Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Redox Biol ; 69: 103031, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38184997

ABSTRACT

The Kelch-like ECH-associated protein 1 (KEAP1) - Nuclear factor erythroid 2 -related factor 2 (NRF2) pathway is the major transcriptional stress response system in cells against oxidative and electrophilic stress. NRF2 is frequently constitutively active in many cancers, rendering the cells resistant to chemo- and radiotherapy. Loss-of-function (LOF) mutations in the repressor protein KEAP1 are common in non-small cell lung cancer, particularly adenocarcinoma. While the mutations can occur throughout the gene, they are enriched in certain areas, indicating that these may have unique functional importance. In this study, we show that in the GSEA analysis of TCGA lung adenocarcinoma RNA-seq data, the KEAP1 mutations in R320 and R470 were associated with enhanced Tumor Necrosis Factor alpha (TNFα) - Nuclear Factor kappa subunit B (NFκB) signaling as well as MYC and MTORC1 pathways. To address the functional role of these hotspot mutations, affinity purification and mass spectrometry (AP-MS) analysis of wild type (wt) KEAP1 and its mutation forms, R320Q and R470C were employed to interrogate differences in the protein interactome. We identified TNF receptor associated factor 2 (TRAF2) as a putative protein interaction partner. Both mutant KEAP1 forms showed increased interaction with TRAF2 and other anti-apoptotic proteins, suggesting that apoptosis signalling could be affected by the protein interactions. A549 lung adenocarcinoma cells overexpressing mutant KEAP1 showed high TRAF2-mediated NFκB activity and increased protection against apoptosis, XIAP being one of the key proteins involved in anti-apoptotic signalling. To conclude, KEAP1 R320Q and R470C and its interaction with TRAF2 leads to activation of NFκB pathway, thereby protecting against apoptosis.


Subject(s)
Adenocarcinoma of Lung , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Lung Neoplasms/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , TNF Receptor-Associated Factor 2/genetics , TNF Receptor-Associated Factor 2/metabolism , Kelch-Like ECH-Associated Protein 1/genetics , Kelch-Like ECH-Associated Protein 1/metabolism , NF-E2-Related Factor 2/metabolism , Cell Line, Tumor , Intracellular Signaling Peptides and Proteins/metabolism , Adenocarcinoma of Lung/genetics , Apoptosis/genetics , NF-kappa B/genetics , NF-kappa B/metabolism , Mutation
2.
Redox Biol ; 63: 102726, 2023 07.
Article in English | MEDLINE | ID: mdl-37146513

ABSTRACT

The KEAP1-NRF2 pathway is the key regulator of cellular defense against both extrinsic and intrinsic oxidative and electrophilic stimuli. Since its discovery in the 1990s, its seminal role in various disease pathologies has become well appreciated, motivating research to elucidate the intricacies of NRF2 signaling and its downstream effects to identify novel targets for therapy. In this graphical review, we present an updated overview of the KEAP1-NRF2 signaling, focusing on the progress made within the past ten years. Specifically, we highlight the advances made in understanding the mechanism of activation of NRF2, resulting in novel discoveries in its therapeutic targeting. Furthermore, we will summarize new findings in the rapidly expanding field of NRF2 in cancer, with important implications for its diagnostics and treatment.


Subject(s)
NF-E2-Related Factor 2 , Neoplasms , Humans , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Kelch-Like ECH-Associated Protein 1/metabolism , Oxidative Stress , Neoplasms/drug therapy , Neoplasms/genetics
3.
Cardiovasc Res ; 117(5): 1339-1357, 2021 04 23.
Article in English | MEDLINE | ID: mdl-32683448

ABSTRACT

AIMS: Oxidized phospholipids and microRNAs (miRNAs) are increasingly recognized to play a role in endothelial dysfunction driving atherosclerosis. NRF2 transcription factor is one of the key mediators of the effects of oxidized phospholipids, but the gene regulatory mechanisms underlying the process remain obscure. Here, we investigated the genome-wide effects of oxidized phospholipids on transcriptional gene regulation in human umbilical vein endothelial cells and aortic endothelial cells with a special focus on miRNAs. METHODS AND RESULTS: We integrated data from HiC, ChIP-seq, ATAC-seq, GRO-seq, miRNA-seq, and RNA-seq to provide deeper understanding of the transcriptional mechanisms driven by NRF2 in response to oxidized phospholipids. We demonstrate that presence of NRF2 motif and its binding is more prominent in the vicinity of up-regulated transcripts and transcriptional initiation represents the most likely mechanism of action. We further identified NRF2 as a novel regulator of over 100 endothelial pri-miRNAs. Among these, we characterize two hub miRNAs miR-21-5p and miR-100-5p and demonstrate their opposing roles on mTOR, VEGFA, HIF1A, and MYC expressions. Finally, we provide evidence that the levels of miR-21-5p and miR-100-5p in exosomes are increased upon senescence and exhibit a trend to correlate with the severity of coronary artery disease. CONCLUSION: Altogether, our analysis provides an integrative view into the regulation of transcription and miRNA function that could mediate the proatherogenic effects of oxidized phospholipids in endothelial cells.


Subject(s)
Atherosclerosis/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , MicroRNAs/metabolism , NF-E2-Related Factor 2/metabolism , Phosphatidylcholines/toxicity , Transcriptome , Atherosclerosis/genetics , Atherosclerosis/pathology , Cells, Cultured , Cellular Senescence , Databases, Genetic , Gene Expression Profiling , Gene Expression Regulation , Gene Regulatory Networks , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/pathology , Humans , MicroRNAs/genetics , NF-E2-Related Factor 2/genetics , Oxidation-Reduction , Plaque, Atherosclerotic
4.
Int J Mol Sci ; 20(21)2019 Oct 29.
Article in English | MEDLINE | ID: mdl-31671916

ABSTRACT

We developed a pipeline for the discovery of transcriptomics-derived disease-modifying therapies and used it to validate treatments in vitro and in vivo that could be repurposed for TBI treatment. Desmethylclomipramine, ionomycin, sirolimus and trimipramine, identified by in silico LINCS analysis as candidate treatments modulating the TBI-induced transcriptomics networks, were tested in neuron-BV2 microglial co-cultures, using tumour necrosis factor α as a monitoring biomarker for neuroinflammation, nitrite for nitric oxide-mediated neurotoxicity and microtubule associated protein 2-based immunostaining for neuronal survival. Based on (a) therapeutic time window in silico, (b) blood-brain barrier penetration and water solubility, (c) anti-inflammatory and neuroprotective effects in vitro (p < 0.05) and (d) target engagement of Nrf2 target genes (p < 0.05), desmethylclomipramine was validated in a lateral fluid-percussion model of TBI in rats. Despite the favourable in silico and in vitro outcomes, in vivo assessment of clomipramine, which metabolizes to desmethylclomipramine, failed to demonstrate favourable effects on motor and memory tests. In fact, clomipramine treatment worsened the composite neuroscore (p < 0.05). Weight loss (p < 0.05) and prolonged upregulation of plasma cytokines (p < 0.05) may have contributed to the worsened somatomotor outcome. Our pipeline provides a rational stepwise procedure for evaluating favourable and unfavourable effects of systems-biology discovered compounds that modulate post-TBI transcriptomics.


Subject(s)
Brain Injuries, Traumatic/drug therapy , Disease , Systems Biology/methods , Animals , Anti-Inflammatory Agents/pharmacology , Biomarkers , Cell Line , Clomipramine/analogs & derivatives , Clomipramine/metabolism , Clomipramine/pharmacology , Coculture Techniques , Cytokines/blood , Gene Expression , In Vitro Techniques , Ionomycin/pharmacology , Machine Learning , Male , Microglia/drug effects , Microglia/metabolism , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Neurons/drug effects , Neurons/metabolism , Neuroprotection , Neuroprotective Agents/pharmacology , Nitrites/metabolism , Rats , Sirolimus/pharmacology , Transcriptome , Trimipramine/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation
5.
Redox Biol ; 21: 101050, 2019 02.
Article in English | MEDLINE | ID: mdl-30654300

ABSTRACT

Cells evolved robust homeostatic mechanisms to protect against oxidation or alkylation by electrophilic species. Glutathione (GSH) is the most abundant intracellular thiol, protects cellular components from oxidation and is maintained in a reduced state by glutathione reductase (GR). Nitro oleic acid (NO2-OA) is an electrophilic fatty acid formed under digestive and inflammatory conditions that both reacts with GSH and induces its synthesis upon activation of Nrf2 signaling. The effects of NO2-OA on intracellular GSH homeostasis were evaluated. In addition to upregulation of GSH biosynthesis, we observed that NO2-OA increased intracellular GSSG in an oxidative stress-independent manner. NO2-OA directly inhibited GR in vitro by covalent modification of the catalytic Cys61, with kon of (3.45 ± 0.04) × 103 M-1 s-1, koff of (4.4 ± 0.4) × 10-4 s-1, and Keq of (1.3 ± 0.1) × 10-7 M. Akin to NO2-OA, the electrophilic Nrf2 activators bardoxolone-imidazole (CDDO-Im), bardoxolone-methyl (CDDO-Me) and dimethyl fumarate (DMF) also upregulated GSH biosynthesis while promoting GSSG accumulation, but without directly inhibiting GR activity. In vitro assays in which GR was treated with increasing GSH concentrations and GSH depletion experiments in cells revealed that GR activity is finely regulated via product inhibition, an observation further supported by theoretical (kinetic modeling of cellular GSSG:GSH levels) approaches. Together, these results describe two independent mechanisms by which electrophiles modulate the GSH/GSSG couple, and provide a novel conceptual framework to interpret experimentally determined values of GSH and GSSG.


Subject(s)
Glutathione Reductase/chemistry , Glutathione Reductase/metabolism , Glutathione/biosynthesis , Algorithms , Alkylation , Amino Acid Sequence , Animals , Catalysis , Catalytic Domain , Glutathione Disulfide/metabolism , Intracellular Space , Kinetics , Mice , Models, Theoretical , Multidrug Resistance-Associated Proteins/antagonists & inhibitors , Multidrug Resistance-Associated Proteins/metabolism , NF-E2-Related Factor 2/metabolism , Nitric Oxide/metabolism , Oxidation-Reduction , Oxidative Stress , RAW 264.7 Cells , Reactive Oxygen Species , Sulfhydryl Compounds
6.
Cardiovasc Res ; 115(1): 243-254, 2019 01 01.
Article in English | MEDLINE | ID: mdl-29917052

ABSTRACT

Aims: Oxidative stress and inflammation play an important role in the progression of atherosclerosis. Transcription factor NF-E2-related factor 2 (Nrf2) has antioxidant and anti-inflammatory effects in the vessel wall, but paradoxically, global loss of Nrf2 in apoE deficient mice alleviates atherosclerosis. In this study, we investigated the effect of global Nrf2 deficiency on early and advanced atherogenesis in alternative models of atherosclerosis, LDL receptor deficient mice (LDLR-/-), and LDLR-/- mice expressing apoB-100 only (LDLR-/- ApoB100/100) having a humanized lipoprotein profile. Methods and results: LDLR-/- mice were fed a high-fat diet (HFD) for 6 or 12 weeks and LDLR-/-ApoB100/100 mice a regular chow diet for 6 or 12 months. Nrf2 deficiency significantly reduced early and more advanced atherosclerosis assessed by lesion size and coverage in the aorta in both models. Nrf2 deficiency in LDLR-/- mice reduced total plasma cholesterol after 6 weeks of HFD and triglycerides in LDLR-/-ApoB100/100 mice on a chow diet. Nrf2 deficiency aggravated aortic plaque maturation in aged LDLR-/-ApoB100/100 mice as it increased plaque calcification. Moreover, ∼36% of Nrf2-/-LDLR-/-ApoB100/100 females developed spontaneous myocardial infarction (MI) or sudden death at 5 to 12 months of age. Interestingly, Nrf2 deficiency increased plaque instability index, enhanced plaque inflammation and calcification, and reduced fibrous cap thickness in brachiocephalic arteries of LDLR-/-ApoB100/100 female mice at age of 12 months. Conclusions: Absence of Nrf2 reduced atherosclerotic lesion size in both atherosclerosis models, likely via systemic effects on lipid metabolism. However, Nrf2 deficiency in aged LDLR-/-ApoB100/100 mice led to an enhanced atherosclerotic plaque instability likely via increased plaque inflammation and oxidative stress, which possibly predisposed to MI and sudden death.


Subject(s)
Aorta/metabolism , Aortic Diseases/metabolism , Atherosclerosis/metabolism , Hypercholesterolemia/complications , NF-E2-Related Factor 2/deficiency , Plaque, Atherosclerotic , Age Factors , Animals , Aorta/pathology , Aortic Diseases/etiology , Aortic Diseases/pathology , Aortic Diseases/prevention & control , Apolipoprotein B-100/genetics , Apolipoprotein B-100/metabolism , Atherosclerosis/etiology , Atherosclerosis/pathology , Atherosclerosis/prevention & control , Cells, Cultured , Diet, High-Fat , Disease Models, Animal , Disease Progression , Female , Hypercholesterolemia/genetics , Inflammation Mediators/metabolism , Macrophages/metabolism , Macrophages/pathology , Male , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout, ApoE , Myocardial Infarction/etiology , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , NF-E2-Related Factor 2/genetics , Oxidative Stress , Receptors, LDL/deficiency , Receptors, LDL/genetics , Triglycerides/blood
7.
Redox Biol ; 18: 77-83, 2018 09.
Article in English | MEDLINE | ID: mdl-29986211

ABSTRACT

Oxidative stress predisposes to several aging-associated diseases, such as cardiovascular diseases and cancer. In aging, increase in the production of reactive oxygen species is typically accompanied with a decline in adaptive stress responses to oxidative stress. The decline is primarily due to a decrease in antioxidant production. Nuclear factor E2-Related Factor 2 (NRF2) is a key transcription factor regulating oxidative and electrophilic stress responses, but it has also been shown to play a role in the regulation of cell metabolism. NRF2 expression declines in aging, but the mechanisms remain unclear. In this study, we show that microRNAs (miRNAs) that are abundant in old endothelial cells decrease NRF2 expression by direct targeting of NRF2 mRNA. The effect is reversed by miRNA inhibition. The senescence-associated downregulation of NRF2 decreases endothelial glycolytic activity and stress tolerance both of which are restored after reinstating NRF2. Manipulation of the senescence-associated miRNA levels affects the glycolytic activity and stress tolerance consistently with the NRF2 results. We conclude that senescence-associated miRNAs are involved in the decline of NRF2 expression, thus contributing to the repression of adaptive responses during cell senescence.


Subject(s)
Cellular Senescence , Endothelial Cells/cytology , MicroRNAs/genetics , NF-E2-Related Factor 2/genetics , Cell Proliferation , Down-Regulation , Endothelial Cells/metabolism , Gene Expression Regulation , Glycolysis , Human Umbilical Vein Endothelial Cells , Humans , MicroRNAs/metabolism , NF-E2-Related Factor 2/metabolism , Oxidative Stress
8.
Nucleic Acids Res ; 46(3): 1124-1138, 2018 02 16.
Article in English | MEDLINE | ID: mdl-29161413

ABSTRACT

Phospholipids, such as 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphocholine (PAPC), are the major components of cell membranes. Their exposure to reactive oxygen species creates oxidized phospholipids, which predispose to the development of chronic inflammatory diseases and metabolic disorders through endothelial activation and dysfunction. Although the effects of oxidized PAPC (oxPAPC) on endothelial cells have been previously studied, the underlying molecular mechanisms evoking biological responses remain largely unknown. Here, we investigated the molecular mechanisms of oxPAPC function with a special emphasis on NRF2-regulated microRNAs (miRNAs) in human umbilical vein endothelial cells (HUVECs) utilizing miRNA profiling, global run-on sequencing (GRO-seq), genome-wide NRF2 binding model, and RNA sequencing (RNA-seq) with miRNA overexpression and silencing. We report that the central regulators of endothelial activity, KLF2 for quiescence, PFKFB3 for glycolysis, and VEGFA, FOXO1 and MYC for growth and proliferation, are regulated by transcription factor NRF2 and the NRF2-regulated miR-106b∼25 cluster member, miR-93, in HUVECs. Mechanistically, oxPAPC was found to induce glycolysis and proliferation NRF2-dependently, and oxPAPC-dependent induction of the miR-106b∼25 cluster was mediated by NRF2. Additionally, several regulatory loops were established between NRF2, miR-93 and the essential regulators of healthy endothelium, collectively implying that NRF2 controls the switch between the quiescent and the proliferative endothelial states together with miR-93.


Subject(s)
Glycolysis/drug effects , Kruppel-Like Transcription Factors/genetics , MicroRNAs/genetics , NF-E2-Related Factor 2/genetics , Phosphatidylcholines/pharmacology , Phosphofructokinase-2/genetics , Antagomirs/genetics , Antagomirs/metabolism , Cell Proliferation/drug effects , Forkhead Box Protein O1/genetics , Forkhead Box Protein O1/metabolism , Gene Expression Profiling , Gene Expression Regulation , Glycolysis/genetics , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Kruppel-Like Transcription Factors/metabolism , MicroRNAs/antagonists & inhibitors , MicroRNAs/metabolism , NF-E2-Related Factor 2/metabolism , Oxidation-Reduction , Phosphatidylcholines/chemistry , Phosphatidylcholines/metabolism , Phosphofructokinase-2/metabolism , Primary Cell Culture , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Reactive Oxygen Species/metabolism , Sequence Analysis, RNA , Signal Transduction , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
9.
Sci Rep ; 7(1): 10943, 2017 09 08.
Article in English | MEDLINE | ID: mdl-28887500

ABSTRACT

Endothelial plasticity enables the cells to switch their phenotype according to the surrounding vascular microenvironment. MicroRNAs (miRNAs) are small noncoding RNAs that control endothelial plasticity. The objective of this study was to investigate the differences in miRNA profiles of tissue-derived cells and cultured endothelial cells. To this end, miRNA expression was profiled from freshly isolated tissue-derived human vascular endothelial cells and endothelial cells cultured until cellular senescence using miRNA sequencing. In addition, the data was searched for putative novel endothelial miRNAs and miRNA isoforms. The data analysis revealed a striking change in endothelial miRNA profile as the cells adapted from tissue to cell culture environment and the overall miRNA expression decreased significantly in cultured compared to tissue-derived endothelial cells. In addition to changes in mechanosensitive miRNA expression, alterations in senescence-associated and endothelial-to-mesenchymal-transition-associated miRNAs were observed in aging cells. Collectively, the data illustrates the adaptability of endothelial cell miRNA expression that mirrors prevailing cellular environment.


Subject(s)
Human Umbilical Vein Endothelial Cells/metabolism , MicroRNAs/genetics , Cellular Senescence , Gene Expression Profiling , Human Umbilical Vein Endothelial Cells/physiology , Humans , Mechanotransduction, Cellular , MicroRNAs/metabolism , Transcriptome
10.
Mol Pharmacol ; 92(4): 481-490, 2017 10.
Article in English | MEDLINE | ID: mdl-28778983

ABSTRACT

Nitro-fatty acids are reactive signaling mediators that are formed when unsaturated fatty acids react with nitric oxide or nitric oxide-derived species. Nitro-fatty acids can modify specific signaling pathways via post-translational modifications of Cys residues in key regulatory proteins. One of the signaling cascades activated by nitro-fatty acids is the Keap1-Nrf2 pathway. We have previously studied the effects of nitro-oleic acid (OA-NO2) on the human endothelial cell transcriptome. We observed that endothelin receptor B [ET-B (gene name EDNRB)], the receptor mediating the vasodilatory effects of endothelin-1 (ET-1) is induced by OA-NO2 Inasmuch as ET-1 is one of the key regulators of vascular tone, we chose to examine in more detail the effect of OA-NO2 on endothelin signaling in human endothelial cells. Nrf2 was found to regulate the OA-NO2-induced transcription of ET-B in human and mouse endothelial cells. Furthermore, chromatin immunoprecipitation analysis revealed that OA-NO2 increased the binding of Nrf2 to an antioxidant response element in the enhancer region of the EDNRB gene. In addition, we show that the overexpression of both OA-NO2 and Nrf2 substantially decreased and that Nrf2 silencing increased the ET-1 concentration in the culture media of endothelial cells. The change in the extracellular ET-1 concentration was dependent on ET-B receptor expression. These data suggest that OA-NO2 modulates endothelin signaling by increasing Nrf2-dependent expression of the ET-B receptor in endothelial cells, which in turn mediates the decrease in extracellular ET-1 concentration. Based on these results, we propose that OA-NO2 and Nrf2 may alleviate the vasoconstrictive effects of ET-1 by removing it from the circulation.


Subject(s)
Endothelial Cells/metabolism , Endothelin-1/metabolism , Nitrogen Dioxide/pharmacology , Oleic Acid/pharmacology , Signal Transduction/physiology , Animals , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Endothelin-1/antagonists & inhibitors , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Mice , Mice, Knockout , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Signal Transduction/drug effects
11.
Nucleic Acids Res ; 44(4): 1760-75, 2016 Feb 29.
Article in English | MEDLINE | ID: mdl-26826707

ABSTRACT

Transcription factor binding specificity is crucial for proper target gene regulation. Motif discovery algorithms identify the main features of the binding patterns, but the accuracy on the lower affinity sites is often poor. Nuclear factor E2-related factor 2 (NRF2) is a ubiquitous redox-activated transcription factor having a key protective role against endogenous and exogenous oxidant and electrophile stress. Herein, we decipher the effects of sequence variation on the DNA binding sequence of NRF2, in order to identify both genome-wide binding sites for NRF2 and disease-associated regulatory SNPs (rSNPs) with drastic effects on NRF2 binding. Interactions between NRF2 and DNA were studied using molecular modelling, and NRF2 chromatin immunoprecipitation-sequence datasets together with protein binding microarray measurements were utilized to study binding sequence variation in detail. The binding model thus generated was used to identify genome-wide binding sites for NRF2, and genomic binding sites with rSNPs that have strong effects on NRF2 binding and reside on active regulatory elements in human cells. As a proof of concept, miR-126-3p and -5p were identified as NRF2 target microRNAs, and a rSNP (rs113067944) residing on NRF2 target gene (Ferritin, light polypeptide, FTL) promoter was experimentally verified to decrease NRF2 binding and result in decreased transcriptional activity.


Subject(s)
Genome, Human , MicroRNAs/genetics , NF-E2-Related Factor 2/genetics , Transcription, Genetic , Algorithms , Binding Sites , Gene Expression Regulation , Humans , MicroRNAs/metabolism , NF-E2-Related Factor 2/metabolism , Polymorphism, Single Nucleotide/genetics , Promoter Regions, Genetic , Protein Binding
12.
Biochem Soc Trans ; 43(4): 645-9, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26551706

ABSTRACT

Accumulating evidence suggests that dysregulation of the Kelch-like ECH-associated protein 1 (Keap1)-nuclear factor E2-related factor 2 (Nrf2) pathway resulting in constitutively active Nrf2 and increased expression of cytoprotective Nrf2 target genes, has a pivotal role in cancer. Cancer cells are able to hijack the Keap1-Nrf2 system via multiple mechanisms leading to enhanced chemo- and radio-resistance and proliferation via metabolic reprogramming as well as inhibition of apoptosis. In this mini-review, we will describe the mechanisms leading to increased Nrf2 activity in cancer with a focus on the information achieved from large-scale multi-omics projects across various cancer types.


Subject(s)
Gene Expression Regulation, Neoplastic , Intracellular Signaling Peptides and Proteins/genetics , NF-E2-Related Factor 2/genetics , Neoplasms/genetics , Apoptosis , Cell Proliferation , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/radiation effects , Genomics , Humans , Kelch-Like ECH-Associated Protein 1 , Radiation Tolerance
13.
Nucleic Acids Res ; 42(20): 12570-84, 2014 Nov 10.
Article in English | MEDLINE | ID: mdl-25352550

ABSTRACT

Vascular endothelial growth factor A (VEGF-A) is a master regulator of angiogenesis, vascular development and function. In this study we investigated the transcriptional regulation of VEGF-A-responsive genes in primary human aortic endothelial cells (HAECs) and human umbilical vein endothelial cells (HUVECs) using genome-wide global run-on sequencing (GRO-Seq). We demonstrate that half of VEGF-A-regulated gene promoters are characterized by a transcriptionally competent paused RNA polymerase II (Pol II). We show that transition into productive elongation is a major mechanism of gene activation of virtually all VEGF-regulated genes, whereas only ∼40% of the genes are induced at the level of initiation. In addition, we report a comprehensive chromatin interaction map generated in HUVECs using tethered conformation capture (TCC) and characterize chromatin interactions in relation to transcriptional activity. We demonstrate that sites of active transcription are more likely to engage in chromatin looping and cell type-specific transcriptional activity reflects the boundaries of chromatin interactions. Furthermore, we identify large chromatin compartments with a tendency to be coordinately transcribed upon VEGF-A stimulation. We provide evidence that these compartments are enriched for clusters of regulatory regions such as super-enhancers and for disease-associated single nucleotide polymorphisms (SNPs). Collectively, these findings provide new insights into mechanisms behind VEGF-A-regulated transcriptional programs in endothelial cells.


Subject(s)
Transcriptional Activation , Vascular Endothelial Growth Factor A/pharmacology , Cell Compartmentation , Cells, Cultured , Chromatin/metabolism , Enhancer Elements, Genetic , Genome, Human , Humans , Phenotype , Transcription Factors/metabolism , Transcription, Genetic/drug effects
14.
Eur J Clin Invest ; 44(10): 989-99, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25208310

ABSTRACT

BACKGROUND: Cardiovascular patients suffer from reduced blood flow leading to ischaemia and impaired tissue metabolism. Unfortunately, an increasing group of elderly patients cannot be treated with current revascularization methods. Thus, new treatment strategies are urgently needed. Hypoxia-inducible factors (HIFs) upregulate the expression of angiogenic mediators together with genes involved in energy metabolism and recovery of ischaemic tissues. Especially, HIF-2α is a novel factor, and only limited information is available about its therapeutic potential. METHODS: Gene transfers with adenoviral HIF-1α and HIF-2α were performed into the mouse heart and rabbit ischaemic hindlimbs. Angiogenesis was evaluated by histology. Left ventricle function was analysed with echocardiography. Perfusion in rabbit skeletal muscles and energy recovery after electrical stimulation-induced exercise were measured with ultrasound and (31)P-magnetic resonance spectroscopy ((31)P-MRS), respectively. RESULTS: HIF-1α and HIF-2α gene transfers increased capillary size up to fivefold in myocardium and ischaemic skeletal muscles. Perfusion in skeletal muscles was increased by fourfold without oedema. Especially, AdHIF-1α enhanced the recovery of ischaemic muscles from electrical stimulation-induced energy depletion. Special characteristic of HIF-2α gene transfer was a strong capillary growth in muscle connective tissue and that HIF-2α gene transfer maintained left ventricle function. CONCLUSIONS: We conclude that both AdHIF-1α and AdHIF-2α gene transfers induced beneficial angiogenesis in vivo. Transient moderate increases in angiogenesis improved energy recovery after exercise in ischaemic muscles. This study shows for the first time that a moderate increase in angiogenesis is enough to improve tissue energy metabolism, which is potentially a very useful feature for cardiovascular gene therapy.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/pharmacology , Muscle, Skeletal/metabolism , Neovascularization, Physiologic/drug effects , Animals , Capillaries/physiology , Coronary Vessels/physiology , Gene Expression/physiology , Gene Transfer Techniques , Genetic Therapy/methods , Hindlimb/blood supply , Ischemia/physiopathology , Ischemia/therapy , Mice, Inbred C57BL , Muscle, Skeletal/blood supply , Myocardium/metabolism , Rabbits
15.
Mol Pharmacol ; 86(5): 522-35, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25140003

ABSTRACT

The primary route of inactivation of the endocannabinoid 2-arachidonoylglycerol in the central nervous system is through enzymatic hydrolysis, mainly carried out by monoacylglycerol lipase (MAGL), along with a small contribution by the α/ß-hydrolase domain (ABHD) proteins ABHD6 and ABHD12. Recent methodological progress allowing kinetic monitoring of glycerol liberation has facilitated substrate profiling of the human endocannabinoid hydrolases, and these studies have revealed that the three enzymes have distinct monoacylglycerol substrate and isomer preferences. Here, we have extended this substrate profiling to cover four prostaglandin glycerol esters, namely, 15-deoxy-Δ(12,14)-prostaglandin J2-2-glycerol (15d-PGJ2-G), PGD2-G, PGE2-G, and PGF2 α-G. We found that the three enzymes hydrolyzed the tested substrates, albeit with distinct rates and preferences. Although human ABHD12 (hABHD12) showed only marginal activity toward PGE2-G, hABHD6 preferentially hydrolyzed PGD2-G, and human MAGL (hMAGL) robustly hydrolyzed all four. This was particularly intriguing for MAGL activity toward 15d-PGJ2-G whose hydrolysis rate rivaled that of the best monoacylglycerol substrates. Molecular modeling studies combined with kinetic analysis supported favorable interaction with the hMAGL active site. Long and short MAGL isoforms shared a similar substrate profile, and hMAGL hydrolyzed 15d-PGJ2-G also in living cells. The ability of 15d-PGJ2-G to activate the canonical nuclear factor erythroid 2-related factor (Nrf2) signaling pathway used by 15d-PGJ2 was assessed, and these studies revealed for the first time that 15d-PGJ2 and 15d-PGJ2-G similarly activated Nrf2 signaling as well as transcription of target genes of this pathway. Our study challenges previous claims regarding the ability of MAGL to catalyze PG-G hydrolysis and extend the MAGL substrate profile beyond the classic monoacylglycerols.


Subject(s)
Esters/metabolism , Glycerol/metabolism , Monoacylglycerol Lipases/metabolism , Prostaglandins/metabolism , Catalytic Domain/physiology , Cells, Cultured , Endocannabinoids/metabolism , HEK293 Cells , Human Umbilical Vein Endothelial Cells , Humans , Hydrolases/metabolism , Hydrolysis , Kinetics , Monoglycerides/metabolism , NF-E2-Related Factor 2/metabolism , Prostaglandin D2/analogs & derivatives , Prostaglandin D2/metabolism , Protein Isoforms/metabolism , Signal Transduction/physiology
16.
Adv Cancer Res ; 122: 281-320, 2014.
Article in English | MEDLINE | ID: mdl-24974185

ABSTRACT

The Kelch-like ECH-associated protein 1 (Keap1)-nuclear factor E2-related factor 2 (Nrf2) pathway is one of the major signaling cascades involved in cell defense and survival against endogenous and exogenous stress. While Nrf2 and its target genes provide protection against various age-related diseases including tumorigenesis, constitutively active Nrf2 in cancer cells increases the expression of cytoprotective genes and, consequently, enhances proliferation via metabolic reprogramming and inhibition of apoptosis. Herein, we review the current understanding of the regulation of Nrf2 in normal cells as well as its dual role in cancer. Furthermore, the mechanisms of Nrf2 dysregulation in cancer, consequences of unchecked Nrf2 activity, and therapies targeting the Keap1-Nrf2 system are discussed.


Subject(s)
Gene Expression Regulation, Neoplastic , Intracellular Signaling Peptides and Proteins/metabolism , NF-E2-Related Factor 2/metabolism , Neoplasms/metabolism , Animals , Antioxidants/metabolism , Apoptosis , Cell Proliferation , Cell Transformation, Neoplastic , Drug Resistance, Neoplasm , Humans , Kelch-Like ECH-Associated Protein 1 , Neoplasms/therapy , Oxidative Stress , Prognosis , Signal Transduction
17.
Free Radic Biol Med ; 71: 196-207, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24681256

ABSTRACT

Redox networks in the cell integrate signaling pathways that control metabolism, energetics, cell survival, and death. The physiological second messengers that modulate these pathways include nitric oxide, hydrogen peroxide, and electrophiles. Electrophiles are produced in the cell via both enzymatic and nonenzymatic lipid peroxidation and are also relatively abundant constituents of the diet. These compounds bind covalently to families of cysteine-containing, redox-sensing proteins that constitute the electrophile-responsive proteome, the subproteomes of which are found in localized intracellular domains. These include those proteins controlling responses to oxidative stress in the cytosol-notably the Keap1-Nrf2 pathway, the autophagy-lysosomal pathway, and proteins in other compartments including mitochondria and endoplasmic reticulum. The signaling pathways through which electrophiles function have unique characteristics that could be exploited for novel therapeutic interventions; however, development of such therapeutic strategies has been challenging due to a lack of basic understanding of the mechanisms controlling this form of redox signaling. In this review, we discuss current knowledge of the basic mechanisms of thiol-electrophile signaling and its potential impact on the translation of this important field of redox biology to the clinic. Emerging understanding of thiol-electrophile interactions and redox signaling suggests replacement of the oxidative stress hypothesis with a new redox biology paradigm, which provides an exciting and influential framework for guiding translational research.


Subject(s)
Antioxidants/metabolism , Autophagy/genetics , Oxidative Stress/genetics , Aldehydes/metabolism , Animals , Antioxidants/therapeutic use , Gene Expression Regulation , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Hydrogen Peroxide/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Kelch-Like ECH-Associated Protein 1 , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Nitric Oxide/metabolism , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Signal Transduction , Unfolded Protein Response/genetics
18.
Redox Biol ; 1: 45-9, 2013 Jan 18.
Article in English | MEDLINE | ID: mdl-24024136

ABSTRACT

The Keap1-Nrf2 pathway is the major regulator of cytoprotective responses to oxidative and electrophilic stress. Although cell signaling pathways triggered by the transcription factor Nrf2 prevent cancer initiation and progression in normal and premalignant tissues, in fully malignant cells Nrf2 activity provides growth advantage by increasing cancer chemoresistance and enhancing tumor cell growth. In this graphical review, we provide an overview of the Keap1-Nrf2 pathway and its dysregulation in cancer cells. We also briefly summarize the consequences of constitutive Nrf2 activation in cancer cells and how this can be exploited in cancer gene therapy.


Subject(s)
Cytoskeletal Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , NF-E2-Related Factor 2/metabolism , Neoplasms/metabolism , Animals , Humans , Neoplasms/pathology , Neoplasms/therapy
19.
Cardiovasc Res ; 98(1): 107-15, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23341579

ABSTRACT

AIMS: The loss of nuclear factor E2-related factor 2 (Nrf2) has been shown to protect against atherogenesis in apoE-deficient mice. The mechanism by which Nrf2 deficiency affords atheroprotection in this model is currently unknown, but combined systemic and local vascular effects on lesion macrophages have been proposed. We investigated the effect of bone marrow-specific loss of Nrf2 on early atherogenesis in low-density lipoprotein (LDL) receptor-deficient (LDLR(-/-)) mice, and assessed the effect of Nrf2 on cellular accumulation of modified LDLs and the expression of inflammatory markers in macrophages. METHODS AND RESULTS: The effect of bone marrow-specific loss of Nrf2 on atherogenesis was studied using bone marrow transplantation of wild-type (WT) or Nrf2(-/-) bone marrow to LDLR(-/-) mice. Mice transplanted with Nrf2(-/-) bone marrow and fed a high-fat diet for 6 weeks exhibited significantly larger atherosclerotic lesions than WT bone marrow transplanted mice. Moreover, in thioglycollate-elicited Nrf2(-/-) macrophages, the uptake of acetylated and malondialdehyde-modified LDLs was increased in comparison with WT controls, with the concomitant increase in the expression of scavenger receptor A and toll-like receptor 4. In addition, the expression of pro-inflammatory monocyte chemoattractant protein-1 and interleukin-6 were increased in Nrf2(-/-) vs. WT macrophages. CONCLUSION: Nrf2 deficiency specific to bone marrow-derived cells aggravates atherosclerosis in LDLR(-/-) mice. Furthermore, the loss of Nrf2 in macrophages enhances foam cell formation and promotes the pro-inflammatory phenotype.


Subject(s)
Atherosclerosis/etiology , Macrophages/physiology , NF-E2-Related Factor 2/physiology , Animals , Chemokine CCL2/genetics , Cholesterol/metabolism , Female , Mice , Mice, Inbred C57BL , Receptors, LDL/physiology , Receptors, Scavenger/analysis
20.
Cancer Res ; 72(23): 6227-35, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-23041549

ABSTRACT

Nuclear factor erythroid-2 related factor 2 (Nrf2) is a transcription factor that regulates protection against a wide variety of toxic insults to cells, including cytotoxic cancer chemotherapeutic drugs. Many lung cancer cells harbor a mutation in either Nrf2 or its inhibitor Keap1 resulting in permanent activation of Nrf2 and chemoresistance. In this study, we sought to examine whether this attribute could be exploited in cancer suicide gene therapy by using a lentiviral (LV) vector expressing herpes simplex virus thymidine kinase (HSV-TK/GCV) under the regulation of antioxidant response element (ARE), a cis-acting enhancer sequence that binds Nrf2. In human lung adenocarcinoma cells in which Nrf2 is constitutively overexpressed, ARE activity was found to be high under basal conditions. In this setting, ARE-HSV-TK was more effective than a vector in which HSV-TK expression was driven by a constitutively active promoter. In a mouse xenograft model of lung cancer, suicide gene therapy with LV-ARE-TK/GCV was effective compared with LV-PGK-TK/GCV in reducing tumor size. We conclude that ARE-regulated HSV-TK/GCV therapy offers a promising approach for suicide cancer gene therapy in cells with high constitutive ARE activity, permitting a greater degree of therapeutic targeting to those cells.


Subject(s)
Adenocarcinoma/therapy , Antioxidant Response Elements , Ganciclovir/pharmacology , Genetic Therapy/methods , Lung Neoplasms/therapy , Oxidative Stress/physiology , Thymidine Kinase/metabolism , Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma of Lung , Animals , Cell Line, Tumor , Ganciclovir/pharmacokinetics , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Kelch-Like ECH-Associated Protein 1 , Lentivirus/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Male , Mice , Mice, Nude , Mutation , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Thymidine Kinase/biosynthesis , Thymidine Kinase/genetics , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...