Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Theranostics ; 13(12): 4288-4302, 2023.
Article in English | MEDLINE | ID: mdl-37554285

ABSTRACT

Rationale: As a key endogenous negative regulator of ferroptosis, glutathione peroxidase 4 (GPX4) can regulate its antioxidant function through multiple post-translational modification pathways. However, the effects of the phosphorylation/dephosphorylation status of GPX4 on the regulation of inducible ferroptosis in hepatocellular carcinoma (HCC) remain unclear. Methods: To investigate the effects and molecular mechanism of GPX4 phosphorylation/dephosphorylation modification on ferroptosis in HCC cells. Sorafenib (Sora) was used to establish the ferroptosis model in HCC cells in vitro. Using the site-directed mutagenesis method, we generated the mimic GPX4 phosphorylation or dephosphorylation HCC cell lines at specific serine sites of GPX4. The effects of GPX4 phosphorylation/dephosphorylation modification on ferroptosis in HCC cells were examined. The interrelationships among GPX4, p53, and protein phosphatase 2A-B55ß subunit (PP2A-B55ß) were also explored. To explore the synergistic anti-tumor effects of PP2A activation on Sora-administered HCC, we established PP2A-B55ß overexpression xenograft tumors in a nude mice model in vivo. Results: In the Sora-induced ferroptosis model of HCC in vitro, decreased levels of cytoplasmic and mitochondrial GPX4, mitochondrial dysfunction, and enhanced p53 retrograde signaling occurred under Sora treatment. Further, we found that mitochondrial p53 retrograded remarkably into the nucleus and aggravated Sora-induced ferroptosis. The phosphorylation status of GPX4 at the serine 2 site (GPX4Ser2) revealed that mitochondrial p-GPX4Ser2 dephosphorylation was positively associated with ferroptosis, and the mechanism might be related to mitochondrial p53 retrograding into the nucleus. In HCC cells overexpressing PP2A-B55ß, it was found that PP2A-B55ß directly interacted with mitochondrial GPX4 and promoted Sora-induced ferroptosis in HCC. Further, PP2A-B55ß reduced the interaction between mitochondrial GPX4 and p53, leading to mitochondrial p53 retrograding into the nucleus. Moreover, it was confirmed that PP2A-B55ß enhanced the ferroptosis-mediated tumor growth inhibition and mitochondrial p53 retrograde signaling in the Sora-treated HCC xenograft tumors. Conclusion: Our data uncovered that the PP2A-B55ß/p-GPX4Ser2/p53 axis was a novel regulatory pathway of Sora-induced ferroptosis. Mitochondrial p-GPX4Ser2 dephosphorylation triggered ferroptosis via inducing mitochondrial p53 retrograding into the nucleus, and PP2A-B55ß was an upstream signal modulator responsible for mitochondrial p-GPX4Ser2 dephosphorylation. Our findings might serve as a potential theranostic strategy to enhance the efficacy of Sora in HCC treatment through the targeted intervention of p-GPX4 dephosphorylation via PP2A-B55ß activation.


Subject(s)
Carcinoma, Hepatocellular , Ferroptosis , Liver Neoplasms , Phospholipid Hydroperoxide Glutathione Peroxidase , Protein Phosphatase 2 , Sorafenib , Animals , Humans , Mice , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Nucleus , Down-Regulation , Drug Resistance, Neoplasm , Heterografts , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mice, Inbred BALB C , Mice, Nude , Mitochondria/pathology , Neoplasm Transplantation , Phospholipid Hydroperoxide Glutathione Peroxidase/chemistry , Phospholipid Hydroperoxide Glutathione Peroxidase/genetics , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Phosphorylation , Signal Transduction/drug effects , Sorafenib/therapeutic use , Protein Phosphatase 2/metabolism
2.
Clin Transl Med ; 11(9): e517, 2021 09.
Article in English | MEDLINE | ID: mdl-34586745

ABSTRACT

BACKGROUND: Platinum-based chemotherapy is effective in inducing shrinkage of primary lung cancer lesions; however, it shows finite therapeutic efficacy in patients suffering from brain metastasis (BM). The intrinsic changes of BM cells, which contribute to the poor results remain unknown. METHODS: Platinum drug-sensitivity was assessed by utilizing a preclinical BM model of PC9 lung adenocarcinoma cells in vitro and in vivo. High consumption of glutathione (GSH) and two associated upregulated proteins (GPX4 and GSTM1) in BM were identified by integrated metabolomics and proteomics in cell lines and verified by clinical serum sample. Gain-of-function and rescue experiments were implemented to reveal the impact and mechanism of GPX4 and GSTM1 on the chemosensitivity in BM. The interaction between GPX4 and GSTM1 was examined by immunoblotting and immunoprecipitation. The mechanism of upregulation of GPX4 was further uncovered by luciferase reporter assay, immunoprecipitation, and electrophoretic mobility shift assay. RESULTS: The derivative brain metastatic subpopulations (PC9-BrMs) of parental cells PC9 developed obvious resistance to platinum. Radically altered profiles of BM metabolism and protein expression compared with primary lung cancer cells were described and GPX4 and GSTM1 were identified as being responsible for the high consumption of GSH, leading to decreased chemosensitivity by negatively regulating ferroptosis. Besides, GSTM1 was found regulated by GPX4, which was transcriptionally activated by the Wnt/NR2F2 signaling axis in BM. CONCLUSIONS: Collectively, our findings demonstrated that Wnt/NR2F2/GPX4 promoted acquired chemoresistance by suppressing ferroptosis with high consumption of GSH. GPX4 inhibitor was found to augment the anticancer effect of platinum drugs in lung cancer BM, providing novel strategies for lung cancer patients with BM.


Subject(s)
Brain Neoplasms , Drug Resistance, Neoplasm , Lung Neoplasms , Phospholipid Hydroperoxide Glutathione Peroxidase , Platinum/pharmacology , Animals , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/secondary , Cell Line, Tumor , Cell Survival/drug effects , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Ferroptosis/genetics , Glutathione/metabolism , Glutathione Transferase/chemistry , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Phospholipid Hydroperoxide Glutathione Peroxidase/antagonists & inhibitors , Phospholipid Hydroperoxide Glutathione Peroxidase/chemistry , Phospholipid Hydroperoxide Glutathione Peroxidase/genetics , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism
3.
Acta Crystallogr D Struct Biol ; 77(Pt 2): 237-248, 2021 Feb 01.
Article in English | MEDLINE | ID: mdl-33559612

ABSTRACT

Wild-type human glutathione peroxidase 4 (GPX4) was co-expressed with SBP2 (selenocysteine insertion sequence-binding protein 2) in human HEK cells to achieve efficient production of this selenocysteine-containing enzyme on a preparative scale for structural biology. The protein was purified and crystallized, and the crystal structure of the wild-type form of GPX4 was determined at 1.0 Šresolution. The overall fold and the active site are conserved compared with previously determined crystal structures of mutated forms of GPX4. A mass-spectrometry-based approach was developed to monitor the reaction of the active-site selenocysteine Sec46 with covalent inhibitors. This, together with the introduction of a surface mutant (Cys66Ser), enabled the crystal structure determination of GPX4 in complex with the covalent inhibitor ML162 [(S)-enantiomer]. The mass-spectrometry-based approach described here opens the path to further co-complex crystal structures of this potential cancer drug target in complex with covalent inhibitors.


Subject(s)
Enzyme Inhibitors , Phospholipid Hydroperoxide Glutathione Peroxidase , Crystallography, X-Ray , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , HEK293 Cells , Humans , Phospholipid Hydroperoxide Glutathione Peroxidase/chemistry , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Protein Binding , Protein Conformation
4.
J Agric Food Chem ; 69(2): 638-645, 2021 Jan 20.
Article in English | MEDLINE | ID: mdl-33398988

ABSTRACT

With widespread applications of the latest neonicotinoid in agriculture, dinotefuran has gradually become a hazardous contaminant for plants through the generation of excessive reactive oxygen species. However, the potential toxic mechanisms of oxidative damages to plants induced by dinotefuran are still unknown. As a core component of the glutathione antioxidant enzyme system, glutathione peroxidases have been used as biomarkers to reflect excessive oxidative stress. In this study, the hazardous effects of dinotefuran on AtGPX6 were investigated at the molecular level. The intrinsic fluorescence intensity of AtGPX6 was quenched using the static quenching mechanism upon binding with dinotefuran. Moreover, a single binding site was predicted for AtGPX6 toward dinotefuran, and the complex formation was presumed to be driven by hydrogen bonds or van der Waals forces, which conformed with the molecular docking results. In addition, AtGPX6 exhibited moderate binding affinity with dinotefuran based on the bio-layer interferometry assay. In addition, the loosening and unfolding of the protein skeleton of AtGPX6 with the addition of dinotefuran were explored along with the increase of hydrophobicity around tryptophan residues. Lastly, the toxic effects of dinotefuran on the root growth of Arabidopsis seedlings were also examined. The exploration of the binding mechanism of dinotefuran with AtGPX6 at the molecular level would provide the toxicity assessment of dinotefuran on plants.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/drug effects , Arabidopsis/enzymology , Guanidines/pharmacology , Insecticides/pharmacology , Neonicotinoids/pharmacology , Nitro Compounds/pharmacology , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Arabidopsis/chemistry , Arabidopsis/genetics , Arabidopsis Proteins/chemistry , Arabidopsis Proteins/genetics , Binding Sites , Insecticides/chemistry , Molecular Docking Simulation , Phospholipid Hydroperoxide Glutathione Peroxidase/chemistry , Phospholipid Hydroperoxide Glutathione Peroxidase/genetics , Seedlings/chemistry , Seedlings/drug effects , Seedlings/enzymology , Seedlings/genetics
5.
Nat Chem Biol ; 16(5): 497-506, 2020 05.
Article in English | MEDLINE | ID: mdl-32231343

ABSTRACT

We recently described glutathione peroxidase 4 (GPX4) as a promising target for killing therapy-resistant cancer cells via ferroptosis. The onset of therapy resistance by multiple types of treatment results in a stable cell state marked by high levels of polyunsaturated lipids and an acquired dependency on GPX4. Unfortunately, all existing inhibitors of GPX4 act covalently via a reactive alkyl chloride moiety that confers poor selectivity and pharmacokinetic properties. Here, we report our discovery that masked nitrile-oxide electrophiles, which have not been explored previously as covalent cellular probes, undergo remarkable chemical transformations in cells and provide an effective strategy for selective targeting of GPX4. The new GPX4-inhibiting compounds we describe exhibit unexpected proteome-wide selectivity and, in some instances, vastly improved physiochemical and pharmacokinetic properties compared to existing chloroacetamide-based GPX4 inhibitors. These features make them superior tool compounds for biological interrogation of ferroptosis and constitute starting points for development of improved inhibitors of GPX4.


Subject(s)
Enzyme Inhibitors/pharmacology , Nitriles/chemistry , Nitriles/pharmacology , Phospholipid Hydroperoxide Glutathione Peroxidase/antagonists & inhibitors , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Animals , Cell Line, Tumor , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacokinetics , Ferroptosis/drug effects , Humans , Lipid Peroxidation/drug effects , Mice, SCID , Molecular Probes/chemistry , Molecular Targeted Therapy , Oxides/chemistry , Phospholipid Hydroperoxide Glutathione Peroxidase/chemistry , Prodrugs/chemistry , Rats, Wistar , Selenocysteine/chemistry , Selenocysteine/metabolism , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Structure-Activity Relationship
6.
Spectrochim Acta A Mol Biomol Spectrosc ; 226: 117597, 2020 Feb 05.
Article in English | MEDLINE | ID: mdl-31629975

ABSTRACT

Along with non-biodegradability and accumulation in agricultural soil, lead (II) ions exert considerable harmful effects on plants even at trace amount, especially for the oxidative damages elicited by the lead ions-induced excessive reactive oxygen species (ROS). The glutathione peroxidases were reported to be correspondent with the oxidative stress induced by heavy metals. However, limited data are available about the potential hazardous mechanisms of the lead ions-induced oxidative damage to plants at molecular level. In this study, the harmful impacts of lead ions on Arabidopsis thaliana glutathione peroxidase 6 (AtGPX6) were assessed based on multi-spectroscopic measurements and molecular docking study. The characteristic fluorescence of AtGPX6 was quenched by lead ions with static mechanism at different temperatures. AtGPX6 exhibits a single binding site with lead ions, and then the complex formation was mainly driven by hydrogen bonding interaction and van der Waals forces on account of the negative ΔH and ΔS. The secondary structural changes were observed from the synchronous fluorescence, UV-visible absorption and Circular dichroism spectra, which led to loosen and unfold of the protein framework accompanied by the incremental hydrophobicity around the vicinity of the tryptophan residues. Therefore, this work illustrates the detailed binding mode between lead (II) ions and glutathione peroxidase 6 from Arabidopsis thaliana and the toxic effects on antioxidative defense system induced by lead ions at molecular level.


Subject(s)
Arabidopsis Proteins/chemistry , Arabidopsis Proteins/metabolism , Lead/chemistry , Lead/pharmacokinetics , Molecular Docking Simulation , Phospholipid Hydroperoxide Glutathione Peroxidase/chemistry , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Arabidopsis/drug effects , Arabidopsis/enzymology , Arabidopsis/metabolism , Binding Sites , Biophysical Phenomena , Circular Dichroism , Fluorescence , Ions/chemistry , Ions/pharmacokinetics , Models, Molecular , Oxidative Stress/drug effects , Protein Binding , Protein Domains/drug effects , Spectrometry, Fluorescence , Spectrophotometry, Ultraviolet
SELECTION OF CITATIONS
SEARCH DETAIL
...