• Title/Summary/Keyword: Dihydroceramide

Search Result 4, Processing Time 0.016 seconds

Dihydroceramide was Highly Elevated by the Fumonisin B1 and Desipramine in Sphingomonas chungbukensis

  • Burenjargal, Munkhtsatsral;Lee, Youn-Sun;Yoo, Jae-Myung;Choi, Mi-Hwa;Ji, So-Young;Lee, Yong-Moon;Kim, Young-Chang;Oh, Sei-Kwan;Yun, Yeo-Pyo;Yoo, Hwan-Soo
    • Biomolecules & Therapeutics
    • /
    • v.16 no.2
    • /
    • pp.100-105
    • /
    • 2008
  • The sphingolipid metabolites act as lipid mediator for cell proliferation and apoptosis in mammalian cells. In bacteria, sphingolipid metabolism remains unknown. The purpose of this study was to investigate whether sphingolipid metabolism is potential target for fumonisin $B_1$($FB_1$) and desipramine in Sphingomonas chungbukensis, Gram-negative bacteria, by comparing the intracellular contents of bacterial sphingolipids with ones of HIT-T15 ${\beta}$-cells, hamster pancreatic cells. The concentrations of ceramide and dihydroceramide were 18.0 ${\pm}$ 12.0 and 0.025 ${\pm}$ 0.018 nmol/mg protein, respectively, in HIT-T15 cells. However, the concentrations of ceramide and dihydroceramide in the bacterial culture were 2.0 ${\pm}$ 1.2 and 10.6 ${\pm}$ 5.5 nmol/mg protein, respectively. $FB_1$ decreased the level of ceramide from 18.0 to 3.8 nmol/mg protein in HIT-T15 ${\beta}$-cells. However, dihydroceramide content in $FB_1$-treated HIT-T15 cells was slightly decreased compared with the control culture. When S. chungbukensis was treated with either $FB_1$ or desipramine, dihydroceramide level was increased by 5- and 4-fold, respectively, compared with the control bacteria. These results indicate that $FB_1$ and desipramine may act as an activator in bacterial sphingolipid biosynthetic pathway, and bacterial sphingolipid metabolism pathway appears to be different from the pathway of mammalian cells.

Study of Complexes of C2- and C6-dihydroceramides with Transition Metal Ions Using Electrospray Ionization Tandem Mass Spectrometry (ESI-MS/MS)

  • Lim, Jin-Yi;Kumar, Avvaru Praveen;Kim, Chang-Dae;Ahn, Chul-Jin;Yoo, Young-Jae;Lee, Yong-Ill
    • Bulletin of the Korean Chemical Society
    • /
    • v.30 no.2
    • /
    • pp.397-401
    • /
    • 2009
  • The complexes of $C_2-\;and\;C_6$-dihydroceramides with transition metal ions have been investigated by using Electrospray ionization-tandem mass spectrometry (ESI-MS/MS). The formation and fragmentation pathways of several doubly charged cluster ions as well as singly charged cluster ions of $C_2-\;and\;C_6$-dihydroceramides with transition metal ions have studied by ESI-MS/MS in the positive mode. Under ESI conditions, dihydroceramides form singly and doubly charged complexes with transition metal ions $(Mn^{2+},\;Fe^{2+},\;Co^{2+},\;Ni^{2+},\;and\;Zn^{2+}\;except\;Cu^{2+})$ with the compositions of $[DHCer+M+2H^2O-H]^+,\;[2DHCer+M+2H2O-H]^+,\;[3DHCer+M+2H2O-H]^+,\;[2DHCer+M]^{2+},\;[3DHCer+M]^{2+},\;[4DHCer+M]^{2+},\;[5DHCer+M]^{2+},\;and\;[6DHCer+M]^{2+}\;(DHCer\;=\;C_2-\;or\;C_6$-dihydroceramide, M = transition metal ion). The different complexation behavior of copper is responsible for relatively lower affinity of dihydroceramides to copper compared to those of other transition metals. It is also found that in the mass spectrum of the dihydroceramide complexes with copper(II), [2DHCer+Cu-H]$^+$ was observed with considerable intensity as well as [2DHCer+Cu+2$H_2O-H]^+$ due to its different geometry from those of other metals.

Celecoxib-mediated activation of endoplasmic reticulum stress induces de novo ceramide biosynthesis and apoptosis in hepatoma HepG2 cells

  • Maeng, Hyo Jin;Song, Jae-Hwi;Kim, Goon-Tae;Song, Yoo-Jeong;Lee, Kangpa;Kim, Jae-Young;Park, Tae-Sik
    • BMB Reports
    • /
    • v.50 no.3
    • /
    • pp.144-149
    • /
    • 2017
  • Ceramides are the major sphingolipid metabolites involved in cell survival and apoptosis. When HepG2 hepatoma cells were treated with celecoxib, the expression of the genes in de novo sphingolipid biosynthesis and sphingomyelinase pathway was upregulated and cellular ceramide was elevated. In addition, celecoxib induced endoplasmic reticulum (ER) stress in a time-dependent manner. SPTLC2, a subunit of serine palmitoyltransferase, was overexpressed by adenovirus. Adenoviral overexpression of SPTLC2 (AdSPTLC2) decreased cell viability of HEK293 and HepG2 cells. In addition, AdSPTLC2 induced apoptosis via the caspase-dependent apoptotic pathway and elevated cellular ceramide, sphingoid bases, and dihydroceramide. However, overexpression of SPTLC2 did not induce ER stress. Collectively, celecoxib activates de novo sphingolipid biosynthesis and the combined effects of elevated ceramide and transcriptional activation of ER stress induce apoptosis. However, activation of de novo sphingolipid biosynthesis does not activate ER stress in hepatoma cells and is distinct from the celecoxib-mediated activation of ER stress.

Inhibition of Sphingolipid Metabolism Enhances Resveratrol Chemotherapy in Human Gastric Cancer Cells

  • Shin, Kyong-Oh;Park, Nam-Young;Seo, Cho-Hee;Hong, Seon-Pyo;Oh, Ki-Wan;Hong, Jin-Tae;Han, Sang-Kil;Lee, Yong-Moon
    • Biomolecules & Therapeutics
    • /
    • v.20 no.5
    • /
    • pp.470-476
    • /
    • 2012
  • Resveratrol, a chemopreventive agent, is rapidly metabolized in the intestine and liver via glucuronidation. Thus, the pharmacokinetics of resveratrol limits its efficacy. To improve efficacy, the activity of resveratrol was investigated in the context of sphingolipid metabolism in human gastric cancer cells. Diverse sphingolipid metabolites, including dihydroceramides (DHCer), were tested for their ability to induce resveratrol cytotoxicity. Exposure to resveratrol ($100{\mu}M$) for 24 hr induced cell death and cell cycle arrest in gastric cancer cells. Exposure to the combination of resveratrol and dimethylsphingosine (DMS) increased cytotoxicity, demonstrating that sphingolipid metabolites intensify resveratrol activity. Specifically, DHCer accumulated in a resveratrol concentration-dependent manner in SNU-1 and HT-29 cells, but not in SNU-668 cells. LC-MS/MS analysis showed that specific DHCer species containing C24:0, C16:0, C24:1, and C22:0 fatty acids chain were increased by up to 30-fold by resveratrol, indicating that resveratrol may partially inhibit DHCer desaturase. Indeed, resveratrol mildly inhibited DHCer desaturase activity compared to the specific inhibitor GT-11 or to retinamide (4-HPR); however, in SNU-1 cells resveratrol alone exhibited a typical cell cycle arrest pattern, which GT-11 did not alter, indicating that inhibition of DHCer desaturase is not essential to the cytotoxicity induced by the combination of resveratrol and sphingolipid metabolites. Resveratrol-induced p53 expression strongly correlated with the enhancement of cytotoxicity observed upon combination of resveratrol with DMS or 4-HPR. Taken together, these results show that DHCer accumulation is a novel lipid biomarker of resveratrol-induced cytotoxicity in human gastric cancer cells.