34 research outputs found

    Schematic representation represents the anti-metastatic potential of ADC.

    No full text
    <p>TGF-β ligand binding with TGF-β receptor II (TGF-βRII) recruit TGF-βRI into a tetrameric receptor complex resulting in transphosphorylation and activation of TGF-βRI, which then phosphorylates Smad2 or Smad3. Phosphorylated Smad2/3 associate with Smad4 and translocate into the nucleus, where they activate transcription of target genes including snail, slug, and Twist. These genes regulate EMT through suppression of epithelial markers and induction of mesenchymal markers. Pretreatment with ADC inhibits TGF-β-induced EMT by inhibiting phosphorylation and transcriptional activation of Smad2/3. In addition, TGF-β activates β-catenin signaling cascade through the suppression of GSK3β, a negative regulator of β-catenin. The free form of β-catenin translocate into the nucleus and transcribe number of migration marker genes including MMP-2, MMP-9, and uPA. Pretreatment with ADC blocks TGF-β1-induced migration and invasion of MCF-7 cells through the suppression of transcriptional activation of β-catenin followed by down-regulation of MMP-2, MMP-9, and uPA. In addition, previous reports demonstrated that MMPs expression also regulated by snail, transcription factor regulates EMT. There is a possible that reduction in snail expression by may influence MMPs activity. Moreover, Twist was reported to be one of the down-stream target of β-catenin. Therefore, we believe that there may be a cross-talk existing between ADC-induced down-regulation of β-catenin and Twist.</p

    Sassarandainol: a new neolignan and anti-inflammatory constituents from the stem of <i>Sassafras randaiense</i>

    No full text
    <div><p>A new neolignan, (<i>R</i>)-( − )-sassarandainol (<b>1</b>), together with 10 known compounds (<b>2</b>–<b>11</b>), was isolated from the stem of <i>Sassafras randaiense</i>. The structures were determined by spectroscopic techniques. Among these isolates, γ-tocopherol (<b>5</b>), subamolide B (<b>7</b>) and β-sitosterone (<b>9</b>) exhibited moderate iNOS inhibitory activity on nitrite production induced (%) value of 30.51, 28.68 and 16.96, respectively.</p></div

    ADC inhibits TGF-β1-induced breast cancer cell migration and invasion.

    No full text
    <p>(A) Cell migration was determined by wound healing assay, the confluent MCF-7 monolayer was pre-treated with ADC (5–20 μM) for 2 h, cells were scratched by 200 μL pipet tips, and washed to remove the debris followed by addition of freshmedium containing 1% FBS and TGF-β1 (20 ng/mL). Cells were then incubated for 48 h. Photographs were taken at 0 h, and 48 h using inverted microscope with 10 × magnification. TGF-β1-induced cell motility was determined by measuring the area of wound closure as shown by histogram. The closure area at 48 h was compared with 0 h in the same samples. (B) For the invasion assay, the pre-treated cells were seed into the upper chamber of 24-well transwell chamber containing DMEM with 1% FBS. The lower chamber was filled with complete serum media. The cells were allowed to invade for 48 h. Invading cells were then fixed, and stained with Giemsa stain solution and counted in 5 random fields. The average invaded cells in each group was presented by histogram. The data reported as mean ± SD of three independent experiments. <sup>Θ</sup><i>P</i>< 0.001, significant difference from control and TGF-β1 alone treated group. *<i>P</i>< 0.05, **<i>P</i>< 0.01, and ***<i>P</i>< 0.001 were significantly different from TGF-β1 alone with the ADC treatment groups.</p

    ADC inhibits TGF-β1-induced transcriptional activity of Smad2/Smad3.

    No full text
    <p>(<b>A</b>) MCF-7 cells were pre-treated with ADC (5–20 μM) for 2 h prior to stimulation with TGF-β1 (20 ng/mL) for 24 h. Western blot analysis was performed to examine the protein expression levels of Snail, Slug, and TWIST with specific antibodies. β-actin was used as an internal loading control. (<b>B</b>) Cells were transfected with pGL3-SBE-4-Luc reporter construct, and then pre-treated with ADC (5–20 μM) prior to stimulation with TGF-β1 for 3 h. The luciferase activity was expressed as a relative value compared to that of the untreated cells which was set to 1-fold. (<b>C</b>) Western blot was performed to measure the total and phosphorylated levels of Smad2 and Smad3 proteins. (<b>D</b>) TGF-β1-induced nuclear translocation of phosphorylated Smad2 and Smad3 were examined by immunofluorescence analysis with confocal microscope. Cells were pre-treated with ADC (20 μM) for 2 h, and then incubated with TGF-β1 for 1 h. After treatment, cells were fixed, permiabilized, and incubated with Phos-Smad2 or Phos-Smad3 primary antibodies for overnight followed by FITC and TRITC secondary antibodies, respectively for 1 h. The cellular DNA was stained with DAPI (1 μg/mL) and images were captured by confocal microscope (magnification 200). Bars, 20 μm. The data reported as mean ± SD of three independent experiments. <sup>Θ</sup><i>P</i>< 0.001, significant difference from control and TGF-β1 alone treated group. *<i>P</i>< 0.05, **<i>P</i>< 0.01, and ***<i>P</i>< 0.001 were significantly different from TGF-β1 alone with the ADC treatment groups.</p

    Chemical structure of Antrodin C (ADC).

    No full text
    <p>Synonym: 3-Isobutyl-4-[4-(methyl-2-butenyloxy)phenyl]-1<i>H</i>-pyrrole-2,5-dione; Molecular formula: C<sub>19</sub>H<sub>23</sub>NO<sub>4</sub>; Molecular weight: 329.39.</p

    ADC blocks TGF-β1-induced EMT in breast cancer cells.

    No full text
    <p>MCF-7 cells were pre-treated with ADC (5–20 μM) for 2 h prior to stimulation with TGF-β1 (20 ng/mL) for 48 h. (<b>A</b>) Morphological changes especially cell scattering was examined by phase-contrast microscope. Photomicrography shown here are from one of the three independent experiments. (<b>B</b>) ADC inhibits TGF-β1-induced actin cytoskeleton reorganization in MCF-7 cells. ADC and TGF-β1 treated cells were fixed, permiabilized, and stained with FITC-phalloidin to visuvalize the F-actin cytoskeleton reorganization. The images are representative of three independent experiments. Bars, 20 μm. (<b>C</b>) Protein samples were isolated from control, ADC, and TGF-β treated cells for the detection of E-cadherin, occluding, vimentin, N-cadherin, and β-actin proteins. β-actin was used as an internal control. Western blot data presented are representative of those obtained in at least three independent experiments.</p

    ADC suppressed TGF-β1-induced transcriptional activation of β-catenin breast cancer cells.

    No full text
    <p>(<b>A</b>) MCF-7 cells were co-transfected with TOP-flash or FOP-flash or pCMV-β-Gal harboring luciferase reporter construct. After transfection, cells were pre-treated with ADC (5–20 μM) for 2 h and then stimulated with TGF-β1 for 3 h. Luciferase activity was determined and normalized with β-gal activity. The histogram shows the relative luciferase activity (fold increase). (<b>B</b>) Cells were pre-treated with ADC (5–20 μM) for 2 h and then stimulated with TGF-β1 for 2 h. β-catenin expression in cytoplasam and the nucleus were determined by western blot analysis using specific cytosolic and nuclear extracts. β-actin and histone were served as an internal control for cytosolic and nuclear fractions, respectively. (<b>C</b>) The nuclear localization of β-catenin in MCF-7 cells were determined by immunofluorescence staining. MCF-7 cells were seeded in a 8-well Tek chamber and allowed to adhere for 24 h. ADC (5–20 μM) for 2 h and then stimulated with TGF-β1 for 2 h. After treatment, cells were fixed, permiabilized, and incubated with β-catenin primary antibody overnight, followed by FITC secondary antibody for 1 h. The cellular DNA was stained with DAPI (1 μg/mL) and images were captured by confocal microscope (magnification 200). (<b>D</b>) Cells were pre-incubated with ADC (5–20 μM) for 2 h, and then stimulated with TGF-β1 for 1 h. The phosphorylated and total protein expression levels of β-catenin and GSK3β were determined by western blot analysis. The data reported as mean ± SD of three independent experiments. <sup>Θ</sup><i>P</i>< 0.001, significant difference from control and TGF-β1 alone treated group in TOP-Flash transfected groups. *<i>P</i>< 0.05, **<i>P</i>< 0.01, and ***<i>P</i>< 0.001 were significantly different from TGF-β1 alone with the ADC treatment groups in TOP-Flash transfected groups.</p

    Differential Gene Expression Network in Terpenoid Synthesis of <i>Antrodia cinnamomea</i> in Mycelia and Fruiting Bodies

    No full text
    <i>Antodia cinnamomea</i>, a precious brown-rot fungus endemic to Taiwan, has pharmaceutical applications due to its diverse array of metabolites. The terpenoids found in <i>A. cinnamomea</i> contribute to its most important bioactivities. We identified several terpenoid compounds in <i>A. cinnamomea</i> and revealed that their content in mycelium and fruiting body were significantly different. Using next-generation sequencing and an in-house transcriptome database, we identified several terpene synthase (TPS) candidates. After sequence analysis and functional characterization, 10 out of 12 candidates were found to have single or multiple terpene synthesis functions. Most of the terpenoid compounds were found to confer important bioactivities. RT-PCR results showed a positive correlation between terpene synthase expression pattern and terpenoid content. In addition, we identified several modification enzyme candidates that may be involved in the postmodification of terpenoid compounds with a genomic DNA scaffold, and a putative genetic network

    Characterization of the 2,3-Oxidosqualene Cyclase Gene from <i>Antrodia cinnamomea</i> and Enhancement of Cytotoxic Triterpenoid Compound Production

    No full text
    <i>Antrodia cinnamomea</i> is a scarce, epiphyte, host-specific, brown-rot fungus that produces diverse bioactive compounds with potent biological activity. Natural wild-type fruiting bodies of <i>A. cinnamomea</i> are rare and highly valued, but their artificial culture poses challenges. Triterpenoids are a group of secondary metabolites that contribute to the bioactivities of <i>A. cinnamomea</i>. 2,3-Oxidosqualene cyclase (OSC) is a key enzyme in triterpenoid biosynthesis, which converts 2,3-oxidosqualene (OS) into polycyclic triterpenoids. In this study, we isolated a 2,3-oxidosqualene cyclase gene from <i>A. cinnamomea</i> with degenerate primers and designated it as <i>AcOSC</i>. The full length <i>AcOSC</i> cDNA was subcloned into a yeast expression vector, and <i>AcOSC</i> activity was confirmed. RT-PCR results showed that <i>AcOSC</i> expression was highest in the wild-type fruiting body and correlated with a higher concentration of triterpenoids. <i>Agrobacterium</i>-mediated gene transformation was conducted to enhance the triterpenoid synthesis capacity of the cultured mycelium. Metabolite profiling was conducted by LC-MS/MS and principal component analysis (PCA). The compositions and contents of metabolites in the <i>AcOSC</i> transgenic lines were different from those in the wild-type mycelium and vector control. The levels of two important triterpenoids, dehydrosulphurenic acid (DSA) and dehydroeburicoic acid (DEA), were increased in <i>A. cinnamomea</i> oxidosqualene cyclase overexpression strains compared to controls. In summary an <i>Agrobacterium</i>-mediated gene transformation procedure was established that successfully increased the level of transgene expression and enhanced the triterpenoid content in cultured <i>A. cinnamomea.</i

    Bioassay Guided Isolation and Identification of Anti-inflammatory Active Compounds from the Root of Ficus formosana

    No full text
    Activity-directed fractionation and purification processes were employed to identify the anti-inflammatory active compounds using lipopolysaccharide (LPS)-stimulated mouse macrophage (RAW264.7) in vitro. Air-dried roots of Ficus formosana were extracted with methanol and separated into <i>n</i>-hexane, chloroform, ethyl acetate, <i>n</i>-butanol, and water layers. Among them, the chloroform layer showed strong activity and was subjected to separation and purification by using various chromatographic techniques. Five compounds showing potent activity were identified by comparing spectral data to be β-sitosterol, stigmasterol, psoralen, kaempferol, carpachromene, and syringic aldehyde. When macrophages were treated with psoralen and kaempferol together with LPS, a concentration-dependent inhibition of nitric oxide (NO) and tumor necrosis factor (TNF-α) productions were detected. Western blotting revealed that kaempferol, psoralen, and carpachromene blocked protein expression of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) in LPS-stimulated macrophages. The results confirmed that the traditional use of F. formosana could be a potential anti-inflammatory agent
    corecore