Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
J Biol Chem ; 289(3): 1866-75, 2014 Jan 17.
Article in English | MEDLINE | ID: mdl-24302727

ABSTRACT

The aryl hydrocarbon receptor (AhR) is involved in the regulation of immune responses, T-cell differentiation, and immunity. Here, we show that inflammatory stimuli such as LPS induce the expression of AhR in human dendritic cells (DC) associated with an AhR-dependent increase of CYP1A1 (cytochrome P4501A1). In vivo data confirmed the elevated expression of AhR by LPS and the LPS-enhanced 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)-mediated induction of CYP1A1 in thymus of B6 mice. Inhibition of nuclear factor-κB (NF-κB) repressed both normal and LPS-enhanced, TCDD-inducible, AhR-dependent gene expression and canonical pathway control of RelA-regulated AhR-responsive gene expression. LPS-mediated induction of AhR was NF-κB-dependent, as shown in mouse embryonic fibroblasts (MEFs) derived from Rel null mice. AhR expression and TCDD-mediated induction of CYP1A1 was significantly reduced in RelA-deficient MEF compared with wild type MEF cells and ectopic expression of RelA restored the expression of AhR and induction of CYP1A1 in MEF RelA null cells. Promoter analysis of the human AhR gene identified three putative NF-κB-binding elements upstream of the transcription start site. Mutation analysis of the AhR promoter identified one NF-κB site as responsible for mediating the induction of AhR expression by LPS and electrophoretic shift assays demonstrated that this NF-κB motif is recognized by the RelA/p50 heterodimer. Our results show for the first time that NF-κB RelA is a critical component regulating the expression of AhR and the induction of AhR-dependent gene expression in immune cells illustrating the interaction of AhR and NF-κB signaling.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Dendritic Cells/metabolism , Receptors, Aryl Hydrocarbon/biosynthesis , Signal Transduction , Transcription Factor RelA/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Line , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP1A1/metabolism , Dendritic Cells/pathology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Humans , Inflammation/chemically induced , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Lipopolysaccharides/toxicity , Mice , Mice, Knockout , Mutation , Polychlorinated Dibenzodioxins/analogs & derivatives , Polychlorinated Dibenzodioxins/toxicity , Receptors, Aryl Hydrocarbon/genetics , Response Elements , Transcription Factor RelA/genetics , Transcription Factor RelA/immunology
2.
PLoS One ; 6(8): e23240, 2011.
Article in English | MEDLINE | ID: mdl-21853092

ABSTRACT

Crystallographic studies have offered understanding of how receptor tyrosine kinases from the ErbB family are regulated by their growth factor ligands. A conformational change of the EGFR (ErbB1) was shown to occur upon ligand binding, where a solely ligand-mediated mode of dimerization/activation was documented. However, this dogma of dimerization/activation was revolutionized by the discovery of constitutively active ligand-independent EGFR mutants. In addition, other ligand-independent activation mechanisms may occur. We have shown that oxidative stress (ox-stress), induced by hydrogen peroxide or cigarette smoke, activates EGFR differently than its ligand, EGF, thereby inducing aberrant phosphorylation and impaired trafficking and degradation of EGFR. Here we demonstrate that ox-stress activation of EGFR is ligand-independent, does not induce "classical" receptor dimerization and is not inhibited by the tyrosine kinase inhibitor AG1478. Thus, an unprecedented, apparently activated, state is found for EGFR under ox-stress. Furthermore, this activation mechanism is temperature-dependent, suggesting the simultaneous involvement of membrane structure. We propose that ceramide increase under ox-stress disrupts cholesterol-enriched rafts leading to EGFR re-localization into the rigid, ceramide-enriched rafts. This increase in ceramide also supports EGFR aberrant trafficking to a peri-nuclear region. Therefore, the EGFR unprecedented and activated conformation could be sustained by simultaneous alterations in membrane structure under ox-stress.


Subject(s)
ErbB Receptors/chemistry , ErbB Receptors/metabolism , Oxidative Stress , Protein Multimerization/drug effects , Animals , Cell Membrane/drug effects , Cell Membrane/metabolism , Ceramides/metabolism , Cholesterol/metabolism , Enzyme Activation/drug effects , Humans , Hydrogen Peroxide/pharmacology , Ligands , Mice , Models, Biological , NIH 3T3 Cells , Oxidative Stress/drug effects , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Structure, Tertiary , Protein Transport/drug effects , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Quinazolines , Temperature , Tyrphostins/pharmacology , src-Family Kinases/metabolism
3.
Int J Biochem Cell Biol ; 43(7): 1065-70, 2011 Jul.
Article in English | MEDLINE | ID: mdl-19497383

ABSTRACT

Scavenger Receptor B1 has been shown to play a prominent role in the uptake and delivery of vitamin E from HDL and is likely involved in regulating vitamin E in the lung. We have previously demonstrated that lung Scavenger Receptor B1 levels (protein and mRNA) are modulated by cigarette smoke in mice and this was accompanied by changes in lung vitamin E. To further characterize the molecular mechanism(s) involved in this process, human alveolar epithelial cells were exposed to cigarette smoke and Scavenger Receptor B1 cellular levels and distribution were assessed. Results demonstrated that Scavenger Receptor B1 localizes in patches on the cellular membrane and in the per nuclear area of control cells. Upon cigarette smoke exposure, Scavenger Receptor B1 first translocated to the cell surface (within the first 12h of exposure) and then cell levels (protein and mRNA levels) decreased significantly at 24h. This decline was accompanied by increased Scavenger Receptor B1 ubiquitination which may explain the decrease in the protein levels. Cigarette smoke induced changes in both sub-cellular redistribution and ubiquitination of Scavenger Receptor B1 together with our previous in vivo data provides evidence that cigarette smoke exposure may alter lung's ability to control its tocopherol levels.


Subject(s)
Biological Transport/drug effects , Lung Neoplasms/etiology , Scavenger Receptors, Class B , Smoking/adverse effects , Tocopherols/metabolism , Ubiquitination/drug effects , Animals , Cell Line, Tumor , Cell Membrane/metabolism , Humans , Immunohistochemistry , Lung/cytology , Mice , Microscopy, Confocal , RNA, Messenger/genetics , RNA, Messenger/metabolism , Scavenger Receptors, Class B/genetics , Scavenger Receptors, Class B/metabolism , Time Factors
4.
Biophys J ; 95(8): 3559-62, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18641073

ABSTRACT

Apoptosis, or genetically programmed cell death, is a crucial cellular process that maintains the balance between life and death in cells. The precise molecular mechanism of apoptosis signaling and the manner in which type 1 and type 2 pathways of the apoptosis signaling network are differentially activated under distinct apoptotic stimuli is poorly understood. Based on Monte Carlo stochastic simulations, we show that the type 1 pathway becomes activated under strong apoptotic stimuli, whereas the type 2 mitochondrial pathway dominates apoptotic signaling in response to a weak death signal. Our results also show signaling in the type 2 pathway is stochastic; the population average over many cells does not capture the cell-to-cell fluctuations in the time course (approximately 1-10 h) of downstream caspase-3 activation. On the contrary, the probability distribution of caspase-3 activation for the mitochondrial pathway shows a distinct bimodal behavior that can be used to characterize the stochastic signaling in type 2 apoptosis and other similar complex signaling processes. Interestingly, such stochastic fluctuations in apoptosis signaling occur even in the presence of large numbers of signaling molecules.


Subject(s)
Apoptosis , Computer Simulation , Monte Carlo Method , Signal Transduction , Caspase 3/metabolism , Enzyme Activation , Humans , Models, Biological , Stochastic Processes , Time Factors
5.
FASEB J ; 22(3): 910-7, 2008 Mar.
Article in English | MEDLINE | ID: mdl-17971399

ABSTRACT

Exposure to hydrogen peroxide (H2O2), one of the reactive oxidants in the gas phase of cigarette smoke (CS), induces aberrant phosphorylation of the epidermal growth factor receptor (EGFR), resulting in the lack of ubiquitination by c-Cbl, and impaired degradation. EGFR activation without the feedback regulation of normal degradation leads to uncontrolled cell growth and tumor promotion. Using immunoprecipitation, immunoblotting, and confocal microscopy, we now demonstrate that the pattern of EGFR activation by CS is similar to H2O2. We found that exposure of human airway epithelial cells to CS, as with exposure to H2O2, not only results in an increase in EGFR activation over time, but the EGFR activated by H2O2 or CS is neither ubiquitinated nor subsequently degraded due to its inability to bind the E3 ubiquitin ligase, c-Cbl, either directly or indirectly via the Grb2 adapter protein. Moreover, the stabilized H2O2- and CS-activated EGFR remains plasma membrane-bound, while a population of the receptor is trafficked to a perinuclear region. Concomitantly, CS exposure results in the activation of downstream Akt and ERK1/2 survival and proliferation pathways. Therefore, exposure to CS, like exposure to H2O2, results in prolonged signaling by the EGFR and may contribute to uncontrolled lung cell growth.


Subject(s)
Cell Nucleus/metabolism , Epithelial Cells/drug effects , ErbB Receptors/metabolism , Hydrogen Peroxide/pharmacology , Nicotiana , Smoke , Cell Line, Tumor , Cell Transformation, Neoplastic , Dose-Response Relationship, Drug , Epithelial Cells/cytology , Epithelial Cells/metabolism , ErbB Receptors/drug effects , Humans , Hydrogen Peroxide/adverse effects , Lung/cytology , Phosphorylation/drug effects , Protein Transport/drug effects , Proto-Oncogene Proteins c-cbl/metabolism , Signal Transduction/drug effects , Time Factors
6.
J Biol Chem ; 281(20): 14486-93, 2006 May 19.
Article in English | MEDLINE | ID: mdl-16407214

ABSTRACT

The epidermal growth factor (EGF) receptor (EGFR) has been found to be overexpressed in several types of cancer cells, and the regulation of its oncogenic potential has been widely studied. The paradigm for EGFR down-regulation involves the trafficking of activated receptor molecules from the plasma membrane, through clathrin-coated pits, and into the cell for lysosomal degradation. We have previously shown that oxidative stress generated by H2O2 results in aberrant phosphorylation of the EGFR. This leads to the loss of c-Cbl-mediated ubiquitination of the EGFR and, consequently, prevents its degradation. However, we have found that c-Cbl-mediated ubiquitination is required solely for degradation but not for internalization of the EGFR under oxidative stress. To further examine the fate of the EGFR under oxidative stress, we used confocal analysis to show that the receptor not only remains co-localized with caveolin-1 at the plasma membrane, but at longer time points, is also sorted to a perinuclear compartment via a clathrin-independent, caveolae-mediated pathway. Our findings indicate that although the EGFR associates with caveolin-1 constitutively, caveolin-1 is hyperphosphorylated only under oxidative stress, which is essential in transporting the EGFR to a perinuclear location, where it is not degraded and remains active. Thus, oxidative stress may have a role in tumorigenesis by not only activating the EGFR but also by promoting prolonged activation of the receptor both at the plasma membrane and within the cell.


Subject(s)
Caveolin 1/metabolism , Cell Nucleus/metabolism , ErbB Receptors/physiology , src-Family Kinases/metabolism , Active Transport, Cell Nucleus , Cell Line, Tumor , Cell Membrane/metabolism , Clathrin/metabolism , Down-Regulation , Humans , Hydrogen Peroxide/metabolism , Lysosomes/metabolism , Models, Biological , Oxidative Stress , Signal Transduction
7.
Antioxid Redox Signal ; 7(1-2): 119-28, 2005.
Article in English | MEDLINE | ID: mdl-15650401

ABSTRACT

Reactive oxidants are associated with the pathogenesis of pulmonary diseases and affect various cell functions, from proliferation to apoptosis. We have shown that oxidants exert growth control on airway epithelial cells by modulating upstream receptor function. Additionally, hydrogen peroxide-mediated oxidative stress modulates ceramide levels to induce apoptosis in lung epithelium. Depletion of glutathione in lung epithelial cells results in ceramide accumulation, suggesting that ceramide elevation, coupled to oxidative stress, initiates apoptosis. While it is desirable to prevent cell death and tissue injury induced by oxidants in diseases such as asthma or acute respiratory distress syndrome, the opposite is sought in cancer. But oxidants may also activate growth factor receptors, enhancing cell proliferation and facilitating tumor promotion. Under oxidative stress, phosphorylation of the epidermal growth factor receptor (EGFR) is abrogated at tyrosine 1,045, the docking site for the ubiquitin ligase c-Cbl, rendering EGFR unable to recruit c-Cbl and be ubiquitylated and degraded. We thus proposed that this deficiency, which confers prolonged receptor signaling at the plasma membrane, links oxidative stress, EGFR, and tumorigenesis. Decoding the molecular interactions between oxidative stress and ceramide pathways and characterizing ubiquitylation control of receptor desensitization should provide new strategies for intervention in diverse pulmonary diseases and in diagnosing and eradicating cancer.


Subject(s)
Ceramides/metabolism , ErbB Receptors/metabolism , Oxidative Stress , Animals , Antioxidants/pharmacology , Apoptosis , Binding Sites , Caspase 3 , Caspases/metabolism , Cell Line, Tumor , Cell Membrane/metabolism , Cell Proliferation , Cell Survival , Enzyme Activation , Epithelial Cells/metabolism , Humans , Hydrogen Peroxide/metabolism , Hydrogen Peroxide/pharmacology , Ligands , Models, Biological , Neoplasms/metabolism , Oxidants/metabolism , Oxidants/pharmacology , Phosphorylation , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-cbl , Reactive Oxygen Species , Tyrosine/chemistry , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/metabolism
8.
J Biol Chem ; 279(35): 37153-62, 2004 Aug 27.
Article in English | MEDLINE | ID: mdl-15210722

ABSTRACT

Epidermal growth factor receptor (EGFR) controls cell growth and has a key role in tumorigenic processes. The extent of EGFR signaling is tightly regulated by post-transcriptional modifications leading to down-regulation of the levels of the receptor. Previous studies from our laboratory demonstrated that the reactive oxidant hydrogen peroxide activates the EGFR, yet, without down-regulation of the receptor levels, which results in prolonged receptor signaling. In the present study we examined the role of the E3 ligase c-Cbl, as a possible link between oxidative stress, EGFR signaling, and tumorigenic responses. First, we ectopically expressed a mutant EGFR (Tyr-1045 --> Phe) in cells lacking endogenous receptor, to determine whether the lack of phosphorylation at this site is the cause for EGFR retention at the membrane under oxidative stress, as we have previously suggested. Our findings suggest that abrogation of tyrosine 1045 phosphorylation alone is not enough to retain the EGFR at the plasma membrane under oxidative stress. Second, through the use of the Src inhibitor PP1, our findings establish EGFR movement out of the early endosomes as the exact location where c-Cbl-mediated ubiquitinylation is essential for EGFR trafficking. Finally, our studies substantiate the findings that c-Cbl-mediated ubiquitinylation is needed for degradation, but not for internalization of the EGFR in both transfection-dependent Chinese hamster ovary cells and transfection-independent A549 lung epithelial cells. These findings only begin to explain the features seen under oxidative stress, but they yield a greater understanding of the role of c-Cbl in EGFR trafficking.


Subject(s)
Endosomes/metabolism , ErbB Receptors/metabolism , Proto-Oncogene Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Blotting, Western , CHO Cells , Cell Line, Tumor , Cricetinae , Down-Regulation , Humans , Hydrogen Peroxide/chemistry , Microscopy, Fluorescence , Models, Biological , Mutation , Oxidative Stress , Phenylalanine/chemistry , Phosphoprotein Phosphatases/metabolism , Precipitin Tests , Protein Transport , Proto-Oncogene Proteins c-cbl , RNA Processing, Post-Transcriptional , Signal Transduction , Transfection , Tyrosine/chemistry , Ubiquitin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...