Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Nat Metab ; 5(4): 538-539, 2023 04.
Article in English | MEDLINE | ID: mdl-37024755
2.
bioRxiv ; 2023 Dec 17.
Article in English | MEDLINE | ID: mdl-38168428

ABSTRACT

Mutations in the NRF2-KEAP1 pathway are common in non-small cell lung cancer (NSCLC) and confer broad-spectrum therapeutic resistance, leading to poor outcomes. The cystine/glutamate antiporter, system xc-, is one of the >200 cytoprotective proteins controlled by NRF2, which can be non-invasively imaged by (S)-4-(3-18F-fluoropropyl)-l-glutamate ([18F]FSPG) positron emission tomography (PET). Through genetic and pharmacologic manipulation, we show that [18F]FSPG provides a sensitive and specific marker of NRF2 activation in advanced preclinical models of NSCLC. We validate imaging readouts with metabolomic measurements of system xc- activity and their coupling to intracellular glutathione concentration. A redox gene signature was measured in patients from the TRACERx 421 cohort, suggesting an opportunity for patient stratification prior to imaging. Furthermore, we reveal that system xc- is a metabolic vulnerability that can be therapeutically targeted for sustained tumour growth suppression in aggressive NSCLC. Our results establish [18F]FSPG as predictive marker of therapy resistance in NSCLC and provide the basis for the clinical evaluation of both imaging and therapeutic agents that target this important antioxidant pathway.

3.
Proc Natl Acad Sci U S A ; 119(43): e2202736119, 2022 10 25.
Article in English | MEDLINE | ID: mdl-36252013

ABSTRACT

Copper is an essential metal nutrient for life that often relies on redox cycling between Cu(I) and Cu(II) oxidation states to fulfill its physiological roles, but alterations in cellular redox status can lead to imbalances in copper homeostasis that contribute to cancer and other metalloplasias with metal-dependent disease vulnerabilities. Copper-responsive fluorescent probes offer powerful tools to study labile copper pools, but most of these reagents target Cu(I), with limited methods for monitoring Cu(II) owing to its potent fluorescence quenching properties. Here, we report an activity-based sensing strategy for turn-on, oxidation state-specific detection of Cu(II) through metal-directed acyl imidazole chemistry. Cu(II) binding to a metal and oxidation state-specific receptor that accommodates the harder Lewis acidity of Cu(II) relative to Cu(I) activates the pendant dye for reaction with proximal biological nucleophiles and concomitant metal ion release, thus avoiding fluorescence quenching. Copper-directed acyl imidazole 649 for Cu(II) (CD649.2) provides foundational information on the existence and regulation of labile Cu(II) pools, including identifying divalent metal transporter 1 (DMT1) as a Cu(II) importer, labile Cu(II) increases in response to oxidative stress induced by depleting total glutathione levels, and reciprocal increases in labile Cu(II) accompanied by decreases in labile Cu(I) induced by oncogenic mutations that promote oxidative stress.


Subject(s)
Copper , Fluorescent Dyes , Copper/metabolism , Fluorescent Dyes/chemistry , Glutathione/metabolism , Imidazoles , Oncogenes , Oxidation-Reduction
4.
Annu Rev Pharmacol Toxicol ; 62: 279-300, 2022 01 06.
Article in English | MEDLINE | ID: mdl-34499527

ABSTRACT

The transcription factor NRF2 coordinates the expression of a vast array of cytoprotective and metabolic genes in response to various stress inputs to restore cellular homeostasis. Transient activation of NRF2 in healthy tissues has been long recognized as a cellular defense mechanism and is critical to prevent cancer initiation by carcinogens. However, cancer cells frequently hijack the protective capability of NRF2 to sustain the redox balance and meet their metabolic requirements for proliferation. Further, aberrant activation of NRF2 in cancer cells confers resistance to commonly used chemotherapeutic agents and radiotherapy. During the last decade, many research groups have attempted to block NRF2 activity in tumors to counteract the survival and proliferative advantage of cancer cells and reverse resistance to treatment. In this review, we highlight the role of NRF2 in cancer progression and discuss the past and current approaches to disable NRF2 signaling in tumors.


Subject(s)
NF-E2-Related Factor 2 , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Humans , NF-E2-Related Factor 2/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Signal Transduction
5.
Biomolecules ; 10(10)2020 09 25.
Article in English | MEDLINE | ID: mdl-32992842

ABSTRACT

Aberrant hyperactivation of nuclear factor erythroid 2 (NF-E2) p45-related factor 2 (NRF2) is a common event in many tumour types and associates with resistance to therapy and poor patient prognosis; however, its relevance in colorectal tumours is not well-established. Measuring the expression of surrogate genes for NRF2 activity in silico, in combination with validation in patients' samples, we show that the NRF2 pathway is upregulated in colorectal tumours and that high levels of nuclear NRF2 correlate with a poor patient prognosis. These results highlight the need to overcome the protection provided by NRF2 and present an opportunity to selectively kill cancer cells with hyperactive NRF2. Exploiting the CRISPR/Cas9 technology, we generated colorectal cancer cell lines with hyperactive NRF2 and used them to perform a drug screen. We identified AT9283, an Aurora kinase inhibitor, for its selectivity towards killing cancer cells with hyperactive NRF2 as a consequence to either genetic or pharmacological activation. Our results show that hyperactivation of NRF2 in colorectal cancer cells might present a vulnerability that could potentially be therapeutically exploited by using the Aurora kinase inhibitor AT9283.


Subject(s)
Benzimidazoles/pharmacology , Colorectal Neoplasms/drug therapy , NF-E2-Related Factor 2/genetics , Protein Kinase Inhibitors/pharmacology , Urea/analogs & derivatives , Benzimidazoles/adverse effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Drug Evaluation, Preclinical , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Protein Kinase Inhibitors/adverse effects , Signal Transduction/drug effects , Urea/adverse effects , Urea/pharmacology
6.
Redox Biol ; 30: 101440, 2020 02.
Article in English | MEDLINE | ID: mdl-32007910

ABSTRACT

Alterations in the NRF2/KEAP1 pathway result in the constitutive activation of NRF2, leading to the aberrant induction of antioxidant and detoxification enzymes, including NQO1. The NQO1 bioactivatable agent ß-lapachone can target cells with high NQO1 expression but relies in the generation of reactive oxygen species (ROS), which are actively scavenged in cells with NRF2/KEAP1 mutations. However, whether NRF2/KEAP1 mutations influence the response to ß-lapachone treatment remains unknown. To address this question, we assessed the cytotoxicity of ß-lapachone in a panel of NSCLC cell lines bearing either wild-type or mutant KEAP1. We found that, despite overexpression of NQO1, KEAP1 mutant cells were resistant to ß-lapachone due to enhanced detoxification of ROS, which prevented DNA damage and cell death. To evaluate whether specific inhibition of the NRF2-regulated antioxidant enzymes could abrogate resistance to ß-lapachone, we systematically inhibited the four major antioxidant cellular systems using genetic and/or pharmacologic approaches. We demonstrated that inhibition of the thioredoxin-dependent system or copper-zinc superoxide dismutase (SOD1) could abrogate NRF2-mediated resistance to ß-lapachone, while depletion of catalase or glutathione was ineffective. Interestingly, inhibition of SOD1 selectively sensitized KEAP1 mutant cells to ß-lapachone exposure. Our results suggest that NRF2/KEAP1 mutational status might serve as a predictive biomarker for response to NQO1-bioactivatable quinones in patients. Further, our results suggest SOD1 inhibition may have potential utility in combination with other ROS inducers in patients with KEAP1/NRF2 mutations.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Drug Resistance, Neoplasm , Lung Neoplasms/genetics , NAD(P)H Dehydrogenase (Quinone)/genetics , NF-E2-Related Factor 2/genetics , Naphthoquinones/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , HEK293 Cells , Humans , Kelch-Like ECH-Associated Protein 1/genetics , Lung Neoplasms/drug therapy , Mutation , Superoxide Dismutase-1/antagonists & inhibitors , Thioredoxin Reductase 1/antagonists & inhibitors
8.
Elife ; 82019 05 20.
Article in English | MEDLINE | ID: mdl-31107239

ABSTRACT

NRF2 is emerging as a major regulator of cellular metabolism. However, most studies have been performed in cancer cells, where co-occurring mutations and tumor selective pressures complicate the influence of NRF2 on metabolism. Here we use genetically engineered, non-transformed primary murine cells to isolate the most immediate effects of NRF2 on cellular metabolism. We find that NRF2 promotes the accumulation of intracellular cysteine and engages the cysteine homeostatic control mechanism mediated by cysteine dioxygenase 1 (CDO1), which catalyzes the irreversible metabolism of cysteine to cysteine sulfinic acid (CSA). Notably, CDO1 is preferentially silenced by promoter methylation in human non-small cell lung cancers (NSCLC) harboring mutations in KEAP1, the negative regulator of NRF2. CDO1 silencing promotes proliferation of NSCLC by limiting the futile metabolism of cysteine to the wasteful and toxic byproducts CSA and sulfite (SO32-), and depletion of cellular NADPH. Thus, CDO1 is a metabolic liability for NSCLC cells with high intracellular cysteine, particularly NRF2/KEAP1 mutant cells.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Cysteine Dioxygenase/metabolism , Kelch-Like ECH-Associated Protein 1/metabolism , Animals , Cysteine/analogs & derivatives , Cysteine/metabolism , DNA Methylation , Gene Silencing , Humans , Mice , NF-E2-Related Factor 2/metabolism , Promoter Regions, Genetic
9.
Cancer Cell ; 32(6): 723-725, 2017 12 11.
Article in English | MEDLINE | ID: mdl-29232549

ABSTRACT

The mechanisms by which chronic stress promote the development of pancreatic ductal adenocarcinoma (PDAC) are poorly defined. In this issue of Cancer Cell, Todoric et al. discover a role for impaired autophagy in the development of PDAC through p62-mediated activation of NRF2.


Subject(s)
Carcinoma in Situ , Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Humans , NF-E2-Related Factor 2 , Pancreas , Proto-Oncogene Proteins c-mdm2
10.
Mol Cell Biol ; 36(18): 2403-17, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27354066

ABSTRACT

Heat shock factor 1 (HSF1) monitors the structural integrity of the proteome. Phosphorylation at S326 is a hallmark for HSF1 activation, but the identity of the kinase(s) phosphorylating this site has remained elusive. We show here that the dietary agent phenethyl isothiocyanate (PEITC) inhibits heat shock protein 90 (Hsp90), the main negative regulator of HSF1; activates p38 mitogen-activated protein kinase (MAPK); and increases S326 phosphorylation, trimerization, and nuclear translocation of HSF1, and the transcription of a luciferase reporter, as well as the endogenous prototypic HSF1 target Hsp70. In vitro, all members of the p38 MAPK family rapidly and stoichiometrically catalyze the S326 phosphorylation. The use of stable knockdown cell lines and inhibitors indicated that among the p38 MAPKs, p38γ is the principal isoform responsible for the phosphorylation of HSF1 at S326 in cells. A protease-mass spectrometry approach confirmed S326 phosphorylation and unexpectedly revealed that p38 MAPK also catalyzes the phosphorylation of HSF1 at S303/307, previously known repressive posttranslational modifications. Thus, we have identified p38 MAPKs as highly efficient catalysts for the phosphorylation of HSF1. Furthermore, our findings suggest that the magnitude and persistence of activation of p38 MAPK are important determinants of the extent and duration of the heat shock response.


Subject(s)
DNA-Binding Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Serine/metabolism , Transcription Factors/genetics , Transcription, Genetic , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Cell Line , DNA-Binding Proteins/chemistry , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HeLa Cells , Heat Shock Transcription Factors , Humans , Isothiocyanates/pharmacology , Mice , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Phosphorylation , Protein Multimerization , Protein Transport , Transcription Factors/chemistry
11.
FEBS Lett ; 588(14): 2353-62, 2014 Jun 27.
Article in English | MEDLINE | ID: mdl-24873882

ABSTRACT

In mammals, the linker histone H1, involved in DNA packaging into chromatin, is represented by a family of variants. H1 tails undergo post-translational modifications (PTMs) that can be detected by mass spectrometry. We developed antibodies to analyze several of these as yet unexplored PTMs including the combination of H1.4 K26 acetylation or trimethylation and S27 phosphorylation. H1.2-T165 phosphorylation was detected at S and G2/M phases of the cell cycle and was dispensable for chromatin binding and cell proliferation; while the H1.4-K26 residue was essential for proper cell cycle progression. We conclude that histone H1 PTMs are dynamic over the cell cycle and that the recognition of modified lysines may be affected by phosphorylation of adjacent residues.


Subject(s)
Histones/metabolism , Protein Processing, Post-Translational , Acetylation , Cell Line, Tumor , Humans , Methylation , Phosphorylation
SELECTION OF CITATIONS
SEARCH DETAIL
...