Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters

Database
Language
Affiliation country
Publication year range
1.
Redox Biol ; 69: 103030, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38181705

ABSTRACT

Ferroptosis is a type of programmed cell death resulting from iron overload-dependent lipid peroxidation, and could be promoted by activating transcription factor 3 (ATF3). SIRT1 is an enzyme accounting for removing acetylated lysine residues from target proteins by consuming NAD+, but its role remains elusive in ferroptosis and activating ATF3. In this study, we found SIRT1 was activated during the process of RSL3-induced glioma cell ferroptosis. Moreover, the glioma cell death was aggravated by SIRT1 activator SRT2183, but suppressed by SIRT inhibitor EX527 or when SIRT1 was silenced with siRNA. These indicated SIRT1 sensitized glioma cells to ferroptosis. Furthermore, we found SIRT1 promoted RSL3-induced expressional upregulation and nuclear translocation of ATF3. Silence of ATF3 with siRNA attenuated RSL3-induced increases of ferrous iron and lipid peroxidation, downregulation of SLC7A11 and GPX4 and depletion of cysteine and GSH. Thus, SIRT1 promoted glioma cell ferroptosis by inducting ATF3 activation. Mechanistically, ATF3 activation was reinforced when RSL3-induced decline of NAD+ was aggravated by FK866 that could inhibit NAD + synthesis via salvage pathway, but suppressed when intracellular NAD+ was maintained at higher level by supplement of exogenous NAD+. Notably, the NAD + decline caused by RSL3 was enhanced when SIRT1 was further activated by SRT2183, but attenuated when SIRT1 activation was inhibited by EX527. These indicated SIRT1 promoted ATF3 activation via consumption of NAD+. Finally, we found RSL3 activated SIRT1 by inducing reactive oxygen species-dependent upregulation of AROS. Together, our study revealed SIRT1 activated by AROS sensitizes glioma cells to ferroptosis via activation of ATF3-dependent inhibition of SLC7A11 and GPX4.


Subject(s)
Ferroptosis , Glioma , Humans , NAD , Activating Transcription Factor 3/genetics , Cell Line, Tumor , Sirtuin 1/genetics , Glioma/genetics , Glioma/metabolism , RNA, Small Interfering
2.
Inflammopharmacology ; 30(4): 1335-1349, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35708797

ABSTRACT

Gout is a chronic disease caused by monosodium urate (MSU) crystal deposition in the joints and surrounding tissues. We examined the effects of Taxifolin, a natural flavonoid mainly existing in vegetables and fruits, on MSU-induced gout. Pretreatment with Taxifolin significantly reduced IL-1ß, Caspase-1 and HMGB1 levels, upregulation of autophagy-related protein, LC3, as well as improved phagocytosis of macrophages. This study indicated that Taxifolin-attenuated inflammatory response in MSU-induced acute gout model by decreasing pro-inflammatory cytokine production and promoting the autophagy and phagocytic capacity of macrophages. Dietary supplementation with Taxifolin induces the autophagy and attenuated inflammatory response, which in consequence modulates acute gout. A preventive strategy combining dietary interventions with Taxifolin may offer a potential therapeutic alternative to pharmacological treatment to reduce inflammatory response to gout.


Subject(s)
Arthritis, Gouty , Gout , Arthritis, Gouty/chemically induced , Arthritis, Gouty/drug therapy , Autophagy , Gout/metabolism , Humans , Inflammation/drug therapy , Inflammation/metabolism , Interleukin-1beta/metabolism , Phagocytosis , Quercetin/analogs & derivatives , Uric Acid/metabolism
3.
Cancer Lett ; 425: 31-42, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29608987

ABSTRACT

RIP1 and RIP3 are necroptosis initiators, but their roles in regulation of glycolysis remain elusive. In this study, we found shikonin activated RIP1 and RIP3 in glioma cells in vitro and in vivo, which was accompanied with glycolysis suppression. Further investigation revealed that shikonin-induced decreases of glucose-6-phosphate and pyruvate and downregulation of HK II and PKM2 were significantly prevented when RIP1 or RIP3 was pharmacologically inhibited or genetically knocked down with SiRNA. Moreover, shikonin also triggered accumulation of intracellular H2O2 and depletion of GSH and cysteine. Mitigation of intracellular H2O2 via supplement of GSH reversed shikonin-induced glycolysis suppression. The role of intracellular H2O2 in regulation of glycolysis suppression was further confirmed in the cells treated with exogenous H2O2. Notably, inhibition of RIP1 or RIP3 prevented intracellular H2O2 accumulation, which was correlated with preventing shikonin-induced downregulation of x-CT and depletion of GSH and cysteine. In addition, supplement of pyruvate effectively inhibited shikonin- or exogenous H2O2-induced accumulation of intracellular H2O2 and glioma cell death. Taken together, we demonstrated in this study that RIP1 and RIP3 contributed to shikonin-induced glycolysis suppression via increasing intracellular H2O2.


Subject(s)
Glioma/drug therapy , Glycolysis/drug effects , Hydrogen Peroxide/metabolism , Naphthoquinones/administration & dosage , Nuclear Pore Complex Proteins/metabolism , RNA-Binding Proteins/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Animals , Cell Line, Tumor , Cysteine/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Glioma/metabolism , Glutathione/metabolism , Humans , Mice , Naphthoquinones/pharmacology , Rats , Xenograft Model Antitumor Assays
4.
Cancer Lett ; 428: 21-33, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29702192

ABSTRACT

Ferroptosis is a form of programmed cell death decided by iron-dependent lipid peroxidation, but its role in glioma cell death remains unclear. In this study, we found Pseudolaric acid B (PAB) inhibited the viabilities of glioma cells in vitro and in vivo, which was accompanied by abnormal increases of intracellular ferrous iron, H2O2 and lipid peroxidation, as well as depletion of GSH and cysteine. In vitro studies revealed that the lipid peroxidation and the cell death caused by PAB were both inhibited by iron chelator deferoxamine, but exacerbated by supplement of ferric ammonium citrate. Inhibition of lipid peroxidation with ferrostatin-1 or GSH rescued PAB-induced cell death. Morphologically, the cells treated with PAB presented intact membrane, shrunken mitochondria with increased membrane density, and normal-sized nucleus without chromatin condensation. Mechanistically, PAB improved intracellular iron by upregulation of transferrin receptor. The increased iron activated Nox4, which resulted in overproduction of H2O2 and lipid peroxides. Moreover, PAB depleted intracellular GSH via p53-mediated xCT pathway, which further exacerbated accumulation of H2O2 and lipid peroxides. Thus, PAB triggers ferroptosis in glioma cells and is a potential medicine for glioma treatment.


Subject(s)
Apoptosis/drug effects , Brain Neoplasms/drug therapy , Diterpenes/pharmacology , Glioma/drug therapy , Lipid Peroxidation/drug effects , Amino Acid Transport System y+/metabolism , Animals , Brain Neoplasms/pathology , Cell Line, Tumor/transplantation , Cyclohexylamines/pharmacology , Disease Models, Animal , Diterpenes/therapeutic use , Down-Regulation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Glioma/pathology , Glutathione/metabolism , Humans , Hydrogen Peroxide/metabolism , Iron/metabolism , Mice , Mice, Nude , Mitochondria/drug effects , Mitochondria/metabolism , NADPH Oxidase 4/metabolism , Phenylenediamines/pharmacology , Rats , Reactive Oxygen Species/metabolism , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL