Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters











Database
Language
Publication year range
1.
J Med Chem ; 67(18): 16107-16127, 2024 Sep 26.
Article in English | MEDLINE | ID: mdl-39264826

ABSTRACT

Ferroptosis is a recently discovered cell death mechanism triggered by iron-dependent elevation of reactive oxygen species leading to lipid membrane peroxidation. We previously reported the development of a new class of ferroptosis inducers referred to as CETZOLEs with CC50 values in the low micromolar range. Structure-activity relationship study of these compounds led to the development of more potent analogs with CC50 values in the nanomolar range. Cells exposed to these compounds displayed the hallmarks of ferroptosis including cell death through ROS accumulation. Cancer cells were found to be more sensitive to these compounds than normal cells. Proteomic studies using covalent and affinity probes led to the identification of cystathionine ß-synthase, peroxiredoxins, ADP/ATP carriers, and glucose dehydrogenase as enriched proteins. The binding of CETZOLEs to these proteins as well as GPX4 was validated by Western blotting. This group of proteins is known to be associated with cellular antioxidant pathways.


Subject(s)
Ferroptosis , Ferroptosis/drug effects , Humans , Structure-Activity Relationship , Reactive Oxygen Species/metabolism , Cell Line, Tumor , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Molecular Probes/chemistry , Molecular Probes/pharmacology
2.
Res Sq ; 2023 Jan 30.
Article in English | MEDLINE | ID: mdl-36778475

ABSTRACT

Tumor suppressor RB binds to E2F family proteins and modulates cell cycle progression. Cyclin dependent kinases (CDK) regulate the interaction of RB/E2F by phosphorylating RB. Previously, we have revealed that CDK2, RB and E2F inhibit ferroptosis. Ferroptosis is a non-apoptotic, iron-dependent form of cell death characterized by toxic lipid peroxidation. Here we provide evidence that CDK2 suppresses ferroptosis through phosphorylation of RB. We approach this question by overexpressing WT-RB or a mutant RB that cannot be phosphorylated by CDKs (RBΔCDK) along with CDK2/cyclinE followed by analysis of ferroptosis. We also observed that E2F1 regulates of both pro and anti-ferroptotic proteins including ALOX5, MYC SLC7A11, ATF4, and GPX4 and finally renders a net inhibitory role in ferroptosis. Interestingly, we also found a cell type dependent compensatory effect of E2F3 upon E2F1 depletion. This compensatory effect resulted in no change of ferroptotic target genes after E2F1 knock down in an osteosarcoma cell line. Taken together, our study reveals that cancer cells protect themselves from ferroptosis through cell cycle regulatory proteins.

3.
J Med Chem ; 65(21): 14764-14791, 2022 11 10.
Article in English | MEDLINE | ID: mdl-36306372

ABSTRACT

HDAC inhibitors are an attractive class of cytotoxic agents for the design of hybrid molecules. Several HDAC hybrids have emerged over the years, but none combines HDAC inhibition with ferroptosis, a combination which is being extensively studied because it leads to enhanced cytotoxicity and attenuated neuronal toxicity. We combined the pharmacophores of SAHA and CETZOLE molecules to design the first-in-class dual mechanism hybrid molecules, which induce ferroptosis and inhibit HDAC proteins. The involvement of both mechanisms in cytotoxicity was confirmed by a series of biological assays. The cytotoxic effects were evaluated in a series of cancer and neuronal cell lines. Analogue HY-1 demonstrated the best cytotoxic profile with GI50 values as low as 20 nM. Although the increase in activity of the hybrids over the combinations is modest in cellular systems, they have the potential advantage of homogeneous spatiotemporal distribution in in vivo systems.


Subject(s)
Antineoplastic Agents , Ferroptosis , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Histone Deacetylases/metabolism , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation
4.
J Med Chem ; 65(17): 11788-11817, 2022 09 08.
Article in English | MEDLINE | ID: mdl-35984756

ABSTRACT

Once considered potential liabilities, the modern era witnesses a renaissance of interest in covalent inhibitors in drug discovery. The available toolbox of electrophilic warheads is limited by constraints on tuning reactivity and selectivity. Following our work on a class of ferroptotic agents termed CETZOLEs, we discovered new tunable heterocyclic electrophiles which are capable of inducing ferroptosis. The biological evaluation demonstrated that thiazoles with an alkyne electrophile at the 2-position selectively induce ferroptosis with high potency. Density functional theory calculations and NMR kinetic studies demonstrated the ability of our heterocycles to undergo thiol addition, an apparent prerequisite for cytotoxicity. Chemoproteomic analysis indicated several potential targets, the most prominent among them being GPX4 protein. These results were further validated by western blot analysis and the cellular thermal shift assay. Incorporation of these heterocycles into appropriate pharmacophores generated highly cytotoxic agents such as the analogue BCP-T.A, with low nM IC50 values in ferroptosis-sensitive cell lines.


Subject(s)
Cysteine , Ferroptosis , Cysteine/chemistry , Drug Discovery , Kinetics , Sulfhydryl Compounds/chemistry
5.
J Biol Chem ; 297(6): 101365, 2021 12.
Article in English | MEDLINE | ID: mdl-34728216

ABSTRACT

p53 is a well-established critical cell cycle regulator. By inducing transcription of the gene encoding p21, p53 inhibits cyclin-dependent kinase (CDK)-mediated phosphorylation of cell cycle inhibitor retinoblastoma (RB) proteins. Phosphorylation of RB releases E2F transcription factor proteins that transactivate cell cycle-promoting genes. Here, we sought to uncover the contribution of p53, p21, CDK, RB, and E2F to the regulation of ferroptosis, an oxidative form of cell death. Our studies have uncovered unexpected complexity in this regulation. First, we showed that elevated levels of p53 enhance ferroptosis in multiple inducible and isogenic systems. On the other hand, we found that p21 suppresses ferroptosis. Elevation of CDK activity also suppressed ferroptosis under conditions where p21 suppressed ferroptosis, suggesting that the impact of p21 must extend beyond CDK inhibition. Furthermore, we showed that overexpression of E2F suppresses ferroptosis in part via a p21-dependent mechanism, consistent with reports that this transcription factor can induce transcription of p21. Finally, deletion of RB genes enhanced ferroptosis. Taken together, these results show that signals affecting ferroptotic sensitivity emanate from multiple points within the p53 tumor suppressor pathway.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinases/metabolism , E2F1 Transcription Factor/metabolism , Ferroptosis/physiology , Retinoblastoma Protein/metabolism , Tumor Suppressor Protein p53/physiology , Animals , Cell Line, Tumor , Cells, Cultured , Humans , Mice , Mice, Knockout , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
6.
J Cell Biochem ; 122(3-4): 413-424, 2021 04.
Article in English | MEDLINE | ID: mdl-33377232

ABSTRACT

Ferroptosis is a form of iron-dependent cell death characterized by elevated lipid peroxides and reactive oxygen species (ROS). Glutathione (GSH) plays an essential role in scavenging ROS to maintain cell viability and acts as a cofactor of GSH peroxidase 4 (GPX4) that protects lipids from oxidation. We have previously described a novel class of small molecules that induce ferroptosis in certain types of cancer cells. These compounds induce ferroptosis by blocking the uptake of cystine required for GSH synthesis. Even though ferroptosis is a well-established form of cell death, signaling pathways that modulate this process are not known. Therefore, we used a panel of growth factors/kinase inhibitors to test effects on ferroptosis induced by our lead compound. We discovered that BMS536924, a dual inhibitor of insulin-like growth and insulin receptors, is a potent inhibitor of ferroptosis. Further investigation indicated that the anti-ferroptotic activity of BMS536924 does not lie in its ability to inhibit insulin signal transduction. Instead, we provide evidence that BMS536924 binds iron, an essential cofactor in ferroptosis. Our results suggest caution in interpreting the effects of BMS536924 in investigations of insulin signaling and uncover a novel ferroptosis inhibitor.


Subject(s)
Benzimidazoles/pharmacology , Ferroptosis/drug effects , Pyridones/pharmacology , Cell Survival/drug effects , Cystine/metabolism , Glutathione/metabolism , Humans , Lipid Peroxidation/drug effects , Reactive Oxygen Species/metabolism , Receptor, Insulin/metabolism
7.
Sci Rep ; 9(1): 5926, 2019 04 11.
Article in English | MEDLINE | ID: mdl-30976078

ABSTRACT

Effective management of advanced cancer requires systemic treatment including small molecules that target unique features of aggressive tumor cells. At the same time, tumors are heterogeneous and current evidence suggests that a subpopulation of tumor cells, called tumor initiating or cancer stem cells, are responsible for metastatic dissemination, tumor relapse and possibly drug resistance. Classical apoptotic drugs are less effective against this critical subpopulation. In the course of generating a library of open-chain epothilones, we discovered a new class of small molecule anticancer agents that has no effect on tubulin but instead kills selected cancer cell lines by harnessing reactive oxygen species to induce ferroptosis. Interestingly, we find that drug sensitivity is highest in tumor cells with a mesenchymal phenotype. Furthermore, these compounds showed enhanced toxicity towards mesenchymal breast cancer populations with cancer stem cell properties in vitro. In summary, we have identified a new class of small molecule ferroptotic agents that warrant further investigation.


Subject(s)
Antineoplastic Agents/pharmacology , Ferroptosis , Neoplasms/drug therapy , Neoplastic Stem Cells/drug effects , Reactive Oxygen Species/metabolism , Small Molecule Libraries/pharmacology , Antineoplastic Agents/chemistry , Cell Proliferation , Humans , Mesoderm/drug effects , Mesoderm/pathology , Neoplasms/pathology , Neoplastic Stem Cells/pathology , Small Molecule Libraries/chemistry , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL