Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Cell ; 168(5): 878-889.e29, 2017 02 23.
Article in English | MEDLINE | ID: mdl-28235199

ABSTRACT

Design of small molecules that disrupt protein-protein interactions, including the interaction of RAS proteins and their effectors, may provide chemical probes and therapeutic agents. We describe here the synthesis and testing of potential small-molecule pan-RAS ligands, which were designed to interact with adjacent sites on the surface of oncogenic KRAS. One compound, termed 3144, was found to bind to RAS proteins using microscale thermophoresis, nuclear magnetic resonance spectroscopy, and isothermal titration calorimetry and to exhibit lethality in cells partially dependent on expression of RAS proteins. This compound was metabolically stable in liver microsomes and displayed anti-tumor activity in xenograft mouse cancer models. These findings suggest that pan-RAS inhibition may be an effective therapeutic strategy for some cancers and that structure-based design of small molecules targeting multiple adjacent sites to create multivalent inhibitors may be effective for some proteins.


Subject(s)
Antineoplastic Agents/pharmacology , Molecular Targeted Therapy , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras)/chemistry , Animals , Antineoplastic Agents/chemistry , Calorimetry , Cell Line , Fibroblasts/metabolism , Heterografts , Humans , Mice , Neoplasm Transplantation , Neoplasms/drug therapy , Pancreatic Neoplasms/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Signal Transduction , Small Molecule Libraries
2.
Cell ; 162(2): 441-451, 2015 Jul 16.
Article in English | MEDLINE | ID: mdl-26186195

ABSTRACT

Genome-wide identification of the mechanism of action (MoA) of small-molecule compounds characterizing their targets, effectors, and activity modulators represents a highly relevant yet elusive goal, with critical implications for assessment of compound efficacy and toxicity. Current approaches are labor intensive and mostly limited to elucidating high-affinity binding target proteins. We introduce a regulatory network-based approach that elucidates genome-wide MoA proteins based on the assessment of the global dysregulation of their molecular interactions following compound perturbation. Analysis of cellular perturbation profiles identified established MoA proteins for 70% of the tested compounds and elucidated novel proteins that were experimentally validated. Finally, unknown-MoA compound analysis revealed altretamine, an anticancer drug, as an inhibitor of glutathione peroxidase 4 lipid repair activity, which was experimentally confirmed, thus revealing unexpected similarity to the activity of sulfasalazine. This suggests that regulatory network analysis can provide valuable mechanistic insight into the elucidation of small-molecule MoA and compound similarity.


Subject(s)
Algorithms , Antineoplastic Agents/pharmacology , Molecular Targeted Therapy , Antineoplastic Agents/chemistry , Epistasis, Genetic , Genome-Wide Association Study , Neoplasms/drug therapy , Small Molecule Libraries
3.
Cell ; 156(1-2): 317-331, 2014 Jan 16.
Article in English | MEDLINE | ID: mdl-24439385

ABSTRACT

Ferroptosis is a form of nonapoptotic cell death for which key regulators remain unknown. We sought a common mediator for the lethality of 12 ferroptosis-inducing small molecules. We used targeted metabolomic profiling to discover that depletion of glutathione causes inactivation of glutathione peroxidases (GPXs) in response to one class of compounds and a chemoproteomics strategy to discover that GPX4 is directly inhibited by a second class of compounds. GPX4 overexpression and knockdown modulated the lethality of 12 ferroptosis inducers, but not of 11 compounds with other lethal mechanisms. In addition, two representative ferroptosis inducers prevented tumor growth in xenograft mouse tumor models. Sensitivity profiling in 177 cancer cell lines revealed that diffuse large B cell lymphomas and renal cell carcinomas are particularly susceptible to GPX4-regulated ferroptosis. Thus, GPX4 is an essential regulator of ferroptotic cancer cell death.


Subject(s)
Carbolines/pharmacology , Cell Death/drug effects , Glutathione Peroxidase/antagonists & inhibitors , Piperazines/pharmacology , Animals , Carcinoma, Renal Cell/drug therapy , Cell Line, Tumor , Gene Knockdown Techniques , Glutathione/metabolism , Glutathione Peroxidase/genetics , Glutathione Peroxidase/metabolism , Heterografts , Humans , Lymphoma, B-Cell/drug therapy , Mice , Neoplasm Transplantation , Neoplasms/drug therapy , Phospholipid Hydroperoxide Glutathione Peroxidase
4.
Cell ; 149(5): 1060-72, 2012 May 25.
Article in English | MEDLINE | ID: mdl-22632970

ABSTRACT

Nonapoptotic forms of cell death may facilitate the selective elimination of some tumor cells or be activated in specific pathological states. The oncogenic RAS-selective lethal small molecule erastin triggers a unique iron-dependent form of nonapoptotic cell death that we term ferroptosis. Ferroptosis is dependent upon intracellular iron, but not other metals, and is morphologically, biochemically, and genetically distinct from apoptosis, necrosis, and autophagy. We identify the small molecule ferrostatin-1 as a potent inhibitor of ferroptosis in cancer cells and glutamate-induced cell death in organotypic rat brain slices, suggesting similarities between these two processes. Indeed, erastin, like glutamate, inhibits cystine uptake by the cystine/glutamate antiporter (system x(c)(-)), creating a void in the antioxidant defenses of the cell and ultimately leading to iron-dependent, oxidative death. Thus, activation of ferroptosis results in the nonapoptotic destruction of certain cancer cells, whereas inhibition of this process may protect organisms from neurodegeneration.


Subject(s)
Cell Death , Iron/metabolism , Animals , Cell Death/drug effects , Cyclohexylamines/pharmacology , Fibroblasts/cytology , Fibroblasts/metabolism , Glutamic Acid/metabolism , Hippocampus/cytology , Humans , In Vitro Techniques , Lipid Metabolism , Neoplasms/pathology , Phenylenediamines/pharmacology , Piperazines/metabolism , Rats , Reactive Oxygen Species/metabolism
5.
Nature ; 547(7664): 453-457, 2017 07 27.
Article in English | MEDLINE | ID: mdl-28678785

ABSTRACT

Plasticity of the cell state has been proposed to drive resistance to multiple classes of cancer therapies, thereby limiting their effectiveness. A high-mesenchymal cell state observed in human tumours and cancer cell lines has been associated with resistance to multiple treatment modalities across diverse cancer lineages, but the mechanistic underpinning for this state has remained incompletely understood. Here we molecularly characterize this therapy-resistant high-mesenchymal cell state in human cancer cell lines and organoids and show that it depends on a druggable lipid-peroxidase pathway that protects against ferroptosis, a non-apoptotic form of cell death induced by the build-up of toxic lipid peroxides. We show that this cell state is characterized by activity of enzymes that promote the synthesis of polyunsaturated lipids. These lipids are the substrates for lipid peroxidation by lipoxygenase enzymes. This lipid metabolism creates a dependency on pathways converging on the phospholipid glutathione peroxidase (GPX4), a selenocysteine-containing enzyme that dissipates lipid peroxides and thereby prevents the iron-mediated reactions of peroxides that induce ferroptotic cell death. Dependency on GPX4 was found to exist across diverse therapy-resistant states characterized by high expression of ZEB1, including epithelial-mesenchymal transition in epithelial-derived carcinomas, TGFß-mediated therapy-resistance in melanoma, treatment-induced neuroendocrine transdifferentiation in prostate cancer, and sarcomas, which are fixed in a mesenchymal state owing to their cells of origin. We identify vulnerability to ferroptic cell death induced by inhibition of a lipid peroxidase pathway as a feature of therapy-resistant cancer cells across diverse mesenchymal cell-state contexts.


Subject(s)
Glutathione Peroxidase/metabolism , Lipid Peroxidation/drug effects , Neoplasms/drug therapy , Neoplasms/enzymology , Cadherins/metabolism , Cell Death , Cell Line, Tumor , Cell Lineage , Cell Transdifferentiation , Drug Resistance, Neoplasm/genetics , Epithelial-Mesenchymal Transition , Humans , Iron/metabolism , Lipid Peroxides/metabolism , Male , Melanoma/drug therapy , Melanoma/enzymology , Melanoma/metabolism , Melanoma/pathology , Mesoderm/drug effects , Mesoderm/enzymology , Mesoderm/metabolism , Mesoderm/pathology , Neoplasms/genetics , Neoplasms/pathology , Phospholipid Hydroperoxide Glutathione Peroxidase , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Proteomics , Proto-Oncogene Proteins B-raf/genetics , Reproducibility of Results , Zinc Finger E-box-Binding Homeobox 1/genetics
6.
Proc Natl Acad Sci U S A ; 113(34): E4966-75, 2016 08 23.
Article in English | MEDLINE | ID: mdl-27506793

ABSTRACT

Ferroptosis is form of regulated nonapoptotic cell death that is involved in diverse disease contexts. Small molecules that inhibit glutathione peroxidase 4 (GPX4), a phospholipid peroxidase, cause lethal accumulation of lipid peroxides and induce ferroptotic cell death. Although ferroptosis has been suggested to involve accumulation of reactive oxygen species (ROS) in lipid environments, the mediators and substrates of ROS generation and the pharmacological mechanism of GPX4 inhibition that generates ROS in lipid environments are unknown. We report here the mechanism of lipid peroxidation during ferroptosis, which involves phosphorylase kinase G2 (PHKG2) regulation of iron availability to lipoxygenase enzymes, which in turn drive ferroptosis through peroxidation of polyunsaturated fatty acids (PUFAs) at the bis-allylic position; indeed, pretreating cells with PUFAs containing the heavy hydrogen isotope deuterium at the site of peroxidation (D-PUFA) prevented PUFA oxidation and blocked ferroptosis. We further found that ferroptosis inducers inhibit GPX4 by covalently targeting the active site selenocysteine, leading to accumulation of PUFA hydroperoxides. In summary, we found that PUFA oxidation by lipoxygenases via a PHKG2-dependent iron pool is necessary for ferroptosis and that the covalent inhibition of the catalytic selenocysteine in Gpx4 prevents elimination of PUFA hydroperoxides; these findings suggest new strategies for controlling ferroptosis in diverse contexts.


Subject(s)
Cell Death/drug effects , Epithelial Cells/metabolism , Fatty Acids, Unsaturated/metabolism , Glutathione Peroxidase/metabolism , Lipoxygenases/metabolism , Phosphorylase Kinase/metabolism , Catalytic Domain , Cell Death/genetics , Cell Line, Tumor , Deuterium , Epithelial Cells/cytology , Epithelial Cells/drug effects , Fatty Acids, Unsaturated/pharmacology , Gene Expression Regulation , Glutathione Peroxidase/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Iron/metabolism , Lipid Peroxidation/drug effects , Lipid Peroxides/biosynthesis , Lipoxygenases/genetics , Phospholipid Hydroperoxide Glutathione Peroxidase , Phosphorylase Kinase/genetics , Protein Transport , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Selenocysteine/metabolism , Signal Transduction
7.
Nat Chem Biol ; 12(7): 497-503, 2016 07.
Article in English | MEDLINE | ID: mdl-27159577

ABSTRACT

Apoptosis is one type of programmed cell death. Increasingly, non-apoptotic cell death is recognized as being genetically controlled, or 'regulated'. However, the full extent and diversity of alternative cell death mechanisms remain uncharted. Here we surveyed the landscape of pharmacologically accessible cell death mechanisms. In an examination of 56 caspase-independent lethal compounds, modulatory profiling showed that 10 compounds induced three different types of regulated non-apoptotic cell death. Optimization of one of those ten resulted in the discovery of FIN56, a specific inducer of ferroptosis. Ferroptosis has been found to occur when the lipid-repair enzyme GPX4 is inhibited. FIN56 promoted degradation of GPX4. FIN56 also bound to and activated squalene synthase, an enzyme involved in isoprenoid biosynthesis, independent of GPX4 degradation. These discoveries show that dysregulation of lipid metabolism is associated with ferroptosis. This systematic approach is a means to discover and characterize novel cell death phenotypes.


Subject(s)
Apoptosis/drug effects , Iron/metabolism , Oximes/pharmacology , Sulfonamides/pharmacology , Dose-Response Relationship, Drug , Humans , Lipid Metabolism/drug effects , Oximes/chemistry , Oximes/metabolism , Structure-Activity Relationship , Sulfonamides/chemistry , Sulfonamides/metabolism
8.
Bioorg Med Chem Lett ; 25(21): 4787-4792, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26231156

ABSTRACT

Introducing a reactive carbonyl to a scaffold that does not otherwise have an electrophilic functionality to create a reversible covalent inhibitor is a potentially useful strategy for enhancing compound potency. However, aldehydes are metabolically unstable, which precludes the use of this strategy for compounds to be tested in animal models or in human clinical studies. To overcome this limitation, we designed ketone-based functionalities capable of forming reversible covalent adducts, while displaying high metabolic stability, and imparting improved water solubility to their pendant scaffold. We tested this strategy on the ferroptosis inducer and experimental therapeutic erastin, and observed substantial increases in compound potency. In particular, a new carbonyl erastin analog, termed IKE, displayed improved potency, solubility and metabolic stability, thus representing an ideal candidate for future in vivo cancer therapeutic applications.


Subject(s)
Ketones/chemistry , Piperazines/chemistry , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Water/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Humans , Ketones/metabolism , Molecular Structure , Piperazines/chemical synthesis , Piperazines/pharmacology , Small Molecule Libraries/chemical synthesis , Solubility , Structure-Activity Relationship
9.
Nat Cell Biol ; 2024 Feb 29.
Article in English | MEDLINE | ID: mdl-38424270

ABSTRACT

Ferroptosis, an intricately regulated form of cell death characterized by uncontrolled lipid peroxidation, has garnered substantial interest since this term was first coined in 2012. Recent years have witnessed remarkable progress in elucidating the detailed molecular mechanisms that govern ferroptosis induction and defence, with particular emphasis on the roles of heterogeneity and plasticity. In this Review, we discuss the molecular ecosystem of ferroptosis, with implications that may inform and enable safe and effective therapeutic strategies across a broad spectrum of diseases.

10.
Nature ; 447(7146): 864-8, 2007 Jun 14.
Article in English | MEDLINE | ID: mdl-17568748

ABSTRACT

Therapeutics that discriminate between the genetic makeup of normal cells and tumour cells are valuable for treating and understanding cancer. Small molecules with oncogene-selective lethality may reveal novel functions of oncoproteins and enable the creation of more selective drugs. Here we describe the mechanism of action of the selective anti-tumour agent erastin, involving the RAS-RAF-MEK signalling pathway functioning in cell proliferation, differentiation and survival. Erastin exhibits greater lethality in human tumour cells harbouring mutations in the oncogenes HRAS, KRAS or BRAF. Using affinity purification and mass spectrometry, we discovered that erastin acts through mitochondrial voltage-dependent anion channels (VDACs)--a novel target for anti-cancer drugs. We show that erastin treatment of cells harbouring oncogenic RAS causes the appearance of oxidative species and subsequent death through an oxidative, non-apoptotic mechanism. RNA-interference-mediated knockdown of VDAC2 or VDAC3 caused resistance to erastin, implicating these two VDAC isoforms in the mechanism of action of erastin. Moreover, using purified mitochondria expressing a single VDAC isoform, we found that erastin alters the permeability of the outer mitochondrial membrane. Finally, using a radiolabelled analogue and a filter-binding assay, we show that erastin binds directly to VDAC2. These results demonstrate that ligands to VDAC proteins can induce non-apoptotic cell death selectively in some tumour cells harbouring activating mutations in the RAS-RAF-MEK pathway.


Subject(s)
Mitogen-Activated Protein Kinase Kinases/metabolism , Oxidative Stress/drug effects , Piperazines/pharmacology , Signal Transduction/drug effects , Voltage-Dependent Anion Channel 2/metabolism , raf Kinases/metabolism , ras Proteins/metabolism , Cell Death/drug effects , Cell Line, Tumor , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Ion Channel Gating/drug effects , Phosphorylation/drug effects , Piperazines/toxicity , Sensitivity and Specificity
11.
FEBS Open Bio ; 13(3): 419-433, 2023 03.
Article in English | MEDLINE | ID: mdl-36595221

ABSTRACT

Ferroptosis is a regulated form of cell death driven by the lethal accumulation of lipid peroxides in cell membranes. Several regulators of ferroptosis have been identified using cancer cell lines. However, the cellular pathways of ferroptosis in neurons remain poorly characterized. In this study, we used a mouse embryonic stem cell-derived motor neuron model to investigate how motor neurons respond to ferroptosis inducers. Pharmacological and genetic inhibition of glutathione peroxidase 4 (GPx4) induced ferroptosis in motor neurons, while system xc - inhibition by erastin had no effect. RNA-seq analysis showed that the expression levels of several genes were altered during RSL3-induced ferroptosis. Subsequent bioinformatic analysis revealed alterations in several biological pathways during ferroptosis, including synaptogenesis and calcium signaling. Finally, we found that edaravone, an FDA-approved drug for treating amyotrophic lateral sclerosis (ALS) disease, rescued motor neurons from RSL3-induced ferroptosis. Our data highlight the crucial role of GPx4 in ferroptosis regulation and demonstrate that stem cell-derived motor neuron culture is a valuable model to study ferroptosis at the single-cell level in a neuronal context.


Subject(s)
Ferroptosis , Animals , Mice , Glutathione Peroxidase/metabolism , Mouse Embryonic Stem Cells/metabolism , Motor Neurons/metabolism , Cell Death
12.
Bioorg Med Chem Lett ; 22(4): 1822-6, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22297109

ABSTRACT

Synthetic lethal screening is a chemical biology approach to identify small molecules that selectively kill oncogene-expressing cell lines with the goal of identifying pathways that provide specific targets against cancer cells. We performed a high-throughput screen of 303,282 compounds from the National Institutes of Health-Molecular Libraries Small Molecule Repository (NIH-MLSMR) against immortalized BJ fibroblasts expressing HRAS(G12V) followed by a counterscreen of lethal compounds in a series of isogenic cells lacking the HRAS(G12V) oncogene. This effort led to the identification of two novel molecular probes (PubChem CID 3689413, ML162 and CID 49766530, ML210) with nanomolar potencies and 4-23-fold selectivities, which can potentially be used for identifying oncogene-specific pathways and targets in cancer cells.


Subject(s)
Apoptosis/drug effects , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Animals , Cell Line , Cell Line, Tumor , Drug Screening Assays, Antitumor , Fibroblasts , Humans , Inhibitory Concentration 50 , Molecular Structure , Mutation , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Rats , Structure-Activity Relationship , Thiophenes/chemistry , Thiophenes/pharmacology
13.
Methods Mol Biol ; 2108: 125-130, 2020.
Article in English | MEDLINE | ID: mdl-31939176

ABSTRACT

Ferroptosis is a distinctive form of regulated cell death that is driven by lethal accumulation of lipid peroxides in plasma membranes. Failure to control ferroptosis has been implicated in multiple pathological conditions including cancer development, neurodegeneration, renal injury, ischemia/reperfusion injury, and T-cell immunity. Here we describe a method to detect ferroptosis by determining the amount of lipid peroxides in cellular membranes using BODIPY-C11 probe and flow cytometry. Putative role of ferroptosis in immune modulatory cells can be determined using the same method.


Subject(s)
Biological Assay/methods , Ferroptosis , Apoptosis , Biomarkers , Cell Line, Tumor , Flow Cytometry/methods , Humans , Lipid Metabolism , Lipid Peroxidation , Piperazines/pharmacology , Reactive Oxygen Species/metabolism
14.
Chem Biol ; 15(3): 234-45, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18355723

ABSTRACT

We screened small molecules to identify two compounds, which we named RSL3 and RSL5, that have increased lethality in the presence of oncogenic RAS. Counter screening with biologically active compounds defined aspects of the mechanism of action for RSL3 and RSL5, such as a nonapoptotic, MEK-dependent, and iron-dependent oxidative cell death. Erastin, a previously reported compound with RAS-selective lethality, showed similar properties. RNA interference experiments targeting voltage-dependent anion channel 3 (VDAC3), a target of erastin, demonstrated that RSL5 is a scaffold that acts through VDACs to activate the observed pathway. RSL3 activated a similar death mechanism but in a VDAC-independent manner. We found that cells transformed with oncogenic RAS have increased iron content relative to their normal cell counterparts through upregulation of transferrin receptor 1 and downregulation of ferritin heavy chain 1 and ferritin light chain.


Subject(s)
Iron/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Small Molecule Libraries/pharmacology , Apoptosis , Cell Death/drug effects , Cell Line, Tumor , Drug Evaluation, Preclinical , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mutation , Neoplasms/drug therapy , Piperazines/pharmacology , Substrate Specificity , Voltage-Dependent Anion Channels/metabolism
15.
FEBS Open Bio ; 9(4): 582-593, 2019 04.
Article in English | MEDLINE | ID: mdl-30984534

ABSTRACT

Ferroptosis is a form of regulated cell death that is driven by lethal accumulation of lipid peroxides upon inhibition of glutathione peroxidase 4 (GPx4). Deletion of the Gpx4 gene in mice revealed that neurons are sensitive to ferroptosis in vivo. However, few studies have been conducted on ferroptosis regulation in neurons. Here, we report that cells of a motor neuron-like cell line, NSC-34, became more sensitive to ferroptosis upon differentiation into a more motor neuron-like condition. We identified three factors that influence ferroptosis sensitivity under differentiation conditions: low serum antioxidants, decreased GPx4 protein amount, and inhibition of the transsulfuration pathway. Our results support the hypothesis that neurons, especially motor neurons, are sensitive to ferroptosis, and suggest that ferroptosis in a neuronal context should be investigated further to develop strategies for neuroprotection.


Subject(s)
Cell Death/physiology , Cell Differentiation/drug effects , Motor Neurons/physiology , Animals , Antioxidants/metabolism , Cell Line , Glutathione Peroxidase/metabolism , Mice , Phospholipid Hydroperoxide Glutathione Peroxidase
16.
Front Cell Dev Biol ; 11: 1179245, 2023.
Article in English | MEDLINE | ID: mdl-37274745
17.
Mol Cancer Ther ; 5(6): 1610-9, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16818521

ABSTRACT

We cloned the streptolysin O gene from the Streptococcus pyogenes genome and tested the possibility of using it as an anticancer reagent. Transient transfection of the streptolysin O gene efficiently killed 293T cells after 12 hours of transfection as determined by lactate dehydrogenase release and propidium iodide uptake. No caspase activity was observed and necrosis was prominent during streptolysin O-induced cell death. Biochemical analysis of streptolysin O protein revealed that the deletion of only 5 amino acids from the COOH-terminal region of streptolysin O, which is essential for cholesterol binding activity, abolished its cell-killing activity, whereas the NH2-terminal region was more resilient, i.e., up to 115 amino acids could be deleted without changing its cell-killing activity. We generated a streptolysin O-expressing adenovirus and injected it into human cervical cancer cell-derived tumors grown in a nude mouse model. Twenty-one days postinjection, the average size of tumors in the streptolysin O adenovirus-injected group was 29.3% of that of the control PBS-treated group. Our results show that the genes of pore-forming toxins, like streptolysin O protein, have the potential to establish a novel class of suicide gene therapeutic reagents.


Subject(s)
Genes, Transgenic, Suicide/physiology , Genetic Therapy , Streptococcus pyogenes/genetics , Streptolysins/genetics , Uterine Cervical Neoplasms/therapy , Adenoviridae/genetics , Animals , Apoptosis , Bacterial Proteins/genetics , Female , Gene Expression , Genetic Vectors , Green Fluorescent Proteins/metabolism , Humans , Male , Mice , Mice, Nude , Transfection , Tumor Cells, Cultured , Uterine Cervical Neoplasms/metabolism , Xenograft Model Antitumor Assays
18.
Trends Cell Biol ; 26(3): 165-176, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26653790

ABSTRACT

Ferroptosis is a regulated form of cell death driven by loss of activity of the lipid repair enzyme glutathione peroxidase 4 (GPX4) and subsequent accumulation of lipid-based reactive oxygen species (ROS), particularly lipid hydroperoxides. This form of iron-dependent cell death is genetically, biochemically, and morphologically distinct from other cell death modalities, including apoptosis, unregulated necrosis, and necroptosis. Ferroptosis is regulated by specific pathways and is involved in diverse biological contexts. Here we summarize the discovery of ferroptosis, the mechanism of ferroptosis regulation, and its increasingly appreciated relevance to both normal and pathological physiology.


Subject(s)
Lipid Peroxidation , Animals , Cell Death/physiology , Glutamic Acid/physiology , Glutathione Peroxidase/physiology , Humans , Iron/physiology , Neurons/physiology , Phospholipid Hydroperoxide Glutathione Peroxidase , Reactive Oxygen Species/metabolism
19.
Cell Rep ; 16(2): 545-558, 2016 07 12.
Article in English | MEDLINE | ID: mdl-27373155

ABSTRACT

Suboptimal axonal regeneration contributes to the consequences of nervous system trauma and neurodegenerative disease, but the intrinsic mechanisms that regulate axon growth remain unclear. We screened 50,400 small molecules for their ability to promote axon outgrowth on inhibitory substrata. The most potent hits were the statins, which stimulated growth of all mouse- and human-patient-derived neurons tested, both in vitro and in vivo, as did combined inhibition of the protein prenylation enzymes farnesyltransferase (PFT) and geranylgeranyl transferase I (PGGT-1). Compensatory sprouting of motor axons may delay clinical onset of amyotrophic lateral sclerosis (ALS). Accordingly, elevated levels of PGGT1B, which would be predicted to reduce sprouting, were found in motor neurons of early- versus late-onset ALS patients postmortem. The mevalonate-prenylation pathway therefore constitutes an endogenous brake on axonal growth, and its inhibition provides a potential therapeutic approach to accelerate neuronal regeneration in humans.


Subject(s)
Neurites/physiology , Protein Prenylation , Amyotrophic Lateral Sclerosis/pathology , Animals , Cell Enlargement , Cells, Cultured , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Mevalonic Acid/metabolism , Mice , Motor Neurons/physiology , Nerve Regeneration
20.
FEBS Lett ; 551(1-3): 29-36, 2003 Sep 11.
Article in English | MEDLINE | ID: mdl-12965200

ABSTRACT

Human Bfl-1 is an anti-apoptotic Bcl-2 family member. Here, we found that Bfl-1 was converted into a potent death-promoting protein by green fluorescent protein (GFP) fusion with its N-terminus. The transient expression of GFP-Bfl-1 induced cytochrome c release and triggered apoptosis in 293T cells, which depended on the mitochondrial localization of GFP-Bfl-1. Apoptosis induced by GFP-Bfl-1 was significantly blocked by the pan-caspase inhibitor carbobenzoxy-Val-Ala-Asp-fluoromethyl ketone, but was not blocked by either Bcl-xL or Bfl-1. Our findings provide a useful model for understanding the structural basis of Bcl-2 family proteins that act in an opposite way despite sharing structural similarity between anti-apoptotic and pro-apoptotic proteins.


Subject(s)
Apoptosis , Proto-Oncogene Proteins c-bcl-2/chemistry , Caspases/physiology , Cell Line , Cytochrome c Group/metabolism , Green Fluorescent Proteins , Humans , Luminescent Proteins/genetics , Minor Histocompatibility Antigens , Mitochondria/chemistry , Mitochondria/metabolism , Protein Structure, Tertiary , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Recombinant Fusion Proteins/analysis , Recombinant Fusion Proteins/antagonists & inhibitors , Recombinant Fusion Proteins/metabolism , bcl-X Protein
SELECTION OF CITATIONS
SEARCH DETAIL