33 research outputs found

    FH535, a β-catenin Pathway Inhibitor, Represses Pancreatic Cancer Xenograft Growth and Angiogenesis

    Get PDF
    The WNT/β-catenin pathway plays an important role in pancreatic cancer carcinogenesis. We evaluated the correlation between aberrant β-catenin pathway activation and the prognosis pancreatic cancer, and the potential of applying the β-catenin pathway inhibitor FH535 to pancreatic cancer treatment. Meta-analysis and immunohistochemistry showed that abnormal β-catenin pathway activation was associated with unfavorable outcome. FH535 repressed pancreatic cancer xenograft growth in vivo. Gene Ontology (GO) analysis of microarray data indicated that target genes responding to FH535 participated in stemness maintenance. Real-time PCR and flow cytometry confirmed that FH535 downregulated CD24 and CD44, pancreatic cancer stem cell (CSC) markers, suggesting FH535 impairs pancreatic CSC stemness. GO analysis of β-catenin chromatin immunoprecipitation sequencing data identified angiogenesis-related gene regulation. Immunohistochemistry showed that higher microvessel density correlated with elevated nuclear β-catenin expression and unfavorable outcome. FH535 repressed the secretion of the proangiogenic cytokines vascular endothelial growth factor (VEGF), interleukin (IL)-6, IL-8, and tumor necrosis factor-α, and also inhibited angiogenesis in vitro and in vivo. Protein and mRNA microarrays revealed that FH535 downregulated the proangiogenic genes ANGPT2, VEGFR3, IFN-γ, PLAUR, THPO, TIMP1, and VEGF. FH535 not only represses pancreatic CSC stemness in vitro, but also remodels the tumor microenvironment by repressing angiogenesis, warranting further clinical investigation

    PP2A Inhibitors Arrest G2/M Transition Through JNK/Sp1-Dependent Down-Regulation of CDK1 and Autophagy-Dependent Up-Regulation of p21

    Get PDF
    Protein phosphatase 2A (PP2A) plays an important role in the control of the cell cycle. We previously reported that the PP2A inhibitors, cantharidin and okadaic acid (OA), efficiently repressed the growth of cancer cells. In the present study, we found that PP2A inhibitors arrested the cell cycle at the G2 phase through a mechanism that was dependent on the JNK pathway. Microarrays further showed that PP2A inhibitors induced expression changes in multiple genes that participate in cell cycle transition. To verify whether these expression changes were executed in a PP2A-dependent manner, we targeted the PP2A catalytic subunit (PP2Ac) using siRNA and evaluated gene expression with a microarray. After the cross comparison of these microarray data, we identified that CDK1 was potentially the same target when treated with either PP2A inhibitors or PP2Ac siRNA. In addition, we found that the down-regulation of CDK1 occurred in a JNK-dependent manner. Luciferase reporter gene assays demonstrated that repression of the transcription of CDK1 was executed through the JNK-dependent activation of the Sp1 transcription factor. By constructing deletion mutants of the CDK1 promoter and by using ChIP assays, we identified an element in the CDK1 promoter that responded to the JNK/Sp1 pathway after stimulation with PP2A inhibitors. Cantharidin and OA also up-regulated the expression of p21, an inhibitor of CDK1, via autophagy rather than PP2A/JNK pathway. Thus, this present study found that the PP2A/JNK/Sp1/CDK1 pathway and the autophagy/p21 pathway participated in G2/M cell cycle arrest triggered by PP2A inhibitors

    Stability analysis of gradient-based neural networks for optimization problems

    No full text
    The paper introduces a new approach to analyze the stability of neural network models without using any Lyapunov function. With the new approach, we investigate the stability properties of the general gradient-based neural network model for optimization problems. Our discussion includes both isolated equilibrium points and connected equilibrium sets which could be unbounded. For a general optimization problem, if the objective function is bounded below and its gradient is Lipschitz continuous, we prove that (a) any trajectory of the gradient-based neural network converges to an equilibrium point, and (b) the Lyapunov stability is equivalent to the asymptotical stability in the gradient-based neural networks. For a convex optimization problem, under the same assumptions, we show that any trajectory of gradient-based neural networks will converge to an asymptotically stable equilibrium point of the neural networks. For a general nonlinear objective function, we propose a refined gradient-based neural network, whose trajectory with any arbitrary initial point will converge to an equilibrium point, which satisfies the second order necessary optimality conditions for optimization problems. Promising simulation results of a refined gradient-based neural network on some problems are also reported

    Evidence of nigericin as a potential therapeutic candidate for cancers: A review

    No full text
    Emerging studies have shown that nigericin, an H+, K+ and Pb2+ ionophore, has exhibited a promising anti-cancer activity in various cancers. However, its anti-cancer mechanisms have not been fully elucidated. In this review, the recent progresses on the use of nigericin in human cancers have been summarized. By exchanging H+ and K+ across cell membranes, nigericin shows promising anti-cancer activities in in vitro and in vivo as a single agent or in combination with other anti-cancer drugs through decreasing intracellular pH (pHi). The underlying mechanisms of nigericin also include the inactivation of Wnt/β-catenin signals, blockade of Androgen Receptor (AR) signaling, and activation of Stress-Activated Protein Kinase/c-Jun N-terminal Kinase (SAPK/JNK) signaling pathways. In many cancers, nigericin is proved to specifically target putative Cancer Stem Cells (CSCs), and its synergistic effects on photodynamic therapy are also reported. Other mechanisms of nigericin including influencing the mitochondrial membrane potentials, inducing an increase in drug accumulation and autophagy, controlling insulin accumulation in nuclei, and increasing the cytotoxic activity of liposome-entrapped drugs, are also discussed. Notably, the potential adverse effects such as teratogenic effects, insulin resistance and eryptosis shall not be ignored. Taken together, these reports suggest that treatment of cancer cells with nigericin may offer a novel therapeutic strategy and future potential of translation to clinics

    VEZT, a Novel Putative Tumor Suppressor, Suppresses the Growth and Tumorigenicity of Gastric Cancer

    Get PDF
    <div><p>Vezatin (VEZT), an adherens junctions transmembrane protein, was identified as a putative tumor suppressor in our previous study. However, the role of VEZT in tumorigenesis remains elusive. We aimed to clarify its epigenetic regulation and biological functions in gastric cancer. In this study, we show that the expression level of VEZT is involved in lymphatic metastasis, depth of cancer invasion and TNM stage in 104 gastric cancer patients. Bisulfate sequencing polymerase chain reaction (BSP) methods showed that VEZT was hypermethylated in tissues and corresponding blood of gastric cancer patients compared with healthy controls. <i>Helicobacter pylori</i> (<i>H. pylori</i>) infection induces the methylation and silencing of VEZT in GES-1 cells. Restoring VEZT expression in MKN-45 and NCI-N87 gastric cancer cells inhibited growth, invasion and tumorigenesis <i>in vitro and in vivo</i>. Global microarray analysis was applied to analyze the molecular basis of the biological functions of VEZT after VEZT transfection combined with real-time PCR and chromatin immunoprecipitation assay. G protein-coupled receptor 56(GPR56), cell growth, cell division cycle 42(CDC42), migration/invasion and transcription factor 19(TCF19), cell cycle progression, were identified as direct VEZT target genes. TCF19, a novel target of VEZT, was functionally validated. Overexpression of TCF19 in MKN-45 cells increased cell cycle progress and growth ability. This study provides novel insight into the regulation of the VEZT gene, which could represent a potential target for therapeutic anti-cancer strategies.</p> </div

    <i>Helicobacter pylori</i> infection promotes the methylation and silencing of VEZT.

    No full text
    <p>(A) <i>H. pylori</i>-positive gastritis patients showed higher methylation levels in the VEZT promoter region when compared with <i>H. pylori</i>-negative gastritis patients. Each row of circles represents an integrated methylation ratio from three clones, and each circle represents a single CpG site. Open circle represents unmethylated cytosine, whereas filled circles or partially filled circles represent the methylated ratio of CpG sites. (B) After 24-h infection with <i>H. pylori</i>, the attachment of <i>H. pylori</i> was observed by transmission electron microscopy on the surface of GES-1 cells in the experimental cells relative to the control cells. (C) VEZT expression level in GES-1 cells was reduced after a 24-h <i>H. pylori</i> infection relative to negative control cells; however, <i>H. pylori</i> infection induced the IL-6, AKT, TNF-α, IL-8, ATF3 and IRX5 expression by western blot analysis. (D) The methylation of the VEZT promoter was detected by MSP after a 24-h <i>H. pylori</i> infection in GES-1 cells (marked as M), whereas methylation of the VEZT promoter in GES-1 cells that were not infected with <i>H. pylori</i> was not observed (marked as U). (E) Schematic summary of 34 CpG sites in the promoter region of the VEZT gene from -171 to -428 by BSP analysis. GES-1 cells showed a higher level of methylation after <i>H. pylori</i> infection relative to the control samples. The bar represents 5000 nm.</p

    Methylation analysis of normal gastric tissues, primary gastric cancer tissues and peripheral blood from gastric carcinoma patients and healthy controls.

    No full text
    <p>(A) Online bioinformatics software was used to analyze the promoter region and the methylation status of the VEZT gene. (B) Methylation status of 34 CpG sites of the VEZT promoter from 30 normal gastric tissues and 30 primary gastric cancer tissues. Primary tumor tissues showed higher methylation levels in the VEZT promoter region when compared with normal gastric tissues. (C) Methylation status of 34 CpG sites of the VEZT promoter from peripheral blood plasma DNA of 30 gastric carcinoma patients and 30 healthy controls. Peripheral blood from gastric carcinoma patients showed higher methylation levels in VEZT promoter region when compared with healthy controls. Each row of circles represents an integrated methylation ratio from three clones, and each circle represents a single CpG site. Open circle represents unmethylated cytosine, whereas filled circles or partially filled circles represent the methylated ratio of CpG sites.</p
    corecore