Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Eur J Pharmacol ; 906: 174214, 2021 Sep 05.
Article in English | MEDLINE | ID: mdl-34116044

ABSTRACT

The transcription factor ETS-1 (E26 transformation specific sequence 1) is the key regulator for malignant tumor cell proliferation and invasion by mediating the transcription of the invasion/migration related factors, e.g. MMPs (matrix metalloproteinases). This work aims to identify the novel small molecule inhibitors of ETS-1 using a small molecule compound library and to study the inhibitors' antitumor activity against hepatocellular carcinoma (HCC). The luciferase reporter is used to examine the inhibition and activation of ETS-1's transcription factor activity in HCC cells, including a highly invasive HCC cell line, MHCC97-H, and five lines of patient-derived cells. The inhibition of the proliferation of HCC cells is examined using the MTT assay, while the invasion of HCC cells is examined using the transwell assay. The anti-tumor activity of the selected compound on HCC cells is also examined in a subcutaneous tumor model or intrahepatic tumor model in nude mice. The results show that for the first time, four compounds, EI1~EI-4, can inhibit the transcription factor activation of ETS-1 and the proliferation or invasion of HCC cells. Among the four compounds, EI-4 has the best activation. The results from this paper contribute to expanding our understanding of ETS-1 and provide alternative, the safer and more effective, HCC molecular therapy strategies.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/drug therapy , Gene Expression Regulation, Neoplastic/drug effects , Liver Neoplasms/drug therapy , Proto-Oncogene Protein c-ets-1/antagonists & inhibitors , Animals , Antineoplastic Agents/therapeutic use , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Mice , Proto-Oncogene Protein c-ets-1/metabolism , Xenograft Model Antitumor Assays
2.
Cancer Res ; 81(8): 2071-2085, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33526511

ABSTRACT

The failure of once promising target-specific therapeutic strategies often arises from redundancies in gene expression pathways. Even with new melanoma treatments, many patients are not responsive or develop resistance, leading to disease progression in terms of growth and metastasis. We previously discovered that the transcription factors ETS1 and PAX3 drive melanoma growth and metastasis by promoting the expression of the MET receptor. Here, we find that there are multiple ETS family members expressed in melanoma and that these factors have redundant functions. The small molecule YK-4-279, initially developed to target the ETS gene-containing translocation product EWS-FLI1, significantly inhibited cellular growth, invasion, and ETS factor function in melanoma cell lines and a clinically relevant transgenic mouse model, BrafCA;Tyr-CreERT2;Ptenf/f. One of the antitumor effects of YK-4-279 in melanoma is achieved via interference of multiple ETS family members with PAX3 and the expression of the PAX3-ETS downstream gene MET. Expression of exogenous MET provided partial rescue of the effects of YK-4-279, further supporting that MET loss is a significant contributor to the antitumor effects of the drug. This is the first study identifying multiple overlapping functions of the ETS family promoting melanoma. In addition, targeting all factors, rather than individual members, demonstrated impactful deleterious consequences in melanoma progression. Given that multiple ETS factors are known to have oncogenic functions in other malignancies, these findings have a high therapeutic impact. SIGNIFICANCE: These findings identify YK-4-279 as a promising therapeutic agent against melanoma by targeting multiple ETS family members and blocking their ability to act as transcription factors.


Subject(s)
Indoles/pharmacology , Melanoma/drug therapy , Proto-Oncogene Proteins c-ets/antagonists & inhibitors , Skin Neoplasms/drug therapy , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/metabolism , Disease Progression , Humans , Melanoma/metabolism , Melanoma/pathology , Mice , Mice, Transgenic , Neoplasm Invasiveness , Oncogene Proteins, Fusion/antagonists & inhibitors , PAX3 Transcription Factor/antagonists & inhibitors , PAX3 Transcription Factor/metabolism , Proto-Oncogene Protein c-ets-1/antagonists & inhibitors , Proto-Oncogene Protein c-ets-1/metabolism , Proto-Oncogene Protein c-fli-1/antagonists & inhibitors , Proto-Oncogene Proteins c-ets/metabolism , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , RNA-Binding Protein EWS/antagonists & inhibitors , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism
3.
Blood Cancer Discov ; 1(2): 178-197, 2020 09.
Article in English | MEDLINE | ID: mdl-32924017

ABSTRACT

Notch activation is highly prevalent among cancers, in particular T-cell acute lymphoblastic leukemia (T-ALL). However, the use of pan-Notch inhibitors to treat cancers has been hampered by adverse effects, particularly intestinal toxicities. To circumvent this barrier in T-ALL, we aimed to inhibit ETS1, a developmentally important T-cell transcription factor previously shown to co-bind Notch response elements. Using complementary genetic approaches in mouse models, we show that ablation of Ets1 leads to strong Notch-mediated suppressive effects on T-cell development and leukemogenesis, but milder intestinal effects than pan-Notch inhibitors. Mechanistically, genome-wide chromatin profiling studies demonstrate that Ets1 inactivation impairs recruitment of multiple Notch-associated factors and Notch-dependent activation of transcriptional elements controlling major Notch-driven oncogenic effector pathways. These results uncover previously unrecognized hierarchical heterogeneity of Notch-controlled genes and points to Ets1-mediated enucleation of Notch-Rbpj transcriptional complexes as a target for developing specific anti-Notch therapies in T-ALL that circumvent the barriers of pan-Notch inhibition.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Leukemia, T-Cell , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma , Proto-Oncogene Protein c-ets-1 , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinogenesis/drug effects , Leukemia, T-Cell/drug therapy , Mice , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Proto-Oncogene Protein c-ets-1/antagonists & inhibitors , Receptor, Notch1/antagonists & inhibitors , Signal Transduction/physiology
4.
Acta Neuropathol Commun ; 7(1): 128, 2019 08 07.
Article in English | MEDLINE | ID: mdl-31391125

ABSTRACT

The BRAF gene and the TERT promoter are among the most frequently altered genomic loci in low-grade (LGG) and high-grade-glioma (HGG), respectively. The coexistence of BRAF and TERT promoter aberrations characterizes a subset of aggressive glioma. Therefore, we investigated interactions between those alterations in malignant glioma. We analyzed co-occurrence of BRAFV600E and TERT promoter mutations in our clinical data (n = 8) in addition to published datasets (n = 103) and established a BRAFV600E-positive glioma cell panel (n = 9) for in vitro analyses. We investigated altered gene expression, signaling events and TERT promoter activity upon BRAF- and E-twenty-six (ETS)-factor inhibition by qRT-PCR, chromatin immunoprecipitation (ChIP), Western blots and luciferase reporter assays. TERT promoter mutations were significantly enriched in BRAFV600E-mutated HGG as compared to BRAFV600E-mutated LGG. In vitro, BRAFV600E/TERT promoter double-mutant glioma cells showed exceptional sensitivity towards BRAF-targeting agents. Remarkably, BRAF-inhibition attenuated TERT expression and TERT promoter activity exclusively in double-mutant models, while TERT expression was undetectable in BRAFV600E-only cells. Various ETS-factors were broadly expressed, however, only ETS1 expression and phosphorylation were consistently downregulated following BRAF-inhibition. Knock-down experiments and ChIP corroborated the notion of a functional role for ETS1 and, accordingly, all double-mutant tumor cells were highly sensitive towards the ETS-factor inhibitor YK-4-279. In conclusion, our data suggest that concomitant BRAFV600E and TERT promoter mutations synergistically support cancer cell proliferation and immortalization. ETS1 links these two driver alterations functionally and may represent a promising therapeutic target in this aggressive glioma subgroup.


Subject(s)
Brain Neoplasms/genetics , Glioma/genetics , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Protein c-ets-1/genetics , Proto-Oncogene Proteins B-raf/genetics , Telomerase/genetics , Antineoplastic Agents/pharmacology , Brain Neoplasms/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Glioma/metabolism , HEK293 Cells , Humans , Indoles/pharmacology , Mutation/genetics , Promoter Regions, Genetic/genetics , Proto-Oncogene Protein c-ets-1/antagonists & inhibitors , Proto-Oncogene Protein c-ets-1/biosynthesis , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/biosynthesis , Telomerase/biosynthesis
5.
Cell Physiol Biochem ; 45(5): 1827-1839, 2018.
Article in English | MEDLINE | ID: mdl-29510376

ABSTRACT

BACKGROUND/AIMS: Recently, endothelial-like cells originating directly from tumor cells have been revealed. However, the mechanism remains unclear. ETS-1 (E26 transformation specific-1), a key transcription factor in the generation and maturation of ECs (endothelial cells), has been reported to be overexpressed in several cancers. Here, we reveal novel regulation of the endothelial-like differentiation of NSCLC (non-small cell lung cancer) cells by ETS-1. METHODS: We up-regulated the expression of ETS-1 in NSCLC cell lines by H2O2 or lentiviral vector. Endothelial phenotypes, such as vWF (von Willebrand factor) and VE-cadherin were examined by Western blot analysis and immunofluorescence assay. Tube formation assay and phagocytotic activity assay were performed to evaluate ECs' specific features on NSCLC cells. The effect of ETS-1 on metastasis was determined by wound healing assays, transwell assays and a xenograft tumor model. To explore the role of ETS-1 in the initiation and progression of NSCLC, we examined ETS-1 levels in NSCLC cancerous tissues and paired adjacent normal tissues by immunohistochemstry and analyzed the relationship between ETS-1 levels and clinicopathological parameters, as well as patient survival. Kaplan Meier plotter database was used to assess the prognostic value of ETS-1 in NSCLC. The association between ETS-1 levels and MVD (microvessel density) was analyzed to determine their role in angiogenesis. RESULTS: With ETS-1 up-regulation, the expression of vWF and VE-cadherin was increased in NSCLC cells. Additionally, cells adopted several ECs' specific features, including enhanced tube formation ability and uptake of Dil-ac-LDL (acetylated low-density lipoprotein) and lectin. ETS-1 up-regulation also promoted cell migration, invasion and adhesion. In addition, xenograft mice arising from ETS-1 over-expressing cells had more liver metastases. In the clinical specimens, ETS-1 expression was significantly higher in NSCLC cancerous tissues than adjacent nontumorous tissues and positively associated with tumor size, T stage, N stage and clinical stage. Patients with high levels of ETS-1 expression had significantly poorer OS (overall survival) and FP (first progression) than those with low expression. Furthermore, there was a positive correlation between ETS-1 level and MVD. CONCLUSION: Collectively, our data reveal that ETS-1 can induce the differentiation of tumor cells into endothelial-like cells and further promote metastatic dissemination in NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/pathology , Proto-Oncogene Protein c-ets-1/metabolism , A549 Cells , Adult , Aged , Aged, 80 and over , Animals , Antigens, CD/metabolism , Cadherins/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/mortality , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Female , Humans , Hydrogen Peroxide/pharmacology , Kaplan-Meier Estimate , Lectins/metabolism , Liver Neoplasms/pathology , Liver Neoplasms/secondary , Lung Neoplasms/metabolism , Lung Neoplasms/mortality , Male , Mice , Mice, Nude , Middle Aged , Neoplasm Staging , Proto-Oncogene Protein c-ets-1/antagonists & inhibitors , Proto-Oncogene Protein c-ets-1/genetics , RNA Interference , RNA, Small Interfering/metabolism , Transplantation, Heterologous , Up-Regulation/drug effects , von Willebrand Factor/metabolism
6.
Cancer Res ; 78(5): 1169-1183, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29311158

ABSTRACT

Long noncoding RNAs (lncRNA) play essential roles in tumor progression. However, the functions of lncRNAs in the tumorigenesis and aggressiveness of neuroblastoma still remain to be determined. Here, we report the identification of lncRNA pancEts-1 as a novel driver of neuroblastoma progression by using a public microarray dataset. LncRNA pancEts-1 promoted the growth, invasion, and metastasis of neuroblastoma cells in vitro and in vivo Mechanistically, pancEts-1 bound to hnRNPK to facilitate its physical interaction with ß-catenin, whereas hnRNPK stabilized the ß-catenin by inhibiting proteasome-mediated degradation, resulting in transcriptional alteration of target genes associated with neuroblastoma progression. Both pancEts-1 and hnRNPK were upregulated in clinical neuroblastoma tissues, and were associated with unfavorable outcome of patients. Overall, our results define an oncogenic role of pancEts-1 in neuroblastoma progression through hnRNPK-mediated ß-catenin stabilization, with potential implications for the clinical therapeutics of neuroblastoma.Significance: These findings reveal the oncogenic functions of a long noncoding RNA in neuroblastoma progression, offering a potential target for clinical therapeutics. Cancer Res; 78(5); 1169-83. ©2018 AACR.


Subject(s)
Biomarkers, Tumor/metabolism , Gene Expression Regulation, Neoplastic , Heterogeneous-Nuclear Ribonucleoprotein K/metabolism , Neuroblastoma/pathology , Proto-Oncogene Protein c-ets-1/antagonists & inhibitors , RNA, Long Noncoding/genetics , beta Catenin/chemistry , Animals , Apoptosis , Biomarkers, Tumor/genetics , Carcinogenesis , Cell Proliferation , Disease Progression , Female , Heterogeneous-Nuclear Ribonucleoprotein K/genetics , Humans , Male , Mice , Mice, Inbred BALB C , Neuroblastoma/genetics , Neuroblastoma/metabolism , Prognosis , Promoter Regions, Genetic , Protein Conformation , Proto-Oncogene Protein c-ets-1/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , beta Catenin/metabolism
7.
Biomed Pharmacother ; 96: 634-641, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29035829

ABSTRACT

New evidence suggests that microRNAs (miRNAs) play an important role in regulating the development and progression of prostate cancer. However, their specific functions and mechanisms remained to be further explored. MiR-129 has been reported in gastric cancers, colon cancer and lung cancer. In this study, we disclosed a new tumor suppresser function of miR-129 in prostate cancer. The purpose of our study is to clarify the effects of miR-129 in cellular processes correlated with cancer development and progression of prostate cancer cell by regulating ETS1. MiR-129 and ETS1 expression in prostate cancer tissues, tumor adjacent tissues and cells were tested by quantitative real-time PCR. We validated the target relationship between miR-129 and ETS1 by dual luciferase reporter gene system. MTT, colony formation, tumorigenesis assays, flow cytometry, wound healing and transwell assays were used to analyze cell viability, proliferation, migration, and invasiveness in vivo and in vitro. The level of ETS1 protein expression was detected by western blot. Here we demonstrate that miR-129 have a relatively reduced expression in prostate cancer cell lines and tissues. Morever, the miR-129 inhibits the expression of ETS1 by binding its 3'-UTR. The overexpression of miR-129 can inhibit PC-3 cell viability, proliferation, migration and invasion through targeting ETS1 by PI3K/AKT/mTOR signaling pathway. These findings suggested that miR-129 could directly suppress ETS1, which might be one of potential mechanisms in inhibiting cell processes including viability, proliferation, migration and invasiveness of prostate cancercells and it provides new clues for us to understand the carcinogenesis of prostate cancer. In addition, it may help to develop a treatment approach for ETS1-activated prostate cancer.


Subject(s)
MicroRNAs/biosynthesis , Phosphatidylinositol 3-Kinases/metabolism , Prostatic Neoplasms/metabolism , Proto-Oncogene Protein c-ets-1/metabolism , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Cell Line, Tumor , Cell Proliferation/physiology , Gene Targeting/methods , Humans , Male , MicroRNAs/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphoinositide-3 Kinase Inhibitors , Prostatic Neoplasms/genetics , Proto-Oncogene Protein c-ets-1/antagonists & inhibitors , Proto-Oncogene Protein c-ets-1/genetics , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/genetics
8.
Nucleic Acids Res ; 45(8): 4344-4358, 2017 05 05.
Article in English | MEDLINE | ID: mdl-28334937

ABSTRACT

Although studies of the differentiation from mouse embryonic stem (ES) cells to vascular endothelial cells (ECs) provide an excellent model for investigating the molecular mechanisms underlying vascular development, temporal dynamics of gene expression and chromatin modifications have not been well studied. Herein, using transcriptomic and epigenomic analyses based on H3K4me3 and H3K27me3 modifications at a genome-wide scale, we analysed the EC differentiation steps from ES cells and crucial epigenetic modifications unique to ECs. We determined that Gata2, Fli1, Sox7 and Sox18 are master regulators of EC that are induced following expression of the haemangioblast commitment pioneer factor, Etv2. These master regulator gene loci were repressed by H3K27me3 throughout the mesoderm period but rapidly transitioned to histone modification switching from H3K27me3 to H3K4me3 after treatment with vascular endothelial growth factor. SiRNA knockdown experiments indicated that these regulators are indispensable not only for proper EC differentiation but also for blocking the commitment to other closely aligned lineages. Collectively, our detailed epigenetic analysis may provide an advanced model for understanding temporal regulation of chromatin signatures and resulting gene expression profiles during EC commitment. These studies may inform the future development of methods to stimulate the vascular endothelium for regenerative medicine.


Subject(s)
Endothelial Cells/metabolism , Epigenesis, Genetic , GATA2 Transcription Factor/genetics , Histones/genetics , Mouse Embryonic Stem Cells/metabolism , Proto-Oncogene Protein c-ets-1/genetics , SOXF Transcription Factors/genetics , Animals , Cell Differentiation , Cell Lineage/genetics , Endothelial Cells/cytology , GATA2 Transcription Factor/antagonists & inhibitors , GATA2 Transcription Factor/metabolism , Histones/metabolism , Mice , Mouse Embryonic Stem Cells/cytology , Oligonucleotide Array Sequence Analysis , Primary Cell Culture , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/genetics , Protein Isoforms/metabolism , Proto-Oncogene Protein c-ets-1/antagonists & inhibitors , Proto-Oncogene Protein c-ets-1/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , SOXF Transcription Factors/antagonists & inhibitors , SOXF Transcription Factors/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
9.
Sci Rep ; 7: 42883, 2017 02 21.
Article in English | MEDLINE | ID: mdl-28220825

ABSTRACT

The Notch1 pathway plays important roles in modulating erythroid and megakaryocyte differentiation. To screen the Notch1-related genes that regulate differentiation fate of K562 and HEL cells, the expression of transient receptor potential ankyrin 1 (TRPA1) was induced by Notch1 receptor intracellular domain (N1IC), the activated form of Notch1 receptor. N1IC and v-ets erythroblastosis virus E26 oncogene homolog 1 (Ets-1) bound to TRPA1 promoter region to regulate transcription in K562 cells. Transactivation of TRPA1 promoter by N1IC depended on the methylation status of TRPA1 promoter. N1IC and Ets-1 suppressed the DNA methyltransferase 3B (DNMT3B) level in K562 cells. Inhibition of TRPA1 expression after Notch1 knockdown could be attenuated by nanaomycin A, an inhibitor of DNMT3B, in K562 and HEL cells. Functionally, hemin-induced erythroid differentiation could be suppressed by TRPA1, and the reduction of erythroid differentiation of both cells by N1IC and Ets-1 occurred via TRPA1. However, PMA-induced megakaryocyte differentiation could be enhanced by TRPA1, and the surface markers of megakaryocytes could be elevated by nanaomycin A. Megakaryocyte differentiation could be reduced by Notch1 or Ets-1 knockdown and relieved by TRPA1 overexpression. The results suggest that Notch1 and TRPA1 might be critical modulators that control the fate of erythroid and megakaryocyte differentiation.


Subject(s)
Cell Differentiation , Receptor, Notch1/metabolism , TRPA1 Cation Channel/metabolism , Cell Differentiation/drug effects , Cell Line, Tumor , DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , Erythroid Cells/cytology , Erythroid Cells/metabolism , Humans , K562 Cells , Megakaryocytes/cytology , Megakaryocytes/metabolism , Naphthoquinones/pharmacology , Promoter Regions, Genetic , Proto-Oncogene Protein c-ets-1/antagonists & inhibitors , Proto-Oncogene Protein c-ets-1/genetics , Proto-Oncogene Protein c-ets-1/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Receptor, Notch1/antagonists & inhibitors , Receptor, Notch1/genetics , TRPA1 Cation Channel/analysis , TRPA1 Cation Channel/genetics , Transcriptional Activation , DNA Methyltransferase 3B
10.
Mol Oncol ; 11(5): 491-516, 2017 05.
Article in English | MEDLINE | ID: mdl-28236660

ABSTRACT

Extravasation and metastatic progression are two main reasons for the high mortality rate associated with cancer. The metastatic potential of cancer cells depends on a plethora of metabolic challenges prevailing within the tumor microenvironment. To achieve higher rates of proliferation, cancer cells reprogram their metabolism, increasing glycolysis and biosynthetic activities. Just why this metabolic reprogramming predisposes cells towards increased oncogenesis remains elusive. The accumulation of myriad oncolipids in the tumor microenvironment has been shown to promote the invasiveness of cancer cells, with lysophosphatidic acid (LPA) being one such critical factor enriched in ovarian cancer patients. Cellular bioenergetic studies confirm that oxidative phosphorylation is suppressed and glycolysis is increased with long exposure to LPA in ovarian cancer cells compared with non-transformed epithelial cells. We sought to uncover the regulatory complexity underlying this oncolipid-induced metabolic perturbation. Gene regulatory networking using RNA-Seq analysis identified the oncogene ETS-1 as a critical mediator of LPA-induced metabolic alterations for the maintenance of invasive phenotype. Moreover, LPA receptor-2 specific PtdIns3K-AKT signaling induces ETS-1 and its target matrix metalloproteases. Abrogation of ETS-1 restores cellular bioenergetics towards increased oxidative phosphorylation and reduced glycolysis, and this effect was reversed by the presence of LPA. Furthermore, the bioenergetic status of LPA-treated ovarian cancer cells mimics hypoxia through induction of hypoxia-inducible factor-1α, which was found to transactivate ets-1. Studies in primary tumors generated in syngeneic mice corroborated the in vitro findings. Thus, our study highlights the phenotypic changes induced by the pro-metastatic factor ETS-1 in ovarian cancer cells. The relationship between enhanced invasiveness and metabolic plasticity further illustrates the critical role of metabolic adaptation of cancer cells as a driver of tumor progression. These findings reveal oncolipid-induced metabolic predisposition as a new mechanism of tumorigenesis and propose metabolic inhibitors as a potential approach for future management of aggressive ovarian cancer.


Subject(s)
Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Gene Expression Regulation, Neoplastic/drug effects , Lysophospholipids/pharmacology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Adenocarcinoma/genetics , Animals , Cell Line, Tumor , Female , Gene Expression Profiling , Gene Regulatory Networks/drug effects , Glycolysis/drug effects , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Matrix Metalloproteinase Inhibitors/pharmacology , Matrix Metalloproteinase Inhibitors/therapeutic use , Mice , Mice, Inbred BALB C , Mitochondria/metabolism , Neoplasm Invasiveness , Ovarian Neoplasms/genetics , Oxidative Phosphorylation/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Protein c-ets-1/antagonists & inhibitors , Proto-Oncogene Protein c-ets-1/genetics , Proto-Oncogene Protein c-ets-1/metabolism , Sequence Analysis, RNA
11.
Biochemistry ; 55(29): 4105-18, 2016 07 26.
Article in English | MEDLINE | ID: mdl-27362745

ABSTRACT

The affinity of the Ets-1 transcription factor for DNA is autoinhibited by an intrinsically disordered serine-rich region (SRR) and a helical inhibitory module (IM) appended to its winged helix-turn-helix ETS domain. Using NMR spectroscopy, we investigated how Ets-1 recognizes specific versus nonspecific DNA, with a focus on the roles of protein dynamics and autoinhibition in these processes. Upon binding either DNA, the two marginally stable N-terminal helices of the IM predominantly unfold, but still sample partially ordered conformations. Also, on the basis of amide chemical shift perturbation mapping, Ets-1 associates with both specific and nonspecific DNA through the same canonical ETS domain interface. These interactions are structurally independent of the SRR, and thus autoinhibition does not impart DNA-binding specificity. However, relative to the pronounced NMR spectroscopic changes in Ets-1 resulting from specific DNA binding, the spectra of the nonspecific DNA complexes showed conformational exchange broadening and lacked several diagnostic amide and indole signals attributable to hydrogen bonding interactions seen in reported X-ray crystallographic structures of this transcription factor with its cognate DNA sequences. Such differences are highlighted by the chemical shift and relaxation properties of several interfacial lysine and arginine side chains. Collectively, these data support a general model in which Ets-1 interacts with nonspecific DNA via dynamic electrostatic interactions, whereas hydrogen bonding drives the formation of well-ordered complexes with specific DNA.


Subject(s)
Proto-Oncogene Protein c-ets-1/chemistry , Proto-Oncogene Protein c-ets-1/metabolism , Animals , Arginine/chemistry , Base Sequence , DNA/chemistry , DNA/metabolism , Lysine/chemistry , Mice , Models, Molecular , Molecular Dynamics Simulation , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Protein Conformation , Protein Structure, Secondary , Proto-Oncogene Protein c-ets-1/antagonists & inhibitors , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Static Electricity
12.
Mol Med Rep ; 13(4): 3559-66, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26934912

ABSTRACT

Oncostatin M (OSM) contributes to cartilage degeneration in osteoarthritis (OA) and was demonstrated to be expressed in OA osteoblasts. Endothelin­1 (ET­1) is implicated in the degradation of OA articular cartilage, and osteoblast proliferation and bone development. In the present study, the effects of ET­1 on OSM expression in human OA osteoblasts were investigated, to the best of our knowledge, for the first time. Primary human OA osteoblasts were treated with ET­1 (1, 5, 10, 20 and 30 nM) for 0.5, 1, 2, 3 and 4 h with or without the selective ETA receptor (ETAR) antagonist, BQ123, ETB receptor antagonist, BQ788 or the phosphatidylinositol 3­kinase (PI3K) inhibitor, BKM120. ET­1 treatment induced OSM mRNA expression, and the intracellular and secreted protein levels of OA osteoblasts in a dose­dependent manner. This effect was suppressed by BQ123 and BKM120, but not BQ788 administration. In combination with electrophoretic mobility shift assays, deletional and mutational analyses on the activity of a human OSM promoter/luciferase reporter demonstrated that ET­1 induced OSM expression in OA osteoblasts by trans­activating the OSM gene promoter through specific binding of Ets­1 to an Ets­1 binding site in the OSM promoter in an ETAR­ and PI3K­dependent manner. Furthermore, ET­1 treatment increased the expression of Ets­1 in a dose­dependent manner, however the knockdown of Ets­1 suppressed the ET1­induced expression of OSM in OA osteoblasts. In conclusion, the present study demonstrated that ET­1 induces the expression of OSM in OA osteoblasts by trans­activating the OSM gene promoter primarily through increasing the expression level of Ets­1 in an ETAR­ and PI3K­dependent manner. The current study suggested novel insights into the mechanistic role of ET­1 in the pathophysiology of OA.


Subject(s)
Endothelin-1/pharmacology , Oncostatin M/metabolism , Osteoblasts/metabolism , Proto-Oncogene Protein c-ets-1/metabolism , Transcriptional Activation/drug effects , Aminopyridines/pharmacology , Base Sequence , Binding Sites , Cells, Cultured , Genes, Reporter , Humans , Molecular Sequence Data , Morpholines/pharmacology , Oligopeptides/pharmacology , Oncostatin M/genetics , Osteoarthritis/metabolism , Osteoarthritis/pathology , Osteoblasts/cytology , Peptides, Cyclic/pharmacology , Piperidines/pharmacology , Promoter Regions, Genetic , Protein Binding , Proto-Oncogene Protein c-ets-1/antagonists & inhibitors , Proto-Oncogene Protein c-ets-1/genetics , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism
13.
PLoS One ; 10(12): e0144877, 2015.
Article in English | MEDLINE | ID: mdl-26658785

ABSTRACT

BACKGROUND: Luteolin (LUT), a kind of flavonoid which is extracted from a variety of diets, has been reported to convey protective effects of various diseases. Recent researches have suggested that LUT can carry out cardioprotective effects during ischemia/reperfusion (I/R). However, there have no reports on whether LUT can exert protective effects against myocardial I/R injury through the actions of specific microRNAs (miRs). The purpose of this study was to determine which miRs and target genes LUT exerted such function through. METHODS: Expression of various miRs in perfused rat hearts was detected using a gene chip. Target genes were predicted with TargetScan, MiRDB and MiRanda. Anoxia/reoxygenation was used to simulate I/R. Cells were transfected by miR-208b-3p mimic, inhibitor and small interfering RNA of Ets1 (avian erythroblastosis virus E26 (v ets) oncogene homolog 1). MiR-208b-3p and Ets1 mRNA were quantified by real-time quantitative polymerase chain reaction. The percentage of apoptotic cells was detected by annexin V-fluorescein isothiocyanate/propidium iodide dyeing and flow cytometry. The protein expression levels of cleaved caspase-3, Bcl-2, Bax, and Ets1 were examined by western blot analysis. A luciferase reporter assay was used to verify the combination between miR-208b-3p and the 3'-untranslated region of Ets1. RESULTS: LUT pretreatment reduced miR-208b-3p expression in myocardial tissue, as compared to the I/R group. And LUT decreased miR-208b-3p expression and apoptosis caused by I/R. However, overexpression of miR-208b-3p further aggravated the changes caused by I/R and blocked all the effects of LUT. Knockdown of miR-208b-3p expression also attenuated apoptosis, while knockdown of Ets1 promoted apoptosis. Further, the luciferase reporter assay showed that miR-208b-3p could inhibit Ets1 expression. CONCLUSION: LUT pretreatment conveys anti-apoptotic effects after myocardial I/R injury by decreasing miR-208b-3p and increasing Ets1 expression levels.


Subject(s)
Cardiotonic Agents/pharmacology , Luteolin/pharmacology , MicroRNAs/genetics , Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/drug effects , Proto-Oncogene Protein c-ets-1/genetics , 3' Untranslated Regions , Animals , Apoptosis , Caspase 3/genetics , Caspase 3/metabolism , Cell Line , Gene Expression Regulation , Genes, Reporter , Luciferases/genetics , Luciferases/metabolism , Male , MicroRNAs/antagonists & inhibitors , MicroRNAs/metabolism , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Oligonucleotides/genetics , Oligonucleotides/metabolism , Organ Culture Techniques , Proto-Oncogene Protein c-ets-1/antagonists & inhibitors , Proto-Oncogene Protein c-ets-1/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction
14.
PLoS One ; 10(7): e0133074, 2015.
Article in English | MEDLINE | ID: mdl-26177288

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the common malignancies, which is highly metastatic and the third common cause of cancer deaths in the world. The invasion and metastasis of cancer cells is a multistep and complex process which is mainly initiated by extracellular matrix (ECM) degradation. Aberrant expression of microRNA has been investigated in HCC and shown to play essential roles during HCC progression. In the present study, we found that microRNA-324-5p (miR-324-5p) was downregulated in both HCC cell lines and tissues. Ectopic miR-324-5p led to the reduction of HCC cells invasive and metastatic capacity, whereas inhibition of miR-324-5p promoted the invasion of HCC cells. Matrix metalloproteinase 2 (MMP2) and MMP9, the major regulators of ECM degradation, were found to be downregulated by ectopic miR-324-5p, while upregulated by miR-324-5p inhibitor. E26 transformation-specific 1 (ETS1) and Specificity protein 1 (SP1), both of which could modulate MMP2 and MMP9 expression and activity, were presented as the direct targets of and downregulated by miR-324-5p. Downregulation of ETS1 and SP1 mediated the inhibitory function of miR-324-5p on HCC migration and invasion. Our study demonstrates that miR-324-5p suppresses hepatocellular carcinoma cell invasion and might provide new clues to invasive HCC therapy.


Subject(s)
Carcinoma, Hepatocellular/genetics , Extracellular Matrix/metabolism , Gene Expression Regulation, Neoplastic , Liver Neoplasms/genetics , MicroRNAs/genetics , Proto-Oncogene Protein c-ets-1/genetics , Sp1 Transcription Factor/genetics , Base Sequence , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Transformed , Cell Line, Tumor , Cell Movement , Epithelial Cells/cytology , Epithelial Cells/metabolism , Extracellular Matrix/chemistry , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Liver/metabolism , Liver/pathology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , MicroRNAs/metabolism , Molecular Sequence Data , Neoplasm Invasiveness , Proto-Oncogene Protein c-ets-1/antagonists & inhibitors , Proto-Oncogene Protein c-ets-1/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Sp1 Transcription Factor/antagonists & inhibitors , Sp1 Transcription Factor/metabolism , Transcription, Genetic
15.
Mol Cell Biochem ; 405(1-2): 115-24, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25893733

ABSTRACT

Endothelial cells (ECs) apoptosis induced by oxidized low-density lipoprotein (ox-LDL) is thought to play an essential role in atherosclerosis. MicroRNAs (miRNAs) are a class of short non-coding RNAs, acting as posttranscriptional regulators of protein-coding genes involved in vascular cell biology. MiRNA-221 and miRNA-222 (miR-221/222) are known to be involved in the regulation of endothelial inflammation and angiogenesis. However, the function of miR-221/222 in ox-LDL-induced ECs apoptosis and atherosclerosis is still unknown. Here, we showed that miR-221/222 expression was markedly down-regulated in ox-LDL-induced apoptotic human umbilical cord vein endothelial cells. MiR-221/222 inhibition enhanced apoptosis in ECs, whereas over-expression of miR-221/222 could partly alleviate apoptotic cell death mediated by ox-LDL through suppression of Ets-1 and its downstream target p21. These findings suggest that manipulation of the miR-221/222-Ets-1-p21 pathway may offer a novel strategy for treatment of endothelial apoptosis and atherosclerosis.


Subject(s)
Apoptosis/genetics , Cyclin-Dependent Kinase Inhibitor p21/antagonists & inhibitors , Endothelial Cells/pathology , Lipoproteins, LDL/adverse effects , MicroRNAs/genetics , Proto-Oncogene Protein c-ets-1/antagonists & inhibitors , Atherosclerosis/genetics , Atherosclerosis/pathology , Cell Line , Cyclin-Dependent Kinase Inhibitor p21/genetics , Down-Regulation/genetics , Endothelial Cells/metabolism , Gene Expression Regulation/genetics , Human Umbilical Vein Endothelial Cells , Humans , Proto-Oncogene Protein c-ets-1/genetics
16.
Sci Rep ; 5: 9170, 2015 Mar 16.
Article in English | MEDLINE | ID: mdl-25773070

ABSTRACT

Bone metastasis of breast cancer cells is a major concern, as it causes increased morbidity and mortality in patients. Bone tissue-derived CXCL12 preferentially recruits breast cancer cells expressing CXCR4 to bone metastatic sites. Thus, understanding how CXCR4 expression is regulated in breast cancer cells could suggest approaches to decrease bone metastasis of breast tumor cells. Here, we show that tumor cell-derived angiopoietin-like protein 2 (ANGPTL2) increases responsiveness of breast cancer cells to CXCL12 by promoting up-regulation of CXCR4 in those cells. In addition, we used a xenograft mouse model established by intracardiac injection of tumor cells to show that ANGPTL2 knockdown in breast cancer cells attenuates tumor cell responsiveness to CXCL12 by decreasing CXCR4 expression in those cells, thereby decreasing bone metastasis. Finally, we found that ANGPTL2 and CXCR4 expression levels within primary tumor tissues from breast cancer patients are positively correlated. We conclude that tumor cell-derived ANGPTL2 may increase bone metastasis by enhancing breast tumor cell responsiveness to CXCL12 signaling through up-regulation of tumor cell CXCR4 expression. These findings may suggest novel therapeutic approaches to treat metastatic breast cancer.


Subject(s)
Angiopoietins/metabolism , Bone Neoplasms/pathology , Breast Neoplasms/pathology , Receptors, CXCR4/metabolism , Angiopoietin-Like Protein 2 , Angiopoietin-like Proteins , Angiopoietins/antagonists & inhibitors , Angiopoietins/genetics , Animals , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Bone Neoplasms/secondary , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Culture Techniques , Cell Line, Tumor , Cell Movement , Chemokine CXCL12/metabolism , Female , Humans , Matrix Metalloproteinase 13/metabolism , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , MicroRNAs/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Proto-Oncogene Protein c-ets-1/antagonists & inhibitors , Proto-Oncogene Protein c-ets-1/genetics , Proto-Oncogene Protein c-ets-1/metabolism , Signal Transduction/genetics , Transplantation, Heterologous
17.
PLoS One ; 9(6): e99049, 2014.
Article in English | MEDLINE | ID: mdl-24897113

ABSTRACT

Long-term activation of extracellular-regulated kinase (ERK1/2) pathway has been shown to cause glucotoxicity and inhibit insulin gene expression in ß-cells. Transcription factor Ets1 is activated by ERK1/2-mediated phosphorylation at the Thr38 residue. We hypothesize that Ets1 plays an important role in mediating ERK1/2 induced glucotoxicity in ß-cells. We determined the role of Ets1 in Min6 cells and isolated mouse islets using overexpression and siRNA mediated knockdown of Ets1. The results show that Ets1 was localized in insulin-staining positive cells but not in glucagon-staining positive cells. Overexpression of Ets1 reduced glucose-stimulated insulin secretion in primary mouse islets. Overexpression of Ets1 in Min6 ß-cells and mouse islets increased expression of thioredoxin-interacting protein (TXNIP). Conversely, knockdown of Ets1 by siRNA reduced expression of TXNIP in Min6 cells. Ets1 was associated with the txnip promoter in min6 cells and transfection of 293 cells with Ets1 and p300 synergistically increased txnip promoter reporter activity. Moreover, overexpression of Ets1 inhibited Min6 cell proliferation. Our results suggest that Ets1, by promoting TXNIP expression, negatively regulates ß-cell function. Thus, over-activation of Ets1 may contribute to diet-induced ß-cell dysfunction.


Subject(s)
Carrier Proteins/genetics , Gene Expression Regulation , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Proto-Oncogene Protein c-ets-1/metabolism , Thioredoxins/genetics , Animals , Blotting, Western , Carrier Proteins/metabolism , Cells, Cultured , Chromatin Immunoprecipitation , Fluorescent Antibody Technique , Glucose/metabolism , Insulin Secretion , Insulin-Secreting Cells/cytology , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic/genetics , Proto-Oncogene Protein c-ets-1/antagonists & inhibitors , Proto-Oncogene Protein c-ets-1/genetics , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Thioredoxins/metabolism
18.
Int J Cancer ; 135(10): 2317-28, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-24706481

ABSTRACT

Ets-1 overexpression in human breast cancers is associated with invasiveness and poor prognosis. By overexpressing Ets-1 or a dominant negative mutant in MMT breast cancer cells, we previously highlighted the key role of Ets-1 in coordinating multiple invasive features of these cells. Interestingly, we also noticed that Ets-1 decreased the density of breast cancer cells cultured in three-dimensional extracellular matrix gels. The 3D context was instrumental to this phenomenon, as such downregulation was not observed in cells grown on two-dimensional plastic or matrix-coated dishes. Ets-1 overexpression was deleterious to anchorage-independent growth of MMT cells in soft agar, a standard model for in vitro tumorigenicity. The relevance of this mechanism was confirmed in vivo, during primary tumor growth and in a metastatic assay of lung colonization. In these models, Ets-1 was associated with epithelial-to-mesenchymal transition features and modulated the ratio of Ki67-positive cells, while hardly affecting in vivo apoptotic cell death. Finally, siRNA-mediated knockdown of Ets-1 in human breast cancer cell lines also decreased colony growth, both in anchorage-independent assays and 3D extracellular matrix cultures. These in vitro and in vivo observations shed light on an unsuspected facet of Ets-1 in breast tumorigenesis. They show that while promoting malignancy through the acquisition of invasive features, Ets-1 also attenuates breast tumor cell growth and could therefore repress the growth of primary tumors and metastases. This work also demonstrates that 3D models may reveal mechanisms of tumor biology that are cryptic in standard 2D models.


Subject(s)
Breast Neoplasms/pathology , Cell Movement , Cell Proliferation , Lung Neoplasms/secondary , Proto-Oncogene Protein c-ets-1/metabolism , Animals , Apoptosis , Blotting, Western , Breast Neoplasms/metabolism , Cell Culture Techniques , Cell Cycle , Female , Humans , Lung Neoplasms/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Proto-Oncogene Protein c-ets-1/antagonists & inhibitors , Proto-Oncogene Protein c-ets-1/genetics , RNA, Small Interfering/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
19.
Physiol Genomics ; 45(12): 493-8, 2013 Jun 17.
Article in English | MEDLINE | ID: mdl-23632416

ABSTRACT

It is well recognized that there is sex-dimorphic expression of mRNA and protein in the heart; however, the underlying mechanism is poorly understood. Endothelial nitric oxide synthase (eNOS) is an important regulator of cardiac function, and the expression levels of eNOS differ between male and female hearts. The aim of this study was to examine whether expression of specific microRNA (miRNA, miR) in males and females contributes to changes in the expression of eNOS. miRNA was extracted from the myocardium of male and female C57BL/6 mice and subjected to an Affymetrix miRNA array. Decreased expression of miR-222 was discovered in females and confirmed by qRT-PCR from whole heart or isolated cardiomyocytes. The transcription factor V-ets erythroblastosis virus E26 oncogene homolog-1 (ets-1) was identified as a potential target of miR-222 using TargetScan, and fivefold increased ets-1 protein expression in females was confirmed by Western blot. Targeting of ets-1 by miR-222 was determined in HEK293 cells overexpressing luciferase under regulation of either the ets-1 3'-UTR, a null 3'-UTR control, or a scrambled ets-1 3'-UTR and treated with a small molecule miR-222 mimic or inhibitor. Additionally qRT-PCR confirmed that mRNA levels of the ets-1 transcriptional target, eNOS, were 25% higher in females. Compared with untreated myocyte controls, 50% inhibition of eNOS expression was achieved by treatment with a miR-222 mimic, compared with a 25% increase due to miR-222 inhibitor. Our findings indicate that sex-dependent miR-222 regulation alters the expression of the cardiac regulatory protein eNOS.


Subject(s)
MicroRNAs/metabolism , Myocardium/enzymology , Nitric Oxide Synthase Type III/metabolism , Proto-Oncogene Protein c-ets-1/metabolism , Sex Characteristics , Animals , Female , Gene Expression Profiling , Gene Expression Regulation , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/enzymology , Nitric Oxide Synthase Type III/genetics , Proto-Oncogene Protein c-ets-1/antagonists & inhibitors , Proto-Oncogene Protein c-ets-1/genetics
20.
Cell Signal ; 25(1): 308-18, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23079083

ABSTRACT

Generally, both endoplasmic reticulum (ER) stress and mitochondrial dysregulation are a potential therapeutic target of anticancer agents including bortezomib. The treatment of melanoma cells with bortezomib was found to induce apoptosis together with the upregulation of Noxa, Mcl-1, and HSP70 proteins, and the cleavage of LC3 and autophagic formation. Also, bortezomib induced ER-stress as evidenced by the increase of intracellular Ca(2+) release. In addition, bortezomib enhanced the phosphorylation of inositol-requiring transmembrane kinase and endonuclease 1α (IRE1α), apoptosis signal-regulating kinase 1 (ASK1), c-jun-N-terminal kinase (JNK) and p38, and the activation of the transcription factors AP-1, ATF-2, Ets-1, and HSF1. Bortezomib-induced mitochondrial dysregulation was associated with the accumulation of reactive oxygen species (ROS), the release of both apoptosis inducing factor (AIF) and cytochrome c, the activation of caspase-9 and caspase-3, and cleavage of Poly (ADP-ribose) polymerase (PARP). The pretreatment of melanoma cells with the inhibitor of caspase-3 (Ac-DEVD-CHO) was found to block bortezomib-induced apoptosis that subsequently led to the increase of autophagic formation. In contrast, the inhibition of ASK1 abrogated bortezomib-induced autophagic formation and increased apoptosis induction. Furthermore, the inhibition of JNK, of HSP70 also increased apoptosis induction without influence of bortezomib-induced autophagic formation. Based on the inhibitory experiments, the treatment with bortezomib triggers the activation of both ER-stress-associated pathways, namely IRE1α-ASK1-p38-ATF-2/ets-1-Mcl-1, and IRE1α-ASK1-JNK-AP-1/HSF1-HSP70 as well as mitochondrial dysregulation-associated pathways, namely ROS-ASK1-JNK-AP-1/HSF1-HS70, and AIF-caspase-3-PARP and Cyt.c, and caspase-9-caspase-3-PARP. Taken together, our data demonstrates for the first time the molecular mechanisms, whereby bortezomib triggers both apoptosis and autophagic formation in melanoma cells.


Subject(s)
Antineoplastic Agents/toxicity , Apoptosis/drug effects , Autophagy/drug effects , Boronic Acids/toxicity , Proteasome Inhibitors/toxicity , Pyrazines/toxicity , Bortezomib , Calcium/metabolism , Caspase 3/chemistry , Caspase 3/metabolism , Cell Line, Tumor , Endoplasmic Reticulum Stress/drug effects , HSP70 Heat-Shock Proteins/antagonists & inhibitors , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Humans , MAP Kinase Kinase Kinase 5/metabolism , Melanoma/metabolism , Melanoma/pathology , Membrane Potential, Mitochondrial/drug effects , Microtubule-Associated Proteins/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Myeloid Cell Leukemia Sequence 1 Protein , Proto-Oncogene Protein c-ets-1/antagonists & inhibitors , Proto-Oncogene Protein c-ets-1/genetics , Proto-Oncogene Protein c-ets-1/metabolism , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...