Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Mol Cell ; 84(10): 1964-1979.e6, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38759628

ABSTRACT

The role of the mitochondrial electron transport chain (ETC) in regulating ferroptosis is not fully elucidated. Here, we reveal that pharmacological inhibition of the ETC complex I reduces ubiquinol levels while decreasing ATP levels and activating AMP-activated protein kinase (AMPK), the two effects known for their roles in promoting and suppressing ferroptosis, respectively. Consequently, the impact of complex I inhibitors on ferroptosis induced by glutathione peroxidase 4 (GPX4) inhibition is limited. The pharmacological inhibition of complex I in LKB1-AMPK-inactivated cells, or genetic ablation of complex I (which does not trigger apparent AMPK activation), abrogates the AMPK-mediated ferroptosis-suppressive effect and sensitizes cancer cells to GPX4-inactivation-induced ferroptosis. Furthermore, complex I inhibition synergizes with radiotherapy (RT) to selectively suppress the growth of LKB1-deficient tumors by inducing ferroptosis in mouse models. Our data demonstrate a multifaceted role of complex I in regulating ferroptosis and propose a ferroptosis-inducing therapeutic strategy for LKB1-deficient cancers.


Subject(s)
AMP-Activated Protein Kinases , Electron Transport Complex I , Ferroptosis , Phospholipid Hydroperoxide Glutathione Peroxidase , Protein Serine-Threonine Kinases , Ferroptosis/genetics , Ferroptosis/drug effects , Animals , Humans , AMP-Activated Protein Kinases/metabolism , AMP-Activated Protein Kinases/genetics , Electron Transport Complex I/metabolism , Electron Transport Complex I/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Mice , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/genetics , Cell Line, Tumor , Neoplasms/genetics , Neoplasms/pathology , Neoplasms/metabolism , Neoplasms/drug therapy , AMP-Activated Protein Kinase Kinases/genetics , Mitochondria/metabolism , Mitochondria/genetics , Mitochondria/drug effects , Xenograft Model Antitumor Assays , Signal Transduction , Female
2.
Nature ; 615(7953): 712-719, 2023 03.
Article in English | MEDLINE | ID: mdl-36922590

ABSTRACT

Mitochondria are critical to the governance of metabolism and bioenergetics in cancer cells1. The mitochondria form highly organized networks, in which their outer and inner membrane structures define their bioenergetic capacity2,3. However, in vivo studies delineating the relationship between the structural organization of mitochondrial networks and their bioenergetic activity have been limited. Here we present an in vivo structural and functional analysis of mitochondrial networks and bioenergetic phenotypes in non-small cell lung cancer (NSCLC) using an integrated platform consisting of positron emission tomography imaging, respirometry and three-dimensional scanning block-face electron microscopy. The diverse bioenergetic phenotypes and metabolic dependencies we identified in NSCLC tumours align with distinct structural organization of mitochondrial networks present. Further, we discovered that mitochondrial networks are organized into distinct compartments within tumour cells. In tumours with high rates of oxidative phosphorylation (OXPHOSHI) and fatty acid oxidation, we identified peri-droplet mitochondrial networks wherein mitochondria contact and surround lipid droplets. By contrast, we discovered that in tumours with low rates of OXPHOS (OXPHOSLO), high glucose flux regulated perinuclear localization of mitochondria, structural remodelling of cristae and mitochondrial respiratory capacity. Our findings suggest that in NSCLC, mitochondrial networks are compartmentalized into distinct subpopulations that govern the bioenergetic capacity of tumours.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Energy Metabolism , Lung Neoplasms , Mitochondria , Humans , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/ultrastructure , Fatty Acids/metabolism , Glucose/metabolism , Lipid Droplets/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/ultrastructure , Microscopy, Electron , Mitochondria/metabolism , Mitochondria/ultrastructure , Oxidative Phosphorylation , Phenotype , Positron-Emission Tomography
3.
Nature ; 593(7860): 586-590, 2021 05.
Article in English | MEDLINE | ID: mdl-33981038

ABSTRACT

Ferroptosis, a form of regulated cell death that is induced by excessive lipid peroxidation, is a key tumour suppression mechanism1-4. Glutathione peroxidase 4 (GPX4)5,6 and ferroptosis suppressor protein 1 (FSP1)7,8 constitute two major ferroptosis defence systems. Here we show that treatment of cancer cells with GPX4 inhibitors results in acute depletion of N-carbamoyl-L-aspartate, a pyrimidine biosynthesis intermediate, with concomitant accumulation of uridine. Supplementation with dihydroorotate or orotate-the substrate and product of dihydroorotate dehydrogenase (DHODH)-attenuates or potentiates ferroptosis induced by inhibition of GPX4, respectively, and these effects are particularly pronounced in cancer cells with low expression of GPX4 (GPX4low). Inactivation of DHODH induces extensive mitochondrial lipid peroxidation and ferroptosis in GPX4low cancer cells, and synergizes with ferroptosis inducers to induce these effects in GPX4high cancer cells. Mechanistically, DHODH operates in parallel to mitochondrial GPX4 (but independently of cytosolic GPX4 or FSP1) to inhibit ferroptosis in the mitochondrial inner membrane by reducing ubiquinone to ubiquinol (a radical-trapping antioxidant with anti-ferroptosis activity). The DHODH inhibitor brequinar selectively suppresses GPX4low tumour growth by inducing ferroptosis, whereas combined treatment with brequinar and sulfasalazine, an FDA-approved drug with ferroptosis-inducing activity, synergistically induces ferroptosis and suppresses GPX4high tumour growth. Our results identify a DHODH-mediated ferroptosis defence mechanism in mitochondria and suggest a therapeutic strategy of targeting ferroptosis in cancer treatment.


Subject(s)
Dihydroorotate Dehydrogenase/metabolism , Ferroptosis , Mitochondria/metabolism , Neoplasms/enzymology , Animals , Biphenyl Compounds/pharmacology , Cell Line, Tumor , Dihydroorotate Dehydrogenase/genetics , Female , Gene Deletion , Humans , Lipid Peroxidation , Metabolomics , Mice, Nude , Phospholipid Hydroperoxide Glutathione Peroxidase/antagonists & inhibitors , Phospholipid Hydroperoxide Glutathione Peroxidase/genetics , Xenograft Model Antitumor Assays
4.
Proc Natl Acad Sci U S A ; 119(26): e2121987119, 2022 06 28.
Article in English | MEDLINE | ID: mdl-35749365

ABSTRACT

Mechanisms of defense against ferroptosis (an iron-dependent form of cell death induced by lipid peroxidation) in cellular organelles remain poorly understood, hindering our ability to target ferroptosis in disease treatment. In this study, metabolomic analyses revealed that treatment of cancer cells with glutathione peroxidase 4 (GPX4) inhibitors results in intracellular glycerol-3-phosphate (G3P) depletion. We further showed that supplementation of cancer cells with G3P attenuates ferroptosis induced by GPX4 inhibitors in a G3P dehydrogenase 2 (GPD2)-dependent manner; GPD2 deletion sensitizes cancer cells to GPX4 inhibition-induced mitochondrial lipid peroxidation and ferroptosis, and combined deletion of GPX4 and GPD2 synergistically suppresses tumor growth by inducing ferroptosis in vivo. Mechanistically, inner mitochondrial membrane-localized GPD2 couples G3P oxidation with ubiquinone reduction to ubiquinol, which acts as a radical-trapping antioxidant to suppress ferroptosis in mitochondria. Taken together, these results reveal that GPD2 participates in ferroptosis defense in mitochondria by generating ubiquinol.


Subject(s)
Ferroptosis , Glycerolphosphate Dehydrogenase , Lipid Peroxidation , Mitochondria , Mitochondrial Proteins , Neoplasms , Cell Line, Tumor , Ferroptosis/genetics , Glycerolphosphate Dehydrogenase/antagonists & inhibitors , Glycerolphosphate Dehydrogenase/genetics , Glycerolphosphate Dehydrogenase/metabolism , Humans , Lipid Peroxidation/genetics , Mitochondria/enzymology , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Neoplasms/enzymology , Neoplasms/pathology , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism
5.
J Immunol ; 206(6): 1127-1139, 2021 03 15.
Article in English | MEDLINE | ID: mdl-33558372

ABSTRACT

T effector cells promote inflammation in asthmatic patients, and both Th2 and Th17 CD4 T cells have been implicated in severe forms of the disease. The metabolic phenotypes and dependencies of these cells, however, remain poorly understood in the regulation of airway inflammation. In this study, we show the bronchoalveolar lavage fluid of asthmatic patients had markers of elevated glucose and glutamine metabolism. Further, peripheral blood T cells of asthmatics had broadly elevated expression of metabolic proteins when analyzed by mass cytometry compared with healthy controls. Therefore, we hypothesized that glucose and glutamine metabolism promote allergic airway inflammation. We tested this hypothesis in two murine models of airway inflammation. T cells from lungs of mice sensitized with Alternaria alternata extract displayed genetic signatures for elevated oxidative and glucose metabolism by single-cell RNA sequencing. This result was most pronounced when protein levels were measured in IL-17-producing cells and was recapitulated when airway inflammation was induced with house dust mite plus LPS, a model that led to abundant IL-4- and IL-17-producing T cells. Importantly, inhibitors of the glucose transporter 1 or glutaminase in vivo attenuated house dust mite + LPS eosinophilia, T cell cytokine production, and airway hyperresponsiveness as well as augmented the immunosuppressive properties of dexamethasone. These data show that T cells induce markers to support metabolism in vivo in airway inflammation and that this correlates with inflammatory cytokine production. Targeting metabolic pathways may provide a new direction to protect from disease and enhance the effectiveness of steroid therapy.


Subject(s)
Asthma/drug therapy , Dexamethasone/pharmacology , Glucose Transporter Type 1/antagonists & inhibitors , Glutaminase/antagonists & inhibitors , Immunosuppressive Agents/pharmacology , Adult , Alternaria/immunology , Animals , Asthma/blood , Asthma/immunology , Biomarkers/analysis , Biomarkers/metabolism , Blood Glucose/metabolism , Bronchoalveolar Lavage Fluid/immunology , Case-Control Studies , Cells, Cultured , Dexamethasone/therapeutic use , Disease Models, Animal , Drug Synergism , Female , Glucose Transporter Type 1/metabolism , Glutaminase/metabolism , Glutamine/metabolism , Healthy Volunteers , Humans , Immunosuppressive Agents/therapeutic use , Lung/cytology , Lung/drug effects , Lung/immunology , Male , Mice , Middle Aged , Primary Cell Culture , Pyroglyphidae/immunology , Th17 Cells/drug effects , Th17 Cells/immunology , Th17 Cells/metabolism , Th2 Cells/drug effects , Th2 Cells/immunology , Th2 Cells/metabolism , Young Adult
6.
Proteins ; 90(4): 919-935, 2022 04.
Article in English | MEDLINE | ID: mdl-34773424

ABSTRACT

Detailed description of the mechanism of action of the therapeutic antibodies is essential for the functional characterization and future optimization of potential clinical agents. We recently developed KD035, a fully human antibody targeting vascular endothelial growth factor receptor 2 (VEGFR2). KD035 blocked VEGF-A, and VEGF-C-mediated VEGFR2 activation, as demonstrated by the in vitro binding and competition assays and functional cellular assays. Here, we report a computational model of the complex between the variable fragment of KD035 (KD035(Fv)) and the domains 2 and 3 of the extracellular portion of VEGFR2 (VEGFR2(D2-3)). Our modeling was guided by a priori experimental information including the X-ray structures of KD035 and related antibodies, binding assays, target domain mapping and comparison of KD035 affinity for VEGFR2 from different species. The accuracy of the model was assessed by molecular dynamics simulations, and subsequently validated by mutagenesis and binding analysis. Importantly, the steps followed during the generation of this model can set a precedent for future in silico efforts aimed at the accurate description of the antibody-antigen and more broadly protein-protein complexes.


Subject(s)
Antibodies , Vascular Endothelial Growth Factor A , Humans , Molecular Dynamics Simulation , Vascular Endothelial Growth Factor A/metabolism
8.
Mol Cell ; 35(3): 316-26, 2009 Aug 14.
Article in English | MEDLINE | ID: mdl-19683495

ABSTRACT

MDM2 associates with ribosomal protein S7, and this interaction is required to inhibit MDM2's E3 ligase activity, leading to stabilization of MDM2 and p53. Notably, the MDM2 homolog MDMX facilitates the inhibition of MDM2 E3 ligase activity by S7. Further, ablation of S7 inhibits MDM2 and p53 accumulation induced by different stress signals in some cell types. Thus, ribosomal/nucleolar stress is likely a key integrating event in DNA damage signaling to p53. Interestingly, S7 is itself a substrate for MDM2 E3 ligase activity both in vitro and in vivo. An S7-ubiquitin fusion protein (S7-Ub) selectively inhibits MDM2 degradation of p53 and is unaffected by MDMX. S7-Ub promotes apoptosis to a greater extent than S7 alone. This indicates that MDM2 ubiquitination of S7 is involved in sustaining the p53 response. Thus, S7 functions as both effector and affector of MDM2 to ensure a proper cellular response to different stress signals.


Subject(s)
Proto-Oncogene Proteins c-mdm2/metabolism , Ribosomal Proteins/physiology , Animals , Binding Sites , Cell Line , Down-Regulation , Humans , Mice , Protein Interaction Mapping , Proto-Oncogene Proteins c-mdm2/genetics , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Signal Transduction , Stress, Physiological , Tumor Suppressor Protein p53/metabolism , Two-Hybrid System Techniques , Ubiquitination
9.
Proc Natl Acad Sci U S A ; 111(47): 16814-9, 2014 Nov 25.
Article in English | MEDLINE | ID: mdl-25385601

ABSTRACT

Rho-associated kinase 2 (ROCK2) regulates the secretion of proinflammatory cytokines and the development of autoimmunity in mice. Data from a phase 1 clinical trial demonstrate that oral administration of KD025, a selective ROCK2 inhibitor, to healthy human subjects down-regulates the ability of T cells to secrete IL-21 and IL-17 by 90% and 60%, respectively, but not IFN-γ in response to T-cell receptor stimulation in vitro. Pharmacological inhibition with KD025 or siRNA-mediated inhibition of ROCK2, but not ROCK1, significantly diminished STAT3 phosphorylation and binding to IL-17 and IL-21 promoters and reduced IFN regulatory factor 4 and nuclear hormone RAR-related orphan receptor γt protein levels in T cells derived from healthy subjects or rheumatoid arthritis patients. Simultaneously, treatment with KD025 also promotes the suppressive function of regulatory T cells through up-regulation of STAT5 phosphorylation and positive regulation of forkhead box p3 expression. The administration of KD025 in vivo down-regulates the progression of collagen-induced arthritis in mice via targeting of the Th17-mediated pathway. Thus, ROCK2 signaling appears to be instrumental in regulating the balance between proinflammatory and regulatory T-cell subsets. Targeting of ROCK2 in man may therefore restore disrupted immune homeostasis and have a role in the treatment of autoimmunity.


Subject(s)
CD4-Positive T-Lymphocytes/drug effects , Interleukin-17/metabolism , Interleukins/metabolism , Protein Kinase Inhibitors/pharmacology , STAT3 Transcription Factor/physiology , rho-Associated Kinases/antagonists & inhibitors , Administration, Oral , CD4-Positive T-Lymphocytes/metabolism , Cells, Cultured , Humans , Interleukin-17/genetics , Interleukins/genetics , Phosphorylation , Promoter Regions, Genetic , Protein Kinase Inhibitors/administration & dosage , STAT3 Transcription Factor/metabolism , Transcription, Genetic
10.
Proc Natl Acad Sci U S A ; 110(46): 18626-31, 2013 Nov 12.
Article in English | MEDLINE | ID: mdl-24167278

ABSTRACT

GATA-binding protein 3 (Gata3) controls the differentiation of naive CD4 T cells into T helper 2 (Th2) cells by induction of chromatin remodeling of the Th2 cytokine gene loci, direct transactivation of Il5 and Il13 genes, and inhibition of Ifng. Gata3 also facilitates Th2 cell proliferation via additional mechanisms that are far less well understood. We herein found that Gata3 associates with RuvB-like protein 2 (Ruvbl2) and represses the expression of a CDK inhibitor, cyclin-dependent kinase inhibitor 2c (Cdkn2c) to facilitate the proliferation of Th2 cells. Gata3 directly bound to the Cdkn2c locus in an Ruvbl2-dependent manner. The defect in the proliferation of Gata3-deficient Th2 cells is rescued by the knockdown of Cdkn2c, indicating that Cdkn2c is a key molecule involved in the Gata3-mediated induction of Th2 cell proliferation. Ruvbl2-knockdown Th2 cells showed decreased antigen-induced expansion and caused less airway inflammation in vivo. We therefore have identified a functional Gata3/Ruvbl2 complex that regulates the proliferation of differentiating Th2 cells through the repression of a CDK inhibitor, Cdkn2c.


Subject(s)
Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p18/metabolism , DNA Helicases/immunology , GATA3 Transcription Factor/immunology , Gene Expression Regulation/immunology , Multiprotein Complexes/immunology , Th2 Cells/cytology , ATPases Associated with Diverse Cellular Activities , Animals , Bromodeoxyuridine , Chromatin Immunoprecipitation , DNA Primers/genetics , Gene Knockdown Techniques , Immunoblotting , Immunoprecipitation , Luciferases , Mass Spectrometry , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Real-Time Polymerase Chain Reaction
11.
Nat Commun ; 15(1): 79, 2024 01 02.
Article in English | MEDLINE | ID: mdl-38167301

ABSTRACT

How cells coordinate cell cycling with cell survival and death remains incompletely understood. Here, we show that cell cycle arrest has a potent suppressive effect on ferroptosis, a form of regulated cell death induced by overwhelming lipid peroxidation at cellular membranes. Mechanistically, cell cycle arrest induces diacylglycerol acyltransferase (DGAT)-dependent lipid droplet formation to sequester excessive polyunsaturated fatty acids (PUFAs) that accumulate in arrested cells in triacylglycerols (TAGs), resulting in ferroptosis suppression. Consequently, DGAT inhibition orchestrates a reshuffling of PUFAs from TAGs to phospholipids and re-sensitizes arrested cells to ferroptosis. We show that some slow-cycling antimitotic drug-resistant cancer cells, such as 5-fluorouracil-resistant cells, have accumulation of lipid droplets and that combined treatment with ferroptosis inducers and DGAT inhibitors effectively suppresses the growth of 5-fluorouracil-resistant tumors by inducing ferroptosis. Together, these results reveal a role for cell cycle arrest in driving ferroptosis resistance and suggest a ferroptosis-inducing therapeutic strategy to target slow-cycling therapy-resistant cancers.


Subject(s)
Ferroptosis , Neoplasms , Humans , Lipid Droplets/metabolism , Fatty Acids, Unsaturated/metabolism , Lipid Peroxidation , Triglycerides/metabolism , Cell Cycle Checkpoints , Neoplasms/metabolism , Diacylglycerol O-Acyltransferase/metabolism , Fluorouracil/pharmacology , Fluorouracil/therapeutic use
12.
Proc Natl Acad Sci U S A ; 107(16): 7461-6, 2010 Apr 20.
Article in English | MEDLINE | ID: mdl-20351271

ABSTRACT

We identified a p53 target gene, phosphate-activated mitochondrial glutaminase (GLS2), a key enzyme in conversion of glutamine to glutamate, and thereby a regulator of glutathione (GSH) synthesis and energy production. GLS2 expression is induced in response to DNA damage or oxidative stress in a p53-dependent manner, and p53 associates with the GLS2 promoter. Elevated GLS2 facilitates glutamine metabolism and lowers intracellular reactive oxygen species (ROS) levels, resulting in an overall decrease in DNA oxidation as determined by measurement of 8-OH-dG content in both normal and stressed cells. Further, siRNA down-regulation of either GLS2 or p53 compromises the GSH-dependent antioxidant system and increases intracellular ROS levels. High ROS levels following GLS2 knockdown also coincide with stimulation of p53-induced cell death. We propose that GLS2 control of intracellular ROS levels and the apoptotic response facilitates the ability of p53 to protect cells from accumulation of genomic damage and allows cells to survive after mild and repairable genotoxic stress. Indeed, overexpression of GLS2 reduces the growth of tumor cells and colony formation. Further, compared with normal tissue, GLS2 expression is reduced in liver tumors. Thus, our results provide evidence for a unique metabolic role for p53, linking glutamine metabolism, energy, and ROS homeostasis, which may contribute to p53 tumor suppressor function.


Subject(s)
Glutaminase/metabolism , Glutamine/metabolism , Promoter Regions, Genetic , Reactive Oxygen Species , Tumor Suppressor Protein p53/metabolism , Animals , Antioxidants/metabolism , Apoptosis , Glutathione/metabolism , Humans , Mice , Mitochondria/metabolism , Oligonucleotide Array Sequence Analysis , RNA Interference
13.
Nat Commun ; 14(1): 3673, 2023 06 21.
Article in English | MEDLINE | ID: mdl-37339981

ABSTRACT

The cystine transporter solute carrier family 7 member 11 (SLC7A11; also called xCT) protects cancer cells from oxidative stress and is overexpressed in many cancers. Here we report a surprising finding that, whereas moderate overexpression of SLC7A11 is beneficial for cancer cells treated with H2O2, a common oxidative stress inducer, its high overexpression dramatically increases H2O2-induced cell death. Mechanistically, high cystine uptake in cancer cells with high overexpression of SLC7A11 in combination with H2O2 treatment results in toxic buildup of intracellular cystine and other disulfide molecules, NADPH depletion, redox system collapse, and rapid cell death (likely disulfidptosis). We further show that high overexpression of SLC7A11 promotes tumor growth but suppresses tumor metastasis, likely because metastasizing cancer cells with high expression of SLC7A11 are particularly susceptible to oxidative stress. Our findings reveal that SLC7A11 expression level dictates cancer cells' sensitivity to oxidative stress and suggests a context-dependent role for SLC7A11 in tumor biology.


Subject(s)
Cystine , Neoplasms , Cystine/metabolism , Cell Line, Tumor , Hydrogen Peroxide/pharmacology , Hydrogen Peroxide/metabolism , Oxidative Stress , Disulfides/metabolism , Amino Acid Transport System y+/genetics , Amino Acid Transport System y+/metabolism , Neoplasms/genetics
14.
Sci Transl Med ; 15(715): eade3157, 2023 09 27.
Article in English | MEDLINE | ID: mdl-37756381

ABSTRACT

Obesity is increasing worldwide and leads to a multitude of metabolic diseases, including cardiovascular disease, type 2 diabetes, nonalcoholic fatty liver disease, and nonalcoholic steatohepatitis (NASH). Cysteine-rich angiogenic inducer 61 (CYR61) is associated with the progression of NASH, but it has been described to have anti- and proinflammatory properties. We sought to examine the role of liver CYR61 in NASH progression. CYR61 liver-specific knockout mice on a NASH diet showed improved glucose tolerance, decreased liver inflammation, and reduced fibrosis. CYR61 polarized infiltrating monocytes promoting a proinflammatory/profibrotic phenotype through an IRAK4/SYK/NF-κB signaling cascade. In vitro, CYR61 activated a profibrotic program, including PDGFa/PDGFb expression in macrophages, in an IRAK4/SYK/NF-κB-dependent manner. Furthermore, targeted-antibody blockade reduced CYR61-driven signaling in macrophages in vitro and in vivo, reducing fibrotic development. This study demonstrates that CYR61 is a key driver of liver inflammation and fibrosis in NASH.


Subject(s)
Diabetes Mellitus, Type 2 , Non-alcoholic Fatty Liver Disease , Mice , Animals , Non-alcoholic Fatty Liver Disease/pathology , Interleukin-1 Receptor-Associated Kinases/metabolism , NF-kappa B/metabolism , Diabetes Mellitus, Type 2/metabolism , Disease Models, Animal , Liver/metabolism , Hepatocytes/metabolism , Fibrosis , Macrophages/metabolism , Mice, Knockout , Mice, Inbred C57BL
15.
Nat Cell Biol ; 25(3): 404-414, 2023 03.
Article in English | MEDLINE | ID: mdl-36747082

ABSTRACT

SLC7A11-mediated cystine uptake suppresses ferroptosis yet promotes cell death under glucose starvation; the nature of the latter cell death remains unknown. Here we show that aberrant accumulation of intracellular disulfides in SLC7A11high cells under glucose starvation induces a previously uncharacterized form of cell death distinct from apoptosis and ferroptosis. We term this cell death disulfidptosis. Chemical proteomics and cell biological analyses showed that glucose starvation in SLC7A11high cells induces aberrant disulfide bonds in actin cytoskeleton proteins and F-actin collapse in a SLC7A11-dependent manner. CRISPR screens and functional studies revealed that inactivation of the WAVE regulatory complex (which promotes actin polymerization and lamellipodia formation) suppresses disulfidptosis, whereas constitutive activation of Rac promotes disulfidptosis. We further show that glucose transporter inhibitors induce disulfidptosis in SLC7A11high cancer cells and suppress SLC7A11high tumour growth. Our results reveal that the susceptibility of the actin cytoskeleton to disulfide stress mediates disulfidptosis and suggest a therapeutic strategy to target disulfidptosis in cancer treatment.


Subject(s)
Disulfides , Neoplasms , Humans , Neoplasms/metabolism , Apoptosis , Actin Cytoskeleton/metabolism , Glucose/metabolism
16.
Nucleic Acids Res ; 38(21): 7587-98, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20671028

ABSTRACT

Mdm2, a central negative regulator of the p53 tumor suppressor, possesses a Really Interesting New Gene (RING) domain within its C-terminus. In addition to E3 ubiquitin ligase activity, the Mdm2 RING preferentially binds adenine base nucleotides, and such binding leads to a conformational change in the Mdm2 C-terminus. Here, we present further biochemical analysis of the nucleotide-Mdm2 interaction. We have found that MdmX, an Mdm2 family member with high sequence homology, binds adenine nucleotides with similar affinity and specificity as Mdm2, suggesting that residues involved in nucleotide binding may be conserved between the two proteins and adenosine triphosphate (ATP) binding may have similar functional consequences for both Mdm family members. By generating and testing a series of proteins with deletions and substitution mutations within the Mdm2 RING, we mapped the specific adenine nucleotide binding region of Mdm2 to residues 429-484, encompassing the minimal RING domain. Using a series of ATP derivatives, we demonstrate that phosphate coordination by the Mdm2 P-loop contributes to, but is not primarily responsible for, ATP binding. Additionally, we have identified the 2' and 3' hydroxyls of the ribose and the C6 amino group of the adenine base moiety as being essential for binding.


Subject(s)
Adenosine Triphosphate/chemistry , Proto-Oncogene Proteins c-mdm2/chemistry , Adenine/chemistry , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Binding Sites , Humans , Magnesium/chemistry , Molecular Sequence Data , Protein Binding , Protein Structure, Tertiary , Proto-Oncogene Proteins c-mdm2/metabolism
17.
Nat Commun ; 13(1): 2206, 2022 04 22.
Article in English | MEDLINE | ID: mdl-35459868

ABSTRACT

Targeting ferroptosis, a unique cell death modality triggered by unrestricted lipid peroxidation, in cancer therapy is hindered by our incomplete understanding of ferroptosis mechanisms under specific cancer genetic contexts. KEAP1 (kelch-like ECH associated protein 1) is frequently mutated or inactivated in lung cancers, and KEAP1 mutant lung cancers are refractory to most therapies, including radiotherapy. In this study, we identify ferroptosis suppressor protein 1 (FSP1, also known as AIFM2) as a transcriptional target of nuclear factor erythroid 2-related factor 2 (NRF2) and reveal that the ubiquinone (CoQ)-FSP1 axis mediates ferroptosis- and radiation- resistance in KEAP1 deficient lung cancer cells. We further show that pharmacological inhibition of the CoQ-FSP1 axis sensitizes KEAP1 deficient lung cancer cells or patient-derived xenograft tumors to radiation through inducing ferroptosis. Together, our study identifies CoQ-FSP1 as a key downstream effector of KEAP1-NRF2 pathway and as a potential therapeutic target for treating KEAP1 mutant lung cancers.


Subject(s)
Apoptosis Regulatory Proteins , Ferroptosis , Lung Neoplasms , Mitochondrial Proteins , Ubiquinone , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Ferroptosis/genetics , Humans , Kelch-Like ECH-Associated Protein 1/genetics , Kelch-Like ECH-Associated Protein 1/metabolism , Lipid Peroxidation , Lung Neoplasms/genetics , Lung Neoplasms/radiotherapy , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Ubiquinone/genetics , Ubiquinone/metabolism
18.
Cell Chem Biol ; 29(3): 423-435.e10, 2022 03 17.
Article in English | MEDLINE | ID: mdl-34715056

ABSTRACT

Efforts to target glucose metabolism in cancer have been limited by the poor potency and specificity of existing anti-glycolytic agents and a poor understanding of the glucose dependence of cancer subtypes in vivo. Here, we present an extensively characterized series of potent, orally bioavailable inhibitors of the class I glucose transporters (GLUTs). The representative compound KL-11743 specifically blocks glucose metabolism, triggering an acute collapse in NADH pools and a striking accumulation of aspartate, indicating a dramatic shift toward oxidative phosphorylation in the mitochondria. Disrupting mitochondrial metabolism via chemical inhibition of electron transport, deletion of the malate-aspartate shuttle component GOT1, or endogenous mutations in tricarboxylic acid cycle enzymes, causes synthetic lethality with KL-11743. Patient-derived xenograft models of succinate dehydrogenase A (SDHA)-deficient cancers are specifically sensitive to KL-11743, providing direct evidence that TCA cycle-mutant tumors are vulnerable to GLUT inhibitors in vivo.


Subject(s)
Citric Acid Cycle , Neoplasms , Aspartic Acid/metabolism , Glucose/metabolism , Humans , Mitochondria/metabolism , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism
19.
iScience ; 24(6): 102649, 2021 Jun 25.
Article in English | MEDLINE | ID: mdl-34151236

ABSTRACT

Metabolic reprogramming in cancer cells can create metabolic liabilities. KEAP1-mutant lung cancer is refractory to most current therapies. Here we show that KEAP1 deficiency promotes glucose dependency in lung cancer cells, and KEAP1-mutant/deficient lung cancer cells are more vulnerable to glucose deprivation than their WT counterparts. Mechanistically, KEAP1 inactivation in lung cancer cells induces constitutive activation of NRF2 transcription factor and aberrant expression of NRF2 target cystine transporter SLC7A11; under glucose limitation, high cystine uptake in KEAP1-inactivated lung cancer cells stimulates toxic intracellular disulfide buildup, NADPH depletion, and cell death, which can be rescued by genetic ablation of NRF2-SLC7A11 axis or treatments inhibiting disulfide accumulation. Finally, we show that KEAP1-inactivated lung cancer cells or xenograft tumors are sensitive to glucose transporter inhibitor. Together, our results reveal that KEAP1 deficiency induces glucose dependency in lung cancer cells and uncover a therapeutically relevant metabolic liability.

20.
J Med Chem ; 63(10): 5201-5211, 2020 05 28.
Article in English | MEDLINE | ID: mdl-32282207

ABSTRACT

Aerobic glycolysis, originally identified by Warburg as a hallmark of cancer, has recently been implicated in immune cell activation and growth. Glucose, the starting material for glycolysis, is transported through the cellular membrane by a family of glucose transporters (GLUTs). Therefore, targeting glucose transporters to regulate aerobic glycolysis is an attractive approach to identify potential therapeutic agents for cancers and autoimmune diseases. Herein, we describe the discovery and optimization of a class of potent, orally bioavailable inhibitors of glucose transporters, targeting both GLUT1 and GLUT3.


Subject(s)
Drug Discovery/methods , Glucose Transport Proteins, Facilitative/antagonists & inhibitors , Glucose Transport Proteins, Facilitative/metabolism , Glucose/metabolism , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Caco-2 Cells , Dose-Response Relationship, Drug , Drug Discovery/trends , Glycolysis/drug effects , Glycolysis/physiology , Humans , Mice , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Rats
SELECTION OF CITATIONS
SEARCH DETAIL