• Title/Summary/Keyword: Silymarin

Search Result 183, Processing Time 0.026 seconds

Inhibition of Wnt Signaling by Silymarin in Human Colorectal Cancer Cells

  • Eo, Hyun Ji;Park, Gwang Hun;Jeong, Jin Boo
    • Biomolecules & Therapeutics
    • /
    • v.24 no.4
    • /
    • pp.380-386
    • /
    • 2016
  • Silymarin from milk thistle (Silybum marianum) has been reported to show an anti-cancer activity. In previous study, we reported that silymarin induces cyclin D1 proteasomal degradation through NF-${\kappa}B$-mediated threonine-286 phosphorylation. However, mechanism for the inhibition of Wnt signaling by silymarin still remains unanswered. Thus, we investigated whether silymarin affects Wnt signaling in human colorectal cancer cells to elucidate the additional anti-cancer mechanism of silymarin. Transient transfection with a TOP and FOP FLASH luciferase construct indicated that silymarin suppressed the transcriptional activity of ${\beta}$-catenin/TCF. Silymarin treatment resulted in a decrease of intracellular ${\beta}$-catenin protein but not mRNA. The inhibition of proteasome by MG132 and $GSK3{\beta}$ inhibition by SB216763 blocked silymarin-mediated downregulation of ${\beta}$-catenin. In addition, silymarin increased phosphorylation of ${\beta}$-catenin and a point mutation of S33Y attenuated silymarin-mediated ${\beta}$-catenin downregulation. In addition, silymarin decreased TCF4 and increased Axin expression in both protein and mRNA level. From these results, we suggest that silymarin-mediated downregulation of ${\beta}$-catenin and TCF4 may result in the inhibition of Wnt signaling in human colorectal cancer cells.

Silymarin-Mediated Degradation of c-Myc Contributes to the Inhibition of Cell Proliferation in Human Colorectal Cancer Cells

  • Eo, Hyun Ji;Jeong, Jin Boo;Koo, Jin Suk;Jeong, Hyung Jin
    • Korean Journal of Plant Resources
    • /
    • v.30 no.3
    • /
    • pp.265-271
    • /
    • 2017
  • In this study, we elucidated the molecular mechanism of silymarin by which silymarin may inhibits cell proliferation in human colorectal cancer cells in order to search the new potential anti-cancer target associated with the cell growth arrest. Silymarin reduced the level of c-Myc protein but not mRNA level indicating that silymarin-mediated downregulation of c-Myc may result from the proteasomal degradation. In the confirmation of silymarin-mediated c-Myc degradation, MG132 as a proteasome inhibitor attenuated c-Myc degradation by silymarin. In addition, silymarin phosphorylated the threonine-58 (Thr58) of c-Myc and the point mutation of Thr58 to alanine blocked its degradation by silymarin, which indicates that Thr58 phosphorylation may be an important modification for silymarin-mediated c-Myc degradation. We observed that the inhibition of ERK1/2, p38 and $GSK3{\beta}$ blocked the Thr58 phosphorylation and subsequent c-Myc degradation by silymarin. Finally, the point mutation of Thr58 to alanine attenuated silymarin-mediated inhibition of the cell growth. The results suggest that silymarin induces the cell growth arrest through c-Myc proteasomal degradation via ERK1/2, p38 and $GSK3{\beta}-dependent$ Thr58 phosphorylation.

Antioxidative Effects of Silymarin and Silybin Purified from Silybum marianum on Lipid Peroxidation (엉겅퀴로부터 분리 정제한 Silymarin 및 Silybin의 지질 과산화에 대한 항산화 효과)

  • 이백천;박종옥;류병호
    • The Korean Journal of Food And Nutrition
    • /
    • v.10 no.1
    • /
    • pp.37-43
    • /
    • 1997
  • This study was undertaken to evaluate as antioxidant activity against lipid peroxidation. Silymarin and silybin extracted from Silybum marianum were successively purified wit solvent fractionation by silica gel column chromatography. These isoflavonoid inhibited superoxide anion production in the xanthine oxidase system. In the rat liver microsomes, silymarin or silybin rapidly inhibited lipid peroxidation which was initiated enzymatically by reduced nicotinamide adenine dinucleotide phosphate(NADPH) or non-enzymatically by ascorbic acid or Fenton's reagent (H2O2+Fe2+). Mitochondrial lipid peroxidation was also inhibited by silymarin and silybin. silymarin and silybin inhibited on terminating radical chain reaction during lipid peroxidation in the enzymatic system of microsomes or in the linoleic acid hydroperoxide induced peroxidation system.

  • PDF

The Effect of Silymarin and Ethanol Intake on Vascular Contractility (엉겅퀴 유래 Silymarin의 단독 및 알코올 병용 시 혈압 조절 효과)

  • Je, Hyun Dong;Min, Young Sil
    • Journal of Industrial Convergence
    • /
    • v.20 no.7
    • /
    • pp.131-137
    • /
    • 2022
  • In the study, we endeavored to assess the convergence effect of Silybum marianum-derived silymarin and epidemiologically-correlated alcohol intake on vascular contractility and to determine the mechanism involved. There were few reports addressing the question whether thin or thick filament modulation is included in ethanol and silymarin-induced regulation. We hypothesized that ethanol at a low concentration and silymarin play a role in agonist-dependent regulation of vascular contractility. Denuded arterial muscles of Sprague-Dawley male rats were suspended in organ baths and isometric tensions were transduced and recorded using isometric transducers and an automatic data acquisition system. Interestingly, both silymarin and ethanol didn't encourage silymarin alone-induced inhibition in agonists-induced contraction suggesting that endothelial nitric oxide synthesis might be involved in ethanol or silymarin-induced modulation of vascular contractility and additional pathways besides endothelial nitric oxide synthesis such as ROCK inactivation might be involved in the silymarin-induced modulation of vascular contractility.

Activating Transcription Factor 3 is a Molecular Target for Apoptotic Effect of Silymarin in Human Colorectal Cancer Cells

  • Eo, Hyun Ji;Park, Gwang Hun;Jeong, Jin Boo
    • Korean Journal of Plant Resources
    • /
    • v.29 no.3
    • /
    • pp.281-288
    • /
    • 2016
  • Apoptosis has been regarded as a therapeutic target because apoptosis is typically disturbed in human cancer. Silymarin found in the seeds of the milk thistle (Silybum marianum) has been reported to exert anti-cancer properties through apoptosis. This study was performed to investigate the molecular target for silymarin-mediated apoptosis in human colorectal cancer cells. Silymarin reduced the cell viability and induced an apoptosis in human colorectal cancer cells. ATF3 overexpression increased PARP cleavage by silymarin. Increased ATF3 expression in both protein and mRNA was observed in silymarin-treated cells. In addition, silymarin increased the luciferase activity of ATF3 promoter. Inhibition of JNK and IκK-α blocked silymarin-mediated ATF3 expression. The results suggest that silymarin induces apoptosis through JNK and IκKα-dependent ATF3 expression in human colorectal cancer cells.

Enhanced Antioxidative Potential by Silymarin Treatment through the Inductionof Nrf2/MAPK Mediated HO-1 Signaling Pathway in RAW 264.7 Cells (RAW 264.7 세포에서 Nrf2/MAPK 의 활성을 통한 HO-1 과발현에 의한 silymarin의 항산화 효과)

  • Hyun-Seo Yoon;Hyun An;Chung Mu Park
    • Journal of Life Science
    • /
    • v.33 no.10
    • /
    • pp.776-782
    • /
    • 2023
  • Silymarin, which is derived from dried Silybum marianum (milk thistle) seeds and fruits, possesses various beneficial properties, such as hepatoprotective, antioxidative, anti-inflammatory, and anticancer activity. This research aimed to explore the antioxidative activity of silymarin against oxidative stress and understand its molecular mechanism in RAW 264.7 cells. The study employed cell viability and reactive oxygen species (ROS) formation assays and western blot analysis. The results demonstrated that silymarin effectively reduced intracellular ROS levels induced by lipopolysaccharide (LPS) in a dose-dependent manner without causing any cytotoxic effects. Moreover, silymarin treatment significantly upregulated the expression of heme oxygenase (HO)-1, a phase II enzyme known for its potent antioxidative activity. Additionally, silymarin treatment significantly induced the expression of nuclear factor-erythroid 2 p45-related factor (Nrf) 2, a transcription factor responsible for regulating antioxidative enzymes, which was consistent with the upregulated HO-1 expression. To investigate the involvement of key signaling pathways in maintaining cellular redox homeostasis against oxidative stress, the phosphorylation status of mitogen-activated protein kinase (MAPK) and phosphoinositide 3-kinase (PI3K) was estimated by western blot analysis. The results showed that silymarin potently induced HO-1 expression, which was mediated by the phosphorylation of p38 MAPK. To further validate the antioxidative potential of silymarin-induced HO-1 expression, tert-butyl hydroperoxide (t-BHP)-induced oxidative damage was employed and attenuated by silymarin treatment, as identified by a selective inhibitor for each signaling molecule. In conclusion, silymarin robustly enhanced antioxidative activity by inducing HO-1 via the Nrf2/p38 MAPK signaling pathway in RAW 264.7 cells.

Effects of Polyoxyl 40 Hydrogenated Castor Oil on Solubility and Bioavailability of Silymarin in Combined Preparation Containing Silymarin and Ursodeoxycholic Acid (Polyoxyl 40 Hydrogenated Castor Oil이 실리마린과 우루소데옥시콜린산 복합제제중 실리마린의 용해성 및 생체이용률에 미치는 영향)

  • 장우익;남권호;조재열;이재희;유영호;박명호;김재환
    • Biomolecules & Therapeutics
    • /
    • v.5 no.3
    • /
    • pp.272-277
    • /
    • 1997
  • The effect of nonionic surfactant(polyoxyl 40 hydrogenated castor oil, PHCO), a common solubi-lizer, on the solubility of silymarin in the combined preparation containing ursoseoxycholic acid(UDCA) and silymarin was investigated in vivo using HPLC. The solubility of silybin, a major component of silymarin, was enhanced by increasing the amount of PHCO. The effect of PHCO on bioavailability was also evaluated in rats. The bioavailability was calculated by silybin content in bile juice that was excreted for 24 hr after oral administration. It was found that the bioavailability of silymarin containing PHCO was significantly increased compared to that of control. These results suggest that PHCO may improve the solubility and bioavailabilty of silymarin when it is combined with UDCA and silymarin.

  • PDF

Antioxidative Effect of Silmarin Purified from Silybum Marianum on Modificaion of Human Low Density Lipoprotein (엉겅퀴에서 분리 정제한 Silymarin의 사람 Low Density Lipoprotein에 대한 항산화 효과)

  • 류병호;이백천;진성현;조경자;김동석
    • Journal of Food Hygiene and Safety
    • /
    • v.12 no.1
    • /
    • pp.1-8
    • /
    • 1997
  • This study was performed to evaluate the antioxidant activity of silymarin against human low density lipoproteins(LDL) oxidation. Silymarin extracted from Silybum marianum was successively purified with solvent fractionation and followed by silica gel column chromatography. The active substances were separated by HPLC and the isolated active substances, silymarin were identified by IR, NMR, GC-MS as silymarin. Silymarin inhibited at the 5 $\mu$M Cu2+-mediated oxidation of human low density lipoprotein (LDL) in a dose dependent manner. Silymarin completely inhibited LDL oxidation at 50 $\mu\textrm{g}$/$m\ell$ concentration. These findings suggest that silymarin may protect LDL against oxidation in atherosclerotic lessions.

  • PDF

Silymarin Attenuates Invasion and Migration through the Regulation of Epithelial-mesenchymal Transition in Huh7 Cells (간암세포주에서 상피간엽전환억제를 통한 Silymarin의 침윤 및 전이 억제 효과)

  • Kim, Do-Hoon;Park, So-Jeong;Lee, Seung-Yeon;Yoon, Hyun-Seo;Park, Chung Mu
    • Korean Journal of Clinical Laboratory Science
    • /
    • v.50 no.3
    • /
    • pp.337-344
    • /
    • 2018
  • Hepatocellular carcinoma (HCC), a major type of hepatoma, is associated with high recurrence and mortality because of its uncontrolled metastatic feature. Silymarin is a polyphenolic flavonoid from Silybum marianun (milk thistle) and exhibits anti-carcinogenic activity through modulation of the epithelial-mesenchymal transition (EMT) in several cancer cells. In this study, the inhibitory mechanism of silymarin against migration and invasion was investigated in the Huh7 HCC cell line. Wound healing and in vitro invasion assays were conducted to examine the effects of silymarin on migration and invasion. Western blot analysis was also applied to evaluate the inhibitory effects of silymarin on the EMT-related genes and their upstream signaling molecules. Silymarin inhibited the migratory and invasive activities of Huh7 cells. In addition, silymarin attenuated the protein expression levels of vimentin and matrix metalloproteinase (MMP)-9 as well as their transcription factors, Snail, and nuclear factor $(NF)-{\kappa}B$, while the expression of E-cadherin was increased by the silymarin treatment. Among the upstream signaling molecules, the phosphorylation of Akt was inhibited by the silymarin treatment, which was confirmed by the selective inhibitor, LY294002. Consequently, silymarin inhibited the invasive and migratory activities in Huh7 cells through the modulation of EMT-related gene expression by the PI3K/Akt signaling pathway, which may have potential as a chemopreventive agent against HCC metastasis.

Silymarin Inhibits Cytokine-Stimulated Pancreatic Beta Cells by Blocking the ERK1/2 Pathway

  • Kim, Eun Jeong;Kim, Jeeho;Lee, Min Young;Sudhanva, Muddenahalli Srinivasa;Devakumar, Sundaravinayagam;Jeon, Young Jin
    • Biomolecules & Therapeutics
    • /
    • v.22 no.4
    • /
    • pp.282-287
    • /
    • 2014
  • We show that silymarin, a polyphenolic flavonoid isolated from milk thistle (Silybum marianum), inhibits cytokine mixture (CM: TNF-${\alpha}$, IFN-${\gamma}$, and IL-$1{\beta}$)-induced production of nitric oxide (NO) in the pancreatic beta cell line MIN6N8a. Immunostaining and Western blot analysis showed that silymarin inhibits iNOS gene expression. RT-PCR showed that silymarin inhibits iNOS gene expression in a dose-dependent manner. We also showed that silymarin inhibits extracellular signal-regulated protein kinase-1 and 2 (ERK1/2) phosphorylation. A MEK1 inhibitor abrogated CM-induced nitrite production, similar to silymarin. Treatment of MIN6N8a cells with silymarin also inhibited CM-stimulated activation of NF-${\kappa}B$, which is important for iNOS transcription. Collectively, we demonstrate that silymarin inhibits NO production in pancreatic beta cells, and silymarin may represent a useful anti-diabetic agent.