39 research outputs found

    Inactivation of the glutathione peroxidase GPx4 by the ferroptosis-inducing molecule RSL3 requires the adaptor protein 14-3-3 epsilon

    Get PDF
    RSL3, a drug candidate prototype for cancer chemotherapy, triggers ferroptosis by inactivating GPx4. Here we report the purification of the protein indispensable for GPx4 inactivation by RSL3. MS analysis reveals 14-3-3 isoforms as candidates and recombinant human 14-3-3epsilon confirms the identification. The function of 14-3-3\uf065 is redox-regulated. Moreover, overexpression and silencing of the gene coding for 14-3-3\uf065 consistently control the inactivation of GPx4 by RSL3. The interaction of GPx4 with a redox-regulated adaptor protein, operating in cell signalling, further contributes to frame it within redox-regulated pathways of cell survival and death and opens new therapeutic perspectives

    Insight into the mechanism of ferroptosis inhibition by ferrostatin-1

    Get PDF
    Ferroptosis is a form of cell death primed by iron and lipid hydroperoxides and prevented by GPx4. Ferrostatin-1 (fer-1) inhibits ferroptosis much more efficiently than phenolic antioxidants. Previous studies on the antioxidant efficiency of fer-1 adopted kinetic tests where a diazo compound generates the hydroperoxyl radical scavenged by the antioxidant. However, this reaction, accounting for a chain breaking effect, is only minimally useful for the description of the inhibition of ferrous iron and lipid hydroperoxide dependent peroxidation. Scavenging lipid hydroperoxyl radicals, indeed, generates lipid hydroperoxides from which ferrous iron initiates a new peroxidative chain reaction. We show that when fer-1 inhibits peroxidation, initiated by iron and traces of lipid hydroperoxides in liposomes, the pattern of oxidized species produced from traces of pre-existing hydroperoxides is practically identical to that observed following exhaustive peroxidation in the absence of the antioxidant. This supported the notion that the anti-ferroptotic activity of fer-1 is actually due to the scavenging of initiating alkoxyl radicals produced, together with other rearrangement products, by ferrous iron from lipid hydroperoxides. Notably, fer-1 is not consumed while inhibiting iron dependent lipid peroxidation. The emerging concept is that it is ferrous iron itself that reduces fer-1 radical. This was supported by electroanalytical evidence that fer-1 forms a complex with iron and further confirmed in cells by fluorescence of calcein, indicating a decrease of labile iron in the presence of fer-1. The notion of such as pseudo-catalytic cycle of the ferrostatin-iron complex was also investigated by means of quantum mechanics calculations, which confirmed the reduction of an alkoxyl radical model by fer-1 and the reduction of fer-1 radical by ferrous iron. In summary, GPx4 and fer-1 in the presence of ferrous iron, produces, by distinct mechanism, the most relevant anti-ferroptotic effect, i.e the disappearance of initiating lipid hydroperoxides

    KDM2A and KDM3B as Potential Targets for the Rescue of F508del-CFTR

    No full text
    Cystic fibrosis (CF) is caused by mutations in the gene encoding of the cystic fibrosis transmembrane conductance regulator (CFTR), an anion-selective plasma membrane channel that mainly regulates chloride transport in a variety of epithelia. More than 2000 mutations, most of which presumed to be disease-relevant, have been identified in the CFTR gene. The single CFTR mutation F508del (deletion of phenylalanine in position 508) is present in about 90% of global CF patients in at least one allele. F508del is responsible for the defective folding and processing of CFTR, failing to traffic to the plasma membrane and undergoing premature degradation via the ubiquitin-proteasome system. CFTR is subjected to different post-translational modifications (PTMs), and the possibility to modulate these PTMs has been suggested as a potential therapeutic strategy for the functional recovery of the disease-associated mutants. Recently, the PTM mapping of CFTR has identified some lysine residues that may undergo methylation or ubiquitination, suggesting a competition between these two PTMs. Our work hypothesis moves from the idea that favors methylation over ubiquitination, e.g., inhibiting demethylation could be a successful strategy for preventing the premature degradation of unstable CFTR mutants. Here, by using a siRNA library against all the human demethylases, we identified the enzymes whose downregulation increases F508del-CFTR stability and channel function. Our results show that KDM2A and KDM3B downregulation increases the stability of F508del-CFTR and boosts the functional rescue of the channel induced by CFTR correctors

    Effect of mercury on selenium utilization and selenoperoxidase activity in LNCaP cells

    No full text
    Abstract Formation of stable complexes with protein thiols is the best-known mechanism of mercury toxicity. However, the solubility product of Hg(2+) with sulfides, although very low, is higher than that with selenides, suggesting that the fully reduced form of selenium might also be a relevant target for Hg(2+). In cells, selenide is the suggested intermediate for selenoprotein biosynthesis and selenoenzymes, in turn, contain reduced selenium as the catalytic moiety. Thus, inhibition of biological functions of selenium could be seen as a different mechanism of Hg(2+) toxicity. To address this issue, we investigated selenoperoxidase (SeGPx) activity in LNCaP cells exposed to HgCl(2). Cells growing in standard medium express a low GPx activity, which increases on addition of selenium donors such as selenite, selenomethionine, or methyl-Se-cysteine. HgCl(2) added to the medium has different effects depending on the type of Se donor. A progressive decrease of SeGPx activity is observed in cells grown in standard medium exposed to HgCl(2), while coadministration of suprastoichiometric amounts of HgCl(2) prevents the increase of SeGPx activity only when selenite, but not selenomethionine or methyl-Se-cysteine, is the selenium source. From this evidence we conclude that HgCl(2): (a) does not inhibit directly SeGPxs, as confirmed on isolated enzymes; (b) does not interfere with the intermediates of the metabolic pathway of selenoprotein synthesis; and (c) decreases the bioavailability of selenium only when ionic complexes can be formed

    Glutathione Peroxidase-4

    No full text
    Within the family of glutathione peroxidases (GPxs), GPx-4 is the sole monomeric enzyme that contains Sec at the active site. Phylogenetically, it is closer to the Cys containing homologues (CysGPx) of invertebrata and vertebrata than to the tetrameric GPxs of vertebrata containing Sec. Nonetheless, the catalytic site is fully conserved in the whole family, suggesting a similar reactivity. As the tetrameric homologues, GPx-4 accepts GSH in the reductive steps of the catalytic cycle, while a redoxin is the preferred reducing substrate of the invertebrata CysGPxs. GPx-4 is also competent for oxidizing a quite heterogeneous series of thiol substrates. Reduction of complex membrane phospholipid and cholesterol hydroperoxides in cooperation with vitamin E accounts for the inhibition of lipid peroxidation by GPx-4. By no means, however, GPx-4 can be seen as just an antioxidant enzyme. Indeed reduction of lipid hydroperoxides accounts for the anti-apoptotic and anti-inflammatory effect of GPx-4 activity, and oxidation of specific protein thiols is its peculiar function in the late phase of spermatogenesis. Whether this reaction is relevant in other biochemical pathways, where a redox-switch drives a functional shift in specific proteins, remains as an open and challenging option. In this chapter, the enzymology of GPx-4 will be reviewed focusing on the two best-characterized aspects: i) inhibition of lipid peroxidation, and ii) oxidation of specific protein motifs. We refer to other chapters in this book for insights contributed by inverse genetic studies and for the general aspects of selenium catalysis in peroxidases

    Dependence of HSP27 cellular level on protein kinase CK2 discloses novel therapeutic strategies

    No full text
    Background: HSP27 plays a role in various diseases, including neurodegenerative diseases, ischemia, and atherosclerosis. It is particularly important in the regulation of the development, progression and metastasis of cancer as well as cell apoptosis and drug resistance. However, the absence of an ATP binding domain, that is, instead, present in other HSPs such as HSP90 and HSP70, hampers the development of small molecules as inhibitors of HSP27. Methods: Knockout cell lines generated by Crispr/Cas9 gene editing tool, specific kinase inhibitors and siRNA transfections were exploited to demonstrate that the expression of HSP27 is dependent on the integrity/activity of protein kinase CK2 holoenzyme. The interaction between these proteins has been confirmed by co-immunoprecipitation, confocal immunofluorescence microscopy, and by density gradient separation of protein complexes. Finally, using a proliferation assay this study demonstrates the potential efficacy of a combinatory therapy of heath shock and CK2 inhibitors in cancer treatment. Results: Our data demonstrate that CK2 is able to regulate HSP27 turnover by affecting the expression of its ubiquitin ligase SMURF2 (Smad ubiquitination regulatory factor 2). Moreover, for the first time we show an increased sensitivity of CK2-inhibited tumour cells to hyperthermia treatment. Conclusion: Being HSP27 involved in several pathological conditions, including protein conformational diseases (i.e Cystic Fibrosis) and cancer, the need of drugs to modulate its activity is growing and CK2-targeting could represent a new strategy to reduce cellular HSP27 level. General significance: This study identifies CK2 as a molecular target to control HSP27 cellular expression
    corecore