Cell Survival , Female , Gene Expression Profiling , Gene Expression Regulation , Gene Expression Regulation, Neoplastic , Gene Silencing , Humans , Inhibitory Concentration 50 , Male , Oligonucleotide Array Sequence Analysis , Oligonucleotides/genetics , Phenotype , Polymerase Chain Reaction , Prostatic Neoplasms/metabolism , RNA Interference , Transcription Factors/metabolism , Transcriptome , Up-Regulation
9.
Oncotarget ; 5(23): 11939-56, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25460502

ABSTRACT

Taxol is a mitotoxin widely used to treat human cancers, including of the breast and ovary. However, taxol resistance (txr) limits treatment efficacy in human patients. To study chemoresistance in ovarian cancer, we established txr ovarian carcinoma cells derived from the SKOV3 cell lineage. The cells obtained were cross-resistant to other mitotoxins such as vincristine while they showed no resistance to the genotoxin cisplatin. Transcriptomic analysis identified 112 highly up-regulated genes in txr cells. Surprisingly, FK506-binding protein 5 (FKBP5) was transiently up-regulated 100-fold in txr cells but showed decreased expression in prolonged culture. Silencing of FKBP5 sensitized txr cells to taxol, whereas ectopic expression of FKBP5 increased resistance to the drug. Modulation of FKBP5 expression produced similar effects in response to vincristine but not to cisplatin. We observed that a panel of newly identified txr genes was trancriptionally regulated by FKBP5 and silencing of these genes sensitized cells to taxol. Notably, immunoprecipitation experiments revealed that FKBP5 forms a protein complex with the androgen receptor (AR), and this complex regulates the transcriptional activity of both proteins. Furthermore, we found that the Akt kinase pathway is regulated by FKBP5. These results indicate that the FKBP5/AR complex may affect cancer cell sensitivity to taxol by regulating expression of txr genes. Our findings suggest that mitotoxin-based treatment against ovarian cancer should be avoided when the Akt/FKBP5/AR axis is activated.


Subject(s)
Biomarkers, Tumor/analysis , Drug Resistance, Neoplasm/physiology , Ovarian Neoplasms/metabolism , Receptors, Androgen/metabolism , Tacrolimus Binding Proteins/metabolism , Animals , Antineoplastic Agents, Phytogenic/therapeutic use , Cell Line, Tumor , Female , Humans , Immunoblotting , Mice , Mice, SCID , Oligonucleotide Array Sequence Analysis , Ovarian Neoplasms/genetics , Paclitaxel/therapeutic use , Real-Time Polymerase Chain Reaction , Transcriptome , Transfection , Xenograft Model Antitumor Assays
10.
J Cell Physiol ; 229(3): 309-22, 2014 Mar.
Article in English | MEDLINE | ID: mdl-23939952

ABSTRACT

The oncogenic latent membrane protein 1 (LMP1) of Epstein-Barr virus (EBV) is involved in the pathogenesis of human nasopharyngeal carcinoma (NPC) and lymphoma. We and other authors have shown earlier that LMP1 induces apoptosis and inhibits xenograft tumor growth in mice, but the mechanism underlying these processes has not been investigated so far. In the present study, we show that knockdown of LMP1 renders the EBV-positive NPC cell line CG-1 resistant to various genotoxic drugs (cisplatin, etoposide, and adriamycin). LMP1 inhibits the expression of Cabin1, a Ca(2+) regulated protein shown earlier to inhibit calcineurin. Knockdown of calcineurin binding protein (Cabin1) with small hairpin RNA sensitizes CG-1 cells to genotoxic drugs. In contrast, LMP1 overexpression reduces Cabin1 level and renders both CG-1 cells and EBV-negative NPC cell lines sensitive to cisplatin. The c-Jun-N-terminal kinase (JNK) and ERK pathways are required for LMP1-induced suppression of Cabin1 at the transcriptional level. Chromatin immunoprecipitation assays further confirm that the JNK-activated transcription factor AP-1 mediates the LMP1-induced down-regulation of Cabin1 gene expression. LMP1 knockdown also increases the resistance of xenograph tumors to cisplatin in mice, therefore confirming the relevance of our findings in vivo. This study reveals the molecular mechanism underlying the pro-apoptotic activity of LMP1 during cisplatin-based NPC chemotherapy.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , Nasopharyngeal Neoplasms/drug therapy , Viral Matrix Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Apoptosis/drug effects , Binding Sites , Carcinoma , Cell Line, Tumor , Dose-Response Relationship, Drug , Down-Regulation , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Signaling System/drug effects , Mice , Mice, Nude , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/metabolism , Nasopharyngeal Neoplasms/pathology , Nasopharyngeal Neoplasms/virology , Promoter Regions, Genetic , RNA Interference , Time Factors , Transcription Factor AP-1/metabolism , Transfection , Tumor Burden , Viral Matrix Proteins/genetics , Xenograft Model Antitumor Assays
11.
J Cell Biochem ; 114(8): 1819-31, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23463670

ABSTRACT

Endometrial carcinoma (EC) is one of the main gynecologic malignancies affecting women, but effective treatments are currently lacking. In the present study, we investigated the effect of sorafenib, a general kinase inhibitor, on several EC cell lines (HEC1A, HEC1B, and RL95-2). Sorafenib induced cell death in EC cells with the following order of sensitivity: HEC1A > HEC1B > RL95-2. Sorafenib suppressed several anti-apoptotic proteins in HEC1A cells, including myeloid cell leukemia 1 (Mcl-1). Ectopic overexpression of Mcl-1 prevented the cell killing effect of sorafenib. Sorafenib suppressed Mcl-1 at the gene transactivation level by inactivating the ERK/Elk-1 pathway. Accordingly, the inhibitory effect of sorafenib on Mcl-1 expression decreased following knockdown of Elk-1 using short-hairpin RNA (shRNA). Elk-1 overexpression rescued both the inhibitory effect of sorafenib on Mcl-1 expression and the cell killing effect of sorafenib. Furthermore, sorafenib reduced the stability of the Mcl-1 protein by enhancing its ubiquitination and degradation by the proteasome via the AKT/GSK3ß and the ERK pathways. Similar results were detected in other EC cell lines. These results indicate that sorafenib induces apoptosis in EC cells by down-regulating the anti-apoptotic protein Mcl-1 via transcriptional inhibition and protein degradation. Our results thus support the notion that sorafenib may be used in endometrial cancer therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Endometrial Neoplasms/metabolism , Glycogen Synthase Kinase 3/metabolism , Niacinamide/analogs & derivatives , Phenylurea Compounds/pharmacology , Proteolysis/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Transcription, Genetic/drug effects , ets-Domain Protein Elk-1/metabolism , Apoptosis/genetics , Cell Line, Tumor , Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Female , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3 beta , Humans , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , Myeloid Cell Leukemia Sequence 1 Protein , Niacinamide/pharmacology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Sorafenib , Transcription, Genetic/genetics , ets-Domain Protein Elk-1/genetics
12.
J Neurosci Res ; 91(1): 51-61, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23086717

ABSTRACT

Growth arrest-specific 7 (Gas7) is preferentially expressed in the nervous system and plays an important role during neuritogenesis in mammals. However, the structure and function of Gas7 homologs have not been studied in nonmammalian vertebrates used as models. In this report, we identify a Gas7 gene in zebrafish that we termed zfGas7. The transcript of this gene was produced by canonical splicing, and its protein product contained a Fes/CIP4 homology and a coiled-coil domain. In early zebrafish embryos, RT-PCR analyses revealed that zfGas7 was initially expressed at 5.3 hr postfertilization (hpf), followed by an increase of expression at 10 hpf and further accumulation during somitogenesis at 48 hpf. Spatiotemporal analyses further showed that Gas7 mRNA was detected in the brain, somite, and posterior presomitic mesoderm regions during somitogenesis. At 36 hpf, zfGas7 mRNA was detected in the brain and somite but was later found only in neuronal clusters of the brain at 52 hpf. Gas7 knockdown with morpholino antisense oligonucleotides (Gas7MO) reduced the number of HuC-positive neurons in the trigeminal and statoacoustic ganglions and produced deformed phenotypes, such as flattening of the top of the head. Notably, the neuron reduction and deformed phenotypes observed in Gas7MO embryos were partially rescued by ectopic expression of Gas7. Because altered somitogenesis and pigmentation were also found in the morphants, the neuronal phenotypes observed likely are due to a general developmental delay of embryogenesis. These results indicate that Gas7 is expressed in neuronal cells but is not specifically required for neuronal development in vertebrates.


Subject(s)
Nerve Tissue Proteins/genetics , Nervous System/embryology , Neurogenesis/genetics , Zebrafish Proteins/genetics , Zebrafish/embryology , Animals , Base Sequence , Blotting, Western , Embryo, Nonmammalian , In Situ Hybridization , Molecular Sequence Data , Phylogeny , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Nucleic Acid , Zebrafish/genetics
13.
Biochem J ; 444(2): 303-14, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22397410

ABSTRACT

In the present study, we observed that the Golgi-SNARE (soluble N-ethylmaleimide-sensitive fusion protein-attachment protein receptor) GS28 forms a complex with p53 in HEK (human embryonic kidney)-293 cells. Given that p53 represents a tumour suppressor that affects the sensitivity of cancer cells to various chemotherapeutic drugs, we examined whether GS28 may influence the level of sensitivity to the DNA-damaging drug cisplatin. Indeed, knockdown of GS28 using short-hairpin RNA (shGS28) induced resistance to cisplatin in HEK-293 cells. On the other hand, overexpression of GS28 sensitized HEK-293 cells to cisplatin, whereas no sensitization effect was noted for the mitotic spindle-damaging drugs vincristine and taxol. Accordingly, we observed that knockdown of GS28 reduced the accumulation of p53 and its pro-apoptotic target Bax. Conversely, GS28 overexpression induced the accumulation of p53 and Bax as well as the pro-apoptotic phosphorylation of p53 on Ser(46). Further experiments showed that these cellular responses could be abrogated by the p53 inhibitor PFT-α (pifithrin-α), indicating that GS28 may affect the stability and activity of p53. The modulatory effects of GS28 on cisplatin sensitivity and p53 stability were absent in lung cancer H1299 cells which are p53-null. As expected, ectopic expression of p53 in H1299 cells restored the modulatory effects of GS28 on sensitivity to cisplatin. In addition, GS28 was found to form a complex with the p53 E3 ligase MDM2 (murine double minute 2) in H1299 cells. Furthermore, the ubiquitination of p53 was reduced by overexpression of GS28 in cells, confirming that GS28 enhances the stability of the p53 protein. Taken together, these results suggest that GS28 may potentiate cells to DNA-damage-induced apoptosis by inhibiting the ubiquitination and degradation of p53.


Subject(s)
Apoptosis/physiology , Cisplatin/pharmacology , Multiprotein Complexes/metabolism , Proteolysis , Proto-Oncogene Proteins c-mdm2/metabolism , Qb-SNARE Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Ubiquitination/physiology , Amino Acid Sequence , Apoptosis/drug effects , Cell Line, Tumor , DNA Damage/genetics , Drug Synergism , HEK293 Cells , Humans , Molecular Sequence Data , Proteolysis/drug effects , Proto-Oncogene Proteins c-mdm2/genetics , Qb-SNARE Proteins/genetics , Ubiquitination/drug effects
14.
Biochem Pharmacol ; 82(11): 1630-40, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-21903092

ABSTRACT

We found earlier that NAPA represents an anti-apoptotic protein that promotes resistance to cisplatin in cancer cells by inducing the degradation of the tumor suppressor p53. In the present study, we investigated the cellular mechanism underlying the degradation of p53 by NAPA. Knockdown of NAPA using short-hairpin RNA was shown to induce p53 accumulation and to sensitize HEK293 cells to cisplatin. On the other hand, this sensitization effect was not found in H1299 lung carcinoma cells which lack p53. Expression of exogenous p53 in H1299 cells was increased following knockdown of NAPA and these cells showed increased sensitivity to cisplatin-induced apoptosis. Notably, knockdown of NAPA induced the ubiquitination and degradation of the E3 ubiquitin ligase synoviolin and the accumulation of p53 in unstressed HEK293 cells. Conversely, NAPA overexpression decreased the ubiquitination and degradation of synoviolin, and reduced p53 protein level. Knockdown of NAPA disrupted the interaction between synoviolin and proteins that form the endoplasmic reticulum-associated degradation (ERAD) complex and in turn decreased the ability of this complex to ubiquitinate p53. In addition, knockdown of NAPA induced the activation of the MAPK kinases ERK, JNK and p38, but only inhibition of ERK reduced synoviolin ubiquitination and p53 accumulation. These results indicate that NAPA promotes resistance to cisplatin through synoviolin and the ERAD complex which together induce the degradation of p53 and thus prevent apoptosis. Based on these findings, we propose that the combination of cisplatin and knockdown of NAPA represents a novel and attractive strategy to eradicate p53-sensitive cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Mitogen-Activated Protein Kinase 1/physiology , Mitogen-Activated Protein Kinase 3/physiology , Soluble N-Ethylmaleimide-Sensitive Factor Attachment Proteins/genetics , Tumor Suppressor Protein p53/biosynthesis , Ubiquitin-Protein Ligases/metabolism , Apoptosis/drug effects , Cell Line, Tumor , Endoplasmic Reticulum-Associated Degradation , Gene Knockdown Techniques , Gene Silencing , HEK293 Cells , Humans , Signal Transduction , Soluble N-Ethylmaleimide-Sensitive Factor Attachment Proteins/biosynthesis , Ubiquitination
15.
J Cell Physiol ; 226(9): 2415-28, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21660965

ABSTRACT

CITED2 is a transcriptional modulator which has been implicated in human oncogenesis. In the present study, we examined whether CITED2 is also involved in the resistance of cancer cells to the chemotherapeutic drug cisplatin. We first observed that knockdown of CITED2 using short-hairpin RNA sensitized non-tumorigenic HEK293 cells to cisplatin. Sensitization to cisplatin following knockdown of CITED2 was also observed in cervical carcinoma HeLa cells and in cisplatin-resistant HeLa cells, thereby showing that acquired cisplatin resistance could be reversed by CITED2 knockdown. This sensitization response was dependent on the status of p53 since efficient sensitization was observed in p53-positive hepatocellular carcinoma (HCC) Sk-Hep-1 cells, whereas a negligible response was produced in the two p53-defective cell lines HCC Mahlavu and lung cancer H1299. In contrast, overexpression of CITED2 decreased sensitivity of HEK293 cells to cisplatin, while moderate resistance was produced in HeLa cells. Overexpression of CITED2 also decreased sensitivity to cisplatin in p53-defective H1299 cells when exogenous p53 expression was re-introduced. We observed that knockdown of CITED2-induced CBP/p300-mediated p53 acetylation (Lys373) in HEK293 cells, thereby leading to a decrease of p53 ubiquitination and subsequent accumulation of the p53 protein. Notably, the effects of CITED2 knockdown on p53 accumulation and the increase of p53's target Bax were more pronounced after treatment with cisplatin. Based on these results, we propose that a combination of cisplatin and CITED2 shRNA may represent an effective treatment against p53-sensitive cancer cells.


Subject(s)
Apoptosis/drug effects , Cisplatin/pharmacology , Gene Knockdown Techniques , RNA, Small Interfering/metabolism , Repressor Proteins/genetics , Trans-Activators/genetics , Tumor Suppressor Protein p53/metabolism , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Drug Screening Assays, Antitumor , HEK293 Cells , Humans , Models, Biological , Protein Stability/drug effects , Repressor Proteins/metabolism , Trans-Activators/metabolism
16.
J Biomed Sci ; 17: 27, 2010 Apr 17.
Article in English | MEDLINE | ID: mdl-20398405

ABSTRACT

BACKGROUND: We observed previously that cisplatin-resistant HeLa cells were cross-resistant to UV light due to accumulation of DDB2, a protein implicated in DNA repair. More recently, we found that cFLIP, which represents an anti-apoptotic protein whose level is induced by DDB2, was implicated in preventing apoptosis induced by death-receptor signaling. In the present study, we investigated whether DDB2 has a protective role against UV irradiation and whether cFLIP is also involved in this process. METHODS: We explored the role of DDB2 in mediating UV resistance in both human cells and Drosophila. To do so, DDB2 was overexpressed by using a full-length open reading frame cDNA. Conversely, DDB2 and cFLIP were suppressed by using antisense oligonucleotides. Cell survival was measured using a colony forming assay. Apoptosis was monitored by examination of nuclear morphology, as well as by flow cytometry and Western blot analyses. A transcription reporter assay was also used to assess transcription of cFLIP. RESULTS: We first observed that the cFLIP protein was upregulated in UV-resistant HeLa cells. In addition, the cFLIP protein could be induced by stable expression of DDB2 in these cells. Notably, the anti-apoptotic effect of DDB2 against UV irradiation was largely attenuated by knockdown of cFLIP with antisense oligonucleotides in HeLa cells. Moreover, overexpression of DDB2 did not protect against UV in VA13 and XP-A cell lines which both lack cFLIP. Interestingly, ectopic expression of human DDB2 in Drosophila dramatically inhibited UV-induced fly death compared to control GFP expression. On the other hand, expression of DDB2 failed to rescue a different type of apoptosis induced by the genes Reaper or eiger. CONCLUSION: Our results show that DDB2 protects against UV stress in a cFLIP-dependent manner. In addition, the protective role of DDB2 against UV irradiation was found to be conserved in divergent living organisms such as human and Drosophila. In addition, UV irradiation may activate a cFLIP-regulated apoptotic pathway in certain cells.


Subject(s)
DNA-Binding Proteins/genetics , Drosophila/metabolism , Ultraviolet Rays/adverse effects , Animals , Animals, Genetically Modified , Cell Line , DNA Damage , DNA-Binding Proteins/metabolism , Drosophila/radiation effects , HeLa Cells , Humans , Transfection , Up-Regulation
17.
Cancer Sci ; 98(8): 1206-14, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17517054

ABSTRACT

Our previous study identified a vincristine-selected multidrug resistance (MDR) cell line, HOB1/VCR, derived from a lymphoblastoma HOB1. The HOB1/VCR cells are resistant to typical MDR drugs and are cross-resistant to P-glycoprotein-independent drugs such as cisplatin (cis-diamminedichloroplatinum [II]). The mechanism of this atypical MDR phenotype is uncertain. The present study provides evidence regarding the contribution of reactive oxygen species (ROS) to the resistance of cells in response to treatments (vincristine, cisplatin and H2O2). Notably, the HOB1/VCR cells were cross-resistant to H2O2. High levels of ROS formed in both sensitive and HOB1/VCR cells by H2O2, and moderate levels of ROS were generated by treatment with cisplatin and vincristine. The ROS level in HOB1/VCR cells was lower than that in sensitive cells following treatments. The ROS level was reduced markedly by a non-toxic concentration of N-acetyl-L-cysteine, a ROS scavenger, in drug-treated cells, and was correlated with reduced cytotoxicity. Furthermore, concentrations of glutathione and glutathione peroxidase, but not superoxide dismutase and catalase, increased in HOB/VCR cells. The DL-buthionine-[S,R]-sulfoximine inhibited formation of glutathione and sensitized both cell types to treatments. Therefore, overexpression of an H2O2-reducing system, glutathione-glutathione peroxidase, has a role in resistance. Experimental results further demonstrate that ROS is likely a primary signal in the acquisition of the MDR phenotype and therefore a potential target when designing drugs for chemoresistance.


Subject(s)
Drug Resistance, Multiple , Drug Resistance, Neoplasm , Lymphoma/metabolism , Reactive Oxygen Species/metabolism , Vincristine/pharmacology , Apoptosis , Cell Line, Tumor , Cisplatin/pharmacology , Humans
18.
Chang Gung Med J ; 28(10): 673-82, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16382751

ABSTRACT

High mobility group box 1 (HMGB1), a mobile chromatin protein, passively leaks from necrotic cells and signals neighboring cells that tissue damage has occurred. Resting, non-activated inflammatory cells such as monocytes or macrophages contain HMGB1 in the nuclear compartment. When activated by lipopolysaccharide or inflammatory cytokines, they actively translocate the nuclear HMGB1 into the cytoplasm; HMGB1 is then exocytosed. At least one receptor for extracellular HMGB1 has been identified. HMGB1 acts as a mediator of systematic inflammation; it causes different cells to divide, migrate or elicit an immune response. Here, we give an abridged review of the cytokine activity of HMGB1, including its secretion mechanism, the putative signal transduction pathways, and its role in several inflammatory diseases. Finally, we cite a few examples in which therapeutic administration of HMGB1 antagonists rescued mice from lethal sepsis, arthritis and liver damage. The new findings of HMGB1 as a cytokine provide a better understanding of inflammatory diseases, establishing a clinically relevant therapeutic target that is significantly more efficient than other known cytokines.


Subject(s)
Cytokines/physiology , HMGB1 Protein/physiology , Active Transport, Cell Nucleus , Animals , Cisplatin/pharmacology , HMGB1 Protein/analysis , HMGB1 Protein/antagonists & inhibitors , Humans , Inflammation Mediators/physiology , Nicotine/pharmacology , Pyruvates/pharmacology
19.
Biochem J ; 376(Pt 1): 253-60, 2003 Nov 15.
Article in English | MEDLINE | ID: mdl-12911332

ABSTRACT

Cisplatin-selected cervix carcinoma HeLa cell lines induced less apoptosis, and weaker activation by cisplatin or Fas-activating antibody, of mitochondrial-associated caspase-9 and death receptor-mediated caspase-8 than did parental cells. Furthermore, less DISC (death-inducing signalling complex) was formed in cisplatin-selected cell lines than in parental cells. Ac-IETD-CHO (acetyl-Ile-Glu-Thr-Asp-aldehyde), which has a certain preference for inhibiting caspase-8, or Fas-antagonistic antibody, significantly inhibited cisplatin-induced apoptosis in both parental and cisplatin-selected HeLa cell lines. These results imply that cell-surface death signalling is inducible by cisplatin; that reduction of this pathway is associated with drug resistance, and that cisplatin-selected cells acquire cross-resistance to cell-surface death signalling. Sequential up-regulation of FLIP (FLICE-like inhibitory protein), but not Bcl-2, Bcl-x(L) or inhibitors of apoptosis protein (IAPs), was observed in resistant cells but not in parental cells. The inhibition of FLIP by FLIP antisense oligonucleotides promotes cisplatin and Fas-antibody-induced apoptosis. However, the modulation of apoptosis by FLIP antisense oligonucleotides in resistant cells is greater than that in parental cells. The presented data reveal that the up-regulation of FLIP may contribute to the suppression of apoptosis and thereby change cells that are resistant to cisplatin and Fas-mediated death signals. The results also show that cancer cells that have undergone long-term chemotherapy and become chemoresistant may change the FLIP level, becoming cross-resistant to death factors such as Fas.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis , Carrier Proteins/metabolism , Cisplatin/pharmacology , Intracellular Signaling Peptides and Proteins , fas Receptor/metabolism , Antibodies/pharmacology , Antineoplastic Agents/antagonists & inhibitors , CASP8 and FADD-Like Apoptosis Regulating Protein , Carrier Proteins/antagonists & inhibitors , Caspases/metabolism , Cisplatin/antagonists & inhibitors , Drug Resistance, Neoplasm , HeLa Cells , Humans , Signal Transduction , Up-Regulation , fas Receptor/immunology
20.
J Biomed Sci ; 9(4): 371-80, 2002.
Article in English | MEDLINE | ID: mdl-12145536

ABSTRACT

Recognition and incision of UV-DNA adducts play key roles in the efficacy of nucleotide excision repair. Damaged-DNA recognition activity has been identified from primate cells as a complex of DDB1 (127-kD) and DDB2 (48-kD) subunits. However, the function of damaged-DNA binding proteins (DDBs) in damaged-DNA recognition is not well understood. To assess the functional correlation between DDBs and UV-damaged-DNA recognition activity, we identified UV-damaged-DNA recognition activities in rodent cell lines. There is a cell type-dependent expression of DDB1 and DDB2. Rodent cells had less abundant DDBs and lower UV-damaged-DNA recognition activity than did human tumor cells. Interestingly, the profusion of DDBs is associated with UV-damaged-DNA recognition activity in these cell lines. We also discovered tissue-dependent expression of DDBs and its functional correlation with UV-damaged-DNA recognition activity. cDNA (3850 nucleotides) from rat ddb1 was isolated. It contained the complete length of the open reading frame that encodes an 1140-amino-acid polypeptide with a predicted molecular weight of 126.8 kD. The predicted protein size from the rat ddb1 gene resembles that from human DDB1 (127 kD). Rat DDB1 shares highly conserved sequencing (greater than 98% similarity) with those of mouse, human, and monkey. Rat and fruit fly DDB1 exhibit 62.23% identity and 57.66% homology. The evolutionary conservation of the DDB1 sequence suggests that DDB1 may play a pivotal role in mammals as well as in other eukaryotes. However, overexpression of DDB1 did not augment UV-damaged-DNA recognition activity in human HeLa, hamster V79, or rat PC12 cells. In contrast, restricting DDB2 expression by antisense ddb2 partially inhibited UV-damaged-DNA recognition activity in cells, whereas overexpressing DDB2 through a recombinant ddb2 adenovirus partly restored the recognition activity of these cells. These findings support the notion that DDB abundance is functionally correlated with UV-damaged-DNA recognition activity. These results also suggest that the profusion of DDB2, but not DDB1, may moderate UV-damaged-DNA recognition activity.


Subject(s)
DNA-Binding Proteins/metabolism , Amino Acid Sequence , Animals , Base Sequence , Cell Line , DNA/metabolism , DNA/radiation effects , DNA Damage , DNA Repair , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Humans , Molecular Sequence Data , Protein Subunits , Rats , Sequence Alignment , Ultraviolet Rays
SELECTION OF CITATIONS
SEARCH DETAIL
...