Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
J Vasc Res ; 55(2): 98-110, 2018.
Article in English | MEDLINE | ID: mdl-29455203

ABSTRACT

BACKGROUND: Mitochondrial reactive oxygen species (ROS) contribute to inflammation and vascular remodeling during atherosclerotic plaque formation. C57BL/6N (6N) and C57BL/6J (6J) mice display distinct mitochondrial redox balance due to the absence of nicotinamide nucleotide transhydrogenase (NNT) in 6J mice. We hypothesize that differential NNT expression between these animals alters plaque development. METHODS: 6N and 6J mice were treated with AAV8-PCSK9 (adeno-associated virus serotype 8/proprotein convertase subtilisin/kexin type 9) virus leading to hypercholesterolemia, increased low-density lipoprotein, and atherosclerosis in mice fed a high-fat diet (HFD). Mice were co-treated with the mitochondria-targeted superoxide dismutase mimetic MitoTEMPO to assess the contribution of mitochondrial ROS to atherosclerosis. RESULTS: Baseline and HFD-induced vascular superoxide is increased in 6J compared to 6N mice. MitoTEMPO diminished superoxide in both groups demonstrating differential production of mitochondrial ROS among these strains. PCSK9 treatment and HFD led to similar increases in plasma lipids in both 6N and 6J mice. However, 6J animals displayed significantly higher levels of plaque formation. MitoTEMPO reduced plasma lipids but did not affect plaque formation in 6N mice. In contrast, MitoTEMPO surprisingly increased plaque formation in 6J mice. CONCLUSION: These data indicate that loss of NNT increases vascular ROS production and exacerbates atherosclerotic plaque development.


Subject(s)
Aorta/enzymology , Aortic Diseases/enzymology , Atherosclerosis/enzymology , NADP Transhydrogenase, AB-Specific/deficiency , Animals , Antioxidants/pharmacology , Aorta/drug effects , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/pathology , Atherosclerosis/genetics , Atherosclerosis/pathology , Cholesterol/blood , Disease Models, Animal , Genetic Predisposition to Disease , Hypercholesterolemia/enzymology , Hypercholesterolemia/genetics , Male , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Proteins/deficiency , Mitochondrial Proteins/genetics , NADP Transhydrogenase, AB-Specific/genetics , Organophosphorus Compounds/pharmacology , Phenotype , Piperidines/pharmacology , Plaque, Atherosclerotic , Proprotein Convertase 9/genetics , Proprotein Convertase 9/metabolism , Superoxides/metabolism , Time Factors
2.
Redox Biol ; 70: 103085, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38359746

ABSTRACT

Endothelial dysfunction and endothelial activation are common early events in vascular diseases and can arise from mitochondrial dysfunction. Neurogranin (Ng) is a 17kD protein well known to regulate intracellular Ca2+-calmodulin (CaM) complex signaling, and its dysfunction is significantly implicated in brain aging and neurodegenerative diseases. We found that Ng is also expressed in human aortic endothelial cells (HAECs), and depleting Ng promotes Ca2+-CaM complex-dependent endothelial activation and redox imbalances. Endothelial-specific Ng knockout (Cre-CDH5-Ngf/f) mice demonstrate a significant delay in the flow-mediated dilation (FMD) response. Therefore, it is critical to characterize how endothelial Ng expression regulates reactive oxygen species (ROS) generation and affects cardiovascular disease. Label-free quantification proteomics identified that mitochondrial dysfunction and the oxidative phosphorylation pathway are significantly changed in the aorta of Cre-CDH5-Ngf/f mice. We found that a significant amount of Ng is expressed in the mitochondrial fraction of HAECs using western blotting and colocalized with the mitochondrial marker, COX IV, using immunofluorescence staining. Seahorse assay demonstrated that a lack of Ng decreases mitochondrial respiration. Treatment with MitoEbselen significantly restores the oxygen consumption rate in Ng knockdown cells. With the RoGFP-Orp1 approach, we identified that Ng knockdown increases mitochondrial-specific hydrogen peroxide (H2O2) production, and MitoEbselen treatment significantly reduced mitochondrial ROS (mtROS) levels in Ng knockdown cells. These results suggest that Ng plays a significant role in mtROS production. We discovered that MitoEbselen treatment also rescues decreased eNOS expression and nitric oxide (NO) levels in Ng knockdown cells, which implicates the critical role of Ng in mtROS-NO balance in the endothelial cells.


Subject(s)
Endothelial Cells , Mitochondria , Neurogranin , Animals , Humans , Mice , Endothelial Cells/metabolism , Hydrogen Peroxide/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Diseases/metabolism , Neurogranin/metabolism , Nitric Oxide/metabolism , Oxidation-Reduction , Reactive Oxygen Species/metabolism
3.
J Biol Chem ; 285(21): 16239-47, 2010 May 21.
Article in English | MEDLINE | ID: mdl-20228065

ABSTRACT

Transforming growth factor beta (TGF-beta) stimulates reactive oxygen species (ROS) production in various cell types, which mediates many of the effects of TGF-beta. The molecular mechanisms whereby TGF-beta increases ROS production and ROS modulate the signaling processes of TGF-beta, however, remain poorly defined. In this study, we show that TGF-beta1 stimulates NADPH oxidase 4 (Nox4) expression and ROS generation in the nucleus of murine embryo fibroblasts (NIH3T3 cells). This is associated with an increase in protein thiol modification and inactivation of MAPK phosphatase 1 (MKP-1), a nuclear phosphatase. Furthermore, knockdown of MKP-1 using small interfering RNA enhances TGF-beta1-induced phosphorylation of JNK and p38 as well as the expression of plasminogen activator inhibitor 1 (PAI-1), a TGF-beta-responsive gene involved in the pathogenesis of many diseases. Knockdown of Nox4 with Nox4 small interfering RNA, on the other hand, reduces TGF-beta1-stimulated ROS production, p38 phosphorylation, and PAI-1 expression. TGF-beta also increased the nuclear level of Nox4 protein as well as PAI-1 expression in human lung fibroblasts (CCL-210 cells), suggesting that TGF-beta may induce PAI-1 expression by a similar mechanism in human lung fibroblasts. In summary, in this study we have identified nuclear MAPK phosphatase MKP-1 as a novel molecular target of ROS in TGF-beta signaling pathways. Our data suggest that increased generation of ROS by Nox4 mediates TGF-beta1-induced PAI-1 gene expression at least in part through oxidative modification and inhibition of MKP-1 leading to a sustained activation of JNK and p38 MAPKs.


Subject(s)
Dual Specificity Phosphatase 1/metabolism , Fibroblasts/metabolism , Gene Expression Regulation/physiology , MAP Kinase Signaling System/physiology , Nuclear Proteins/metabolism , Plasminogen Activator Inhibitor 1/biosynthesis , Transforming Growth Factor beta1/metabolism , Animals , Dual Specificity Phosphatase 1/genetics , Fibroblasts/cytology , Gene Expression Regulation/drug effects , Humans , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/metabolism , MAP Kinase Signaling System/drug effects , Mice , NADPH Oxidase 4 , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , NIH 3T3 Cells , Nuclear Proteins/genetics , Oxidation-Reduction/drug effects , Phosphorylation/drug effects , Phosphorylation/physiology , Plasminogen Activator Inhibitor 1/genetics , RNA, Small Interfering/genetics , Reactive Oxygen Species/metabolism , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/pharmacology , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
4.
Lab Invest ; 91(8): 1122-35, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21647091

ABSTRACT

While there is general agreement that cardiovascular disease (CVD) development is influenced by a combination of genetic, environmental, and behavioral contributors, the actual mechanistic basis of how these factors initiate or promote CVD development in some individuals while others with identical risk profiles do not, is not clearly understood. This review considers the potential role for mitochondrial genetics and function in determining CVD susceptibility from the standpoint that the original features that molded cellular function were based upon mitochondrial-nuclear relationships established millions of years ago and were likely refined during prehistoric environmental selection events that today, are largely absent. Consequently, contemporary risk factors that influence our susceptibility to a variety of age-related diseases, including CVD were probably not part of the dynamics that defined the processes of mitochondrial-nuclear interaction, and thus, cell function. In this regard, the selective conditions that contributed to cellular functionality and evolution should be given more consideration when interpreting and designing experimental data and strategies. Finally, future studies that probe beyond epidemiologic associations are required. These studies will serve as the initial steps for addressing the provocative concept that contemporary human disease susceptibility is the result of selection events for mitochondrial function that increased chances for prehistoric human survival and reproductive success.


Subject(s)
Cardiovascular Diseases/etiology , DNA, Mitochondrial/genetics , Mitochondria/physiology , Biological Evolution , Diet/adverse effects , Disease Susceptibility , Humans , Oxidation-Reduction , Selection, Genetic
5.
Redox Biol ; 36: 101650, 2020 09.
Article in English | MEDLINE | ID: mdl-32763515

ABSTRACT

Endothelial dysfunction is a critical, initiating step in the development of hypertension (HTN) and mitochondrial reactive oxygen species (ROS) are important contributors to endothelial dysfunction. Genome-wide association studies (GWAS) have identified single nucleotide polymorphisms (SNPs) in the nicotinamide nucleotide transhydrogenase (Nnt) gene that are associated with endothelial dysfunction and increased risk for HTN. NNT is emerging as an important enzyme that regulates mitochondrial NADPH levels and mitochondrial redox balance by supporting the thiol dependent peroxidase systems in the mitochondria. We have previously shown that the absence of NNT in C57Bl/6J animals promotes a more severe hypertensive phenotype through reductions in •NO and endothelial dependent vessel dilation. However, the impact of NNT on human endothelial cell function remains unclear. We utilized NNT directed shRNA in human aortic endothelial cells to test the hypothesis that NNT critically regulates mitochondrial redox balance and endothelial function in response to angiotensin II (Ang II). We demonstrate that NNT expression and activity are elevated in response to the mitochondrial dysfunction and oxidative stress associated with Ang II treatment. Knockdown of NNT led to a significant elevation of mitochondrial ROS production and impaired glutathione peroxidase and glutathione reductase activities associated with a reduction in the NADPH/NADP+ ratio. Loss of NNT also promoted mitochondrial dysfunction, disruption of the mitochondrial membrane potential, and impaired ATP production in response to Ang II. Finally, we observed that, while the loss of NNT augmented eNOS phosphorylation at Ser1177, neither eNOS activity nor nitric oxide production were similarly increased. The results from these studies clearly demonstrate that NNT is critical for the maintenance of mitochondrial redox balance and mitochondrial function. Loss of NNT and disruption of redox balance leads to oxidative stress that compromises eNOS activity that could have a profound effect on the endothelium dependent regulation of vascular tone.


Subject(s)
NADP Transhydrogenases , Angiotensin II/metabolism , Animals , Endothelial Cells/metabolism , Genome-Wide Association Study , Humans , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , NADP Transhydrogenase, AB-Specific/genetics , NADP Transhydrogenases/metabolism , Oxidative Stress , Reactive Oxygen Species/metabolism
6.
Immunohorizons ; 4(10): 659-669, 2020 10 19.
Article in English | MEDLINE | ID: mdl-33077427

ABSTRACT

Macrophages reprogram their metabolism to promote appropriate responses. Proresolving macrophages primarily use fatty acid oxidation as an energy source. Metabolites generated during the catabolism of fatty acids aid in the resolution of inflammation and tissue repair, but the regulatory mechanisms that control lipid metabolism in macrophages are not fully elucidated. Lipin-1, a phosphatidic acid phosphatase that has transcriptional coregulator activity, regulates lipid metabolism in a variety of cells. In this current study, we show that lipin-1 is required for increased oxidative phosphorylation in IL-4 stimulated mouse (Mus musculus) macrophages. We also show that the transcriptional coregulatory function of lipin-1 is required for ß-oxidation in response to palmitate (free fatty acid) and apoptotic cell (human) stimulation. Mouse bone marrow-derived macrophages lacking lipin-1 have a reduction in critical TCA cycle metabolites following IL-4 stimulation, suggesting a break in the TCA cycle that is supportive of lipid synthesis rather than lipid catabolism. Together, our data demonstrate that lipin-1 regulates cellular metabolism in macrophages in response to proresolving stimuli and highlights the importance of aligning macrophage metabolism with macrophage phenotype.


Subject(s)
Cell Polarity/genetics , Interleukin-4/metabolism , Macrophage Activation , Macrophages/immunology , Phosphatidate Phosphatase/metabolism , Animals , Cell Polarity/immunology , Cells, Cultured , Gene Expression , Gene Knockout Techniques , Inflammation/genetics , Inflammation/immunology , Interleukin-4/genetics , Macrophages/enzymology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphatidate Phosphatase/genetics , Wound Healing/genetics , Wound Healing/immunology
7.
Article in English | MEDLINE | ID: mdl-32663656

ABSTRACT

Cardiac arrest causes neuronal damage and functional impairments that can result in learning/memory dysfunction after ischemia. We previously identified a saturated fatty acid (stearic acid methyl ester, SAME) that was released from the superior cervical ganglion (sympathetic ganglion). The function of stearic acid methyl ester is currently unknown. Here, we show that SAME can inhibit the detrimental effects of global cerebral ischemia (i.e. cardiac arrest). Treatment with SAME in the presence of asphyxial cardiac arrest (ACA) revived learning and working memory deficits. Similarly, SAME-treated hippocampal slices after oxygen-glucose deprivation inhibited neuronal cell death. Moreover, SAME afforded neuroprotection against ACA in the CA1 region of the hippocampus, reduced ionized calcium-binding adapter molecule 1 expression and inflammatory cytokines/chemokines, with restoration in mitochondria respiration. Altogether, we describe a unique and uncharted role of saturated fatty acids in the brain that may have important implications against cerebral ischemia.


Subject(s)
Asphyxia/drug therapy , CA1 Region, Hippocampal/metabolism , Heart Arrest/drug therapy , Neuroprotection/drug effects , Stearic Acids/pharmacology , Animals , Asphyxia/metabolism , Asphyxia/physiopathology , CA1 Region, Hippocampal/physiopathology , Disease Models, Animal , Heart Arrest/metabolism , Heart Arrest/physiopathology , Male , Rats , Rats, Sprague-Dawley
8.
Am J Physiol Lung Cell Mol Physiol ; 297(2): L209-16, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19395667

ABSTRACT

More than 100 million people in the United States live in areas that exceed current ozone air quality standards. In addition to its known pulmonary effects, environmental ozone exposures have been associated with increased hospital admissions related to cardiovascular events, but to date, no studies have elucidated the potential molecular mechanisms that may account for exposure-related vascular impacts. Because of the known pulmonary redox and immune biology stemming from ozone exposure, we hypothesized that ozone inhalation would initiate oxidant stress, mitochondrial damage, and dysfunction within the vasculature. Accordingly, these factors were quantified in mice consequent to a cyclic, intermittent pattern of ozone or filtered air control exposure. Ozone significantly modulated vascular tone regulation and increased oxidant stress and mitochondrial DNA damage (mtDNA), which was accompanied by significantly decreased vascular endothelial nitric oxide synthase protein and indices of nitric oxide production. To examine influences on atherosclerotic lesion formation, apoE-/- mice were exposed as above, and aortic plaques were quantified. Exposure resulted in significantly increased atherogenesis compared with filtered air controls. Vascular mitochondrial damage was additionally quantified in ozone- and filtered air-exposed infant macaque monkeys. These studies revealed that ozone increased vascular mtDNA damage in nonhuman primates in a fashion consistent with known atherosclerotic lesion susceptibility in humans. Consequently, inhaled ozone, in the absence of other environmental toxicants, promotes increased vascular dysfunction, oxidative stress, mitochondrial damage, and atherogenesis.


Subject(s)
Air Pollutants/adverse effects , Atherosclerosis/etiology , Mitochondrial Diseases/etiology , Ozone/adverse effects , Animals , Aorta/metabolism , Atherosclerosis/metabolism , Blood Pressure/physiology , DNA Damage/physiology , DNA, Mitochondrial/genetics , Heart Rate/physiology , Lung Diseases/etiology , Lung Diseases/metabolism , Macaca mulatta , Male , Mice , Mice, Inbred C57BL , Mitochondria/physiology , Mitochondrial Diseases/metabolism , Nitrates/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/metabolism , Nitrites/metabolism , Oxidants/adverse effects , Oxidative Stress/physiology , Superoxide Dismutase/metabolism , Tyrosine/analogs & derivatives , Tyrosine/metabolism
9.
Cardiovasc Toxicol ; 19(6): 500-509, 2019 12.
Article in English | MEDLINE | ID: mdl-31020509

ABSTRACT

Human immunodeficiency virus (HIV)-infected patients undergoing antiretroviral therapy are afforded an increased lifespan but also exhibit an elevated incidence of cardiovascular disease. HIV therapy uses a combination drug approach, and nucleoside reverse transcriptase inhibitors (NRTI) are a backbone of this therapy. Endothelial dysfunction is an initiating event in cardiovascular disease etiology, and in our prior studies, NRTIs induced an endothelial dysfunction that was dependent upon mitochondrial oxidative stress. Moreover, short-term NRTI administration induced a mitophagy-associated endothelial toxicity and increased reactive oxygen species (ROS) production that was rescued by coenzyme Q10 (Q10) or overexpression of a mitochondrial antioxidant enzyme. Thus, our objective was to examine mitochondrial toxicity in endothelial cells after chronic NRTI treatment and evaluate Q10 as a potential adjunct therapy for preventing NRTI-induced mitochondrial toxicity. Human aortic endothelial cells (HAEC) were exposed to chronic NRTI treatment, with or without Q10. ROS production, cell proliferation rate, levels of senescence, and mitochondrial bioenergetic function were determined. Chronic NRTI increased ROS production but decreased population doubling. In addition, NRTI increased the accumulation of ß-galactosidase, indicative of an accelerated rate of senescence. Moreover, ATP-linked respiration was diminished. Co-treatment with Q10 delayed the onset of NRTI-induced senescence, decreased ROS production and rescued the cells' mitochondrial respiration rate. Thus, our findings may suggest antioxidant enrichment approaches for reducing the cardiovascular side effects of NRTI therapy.


Subject(s)
Antioxidants/pharmacology , Cellular Senescence/drug effects , Endothelial Cells/drug effects , Mitochondria/drug effects , Reverse Transcriptase Inhibitors/toxicity , Ubiquinone/analogs & derivatives , Cell Proliferation , Cells, Cultured , Cytoprotection , Endothelial Cells/metabolism , Endothelial Cells/pathology , Energy Metabolism/drug effects , Humans , Mitochondria/metabolism , Mitochondria/pathology , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Signal Transduction , Ubiquinone/pharmacology
10.
J Am Soc Hypertens ; 11(2): 110-121, 2017 02.
Article in English | MEDLINE | ID: mdl-28087333

ABSTRACT

Oxidant stress contributes to the initiation and progression of hypertension (HTN) by enhancing endothelial dysfunction and/or causing perturbations in nitric oxide homeostasis. Differences in mitochondrial function may augment this process and provide insight into why age of onset and clinical outcomes differ among individuals from distinct ethnic groups. We have previously demonstrated that variation in normal mitochondrial function and oxidant production exists in endothelial cells from individuals of Caucasian and African-American ethnicity and that this variation contributes to endothelial dysfunction. To model these distinct mitochondrial redox phenotypes, we used C57Bl/6N (6N) and C57Bl/6J (6J) mice that also display unique mitochondrial functional properties due to the differential expression nicotinamide nucleotide transhydrogenase (NNT). We demonstrate that the absence of NNT in 6J cells led to distinct mitochondrial bioenergetic profiles and a pro-oxidative mitochondrial phenotype characterized by increased superoxide production and reduced glutathione peroxidase activity. Interestingly, we found that 6J animals have significantly higher systolic blood pressure compared to 6N animals, and this difference is exacerbated by angiotensin II treatment. The changes in pressure were accompanied by both mitochondrial and vascular dysfunction revealed by impaired respiratory control ratios and endothelial-dependent vessel dilation. All end points could be significantly ameliorated by treatment with the mitochondria-targeted superoxide dismutase mimetic MitoTEMPO demonstrating a critical role for the production of mitochondrial reactive oxygen species in the development of HTN in these animals. Taken together, these data indicate that the absence of NNT leads to variation in mitochondrial function and contributes to a unique mitochondrial redox phenotype that influences susceptibility to HTN by contributing to endothelial and vascular dysfunction.


Subject(s)
Endothelium/physiopathology , Hypertension/pathology , Mitochondria/metabolism , NADP Transhydrogenase, AB-Specific/metabolism , Oxidative Stress , Superoxides/metabolism , Angiotensin II/pharmacology , Animals , Antioxidants/pharmacology , Blood Pressure/drug effects , Carotid Arteries , Cells, Cultured , Disease Models, Animal , Humans , Male , Mice , Mice, Inbred C57BL , Mitochondrial Proteins/metabolism , Myography , Nitric Oxide/metabolism , Organophosphorus Compounds/pharmacology , Oxidation-Reduction , Piperidines/pharmacology , Primary Cell Culture , Superoxide Dismutase/metabolism , Vasoconstrictor Agents/pharmacology
11.
Circ Cardiovasc Genet ; 9(1): 26-36, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26787433

ABSTRACT

BACKGROUND: We hypothesized that endothelial cells having distinct mitochondrial genetic backgrounds would show variation in mitochondrial function and oxidative stress markers concordant with known differential cardiovascular disease susceptibilities. To test this hypothesis, mitochondrial bioenergetics were determined in endothelial cells from healthy individuals with African versus European maternal ancestries. METHODS AND RESULTS: Bioenergetics and mitochondrial DNA (mtDNA) damage were assessed in single-donor human umbilical vein endothelial cells belonging to mtDNA haplogroups H and L, representing West Eurasian and African maternal ancestries, respectively. Human umbilical vein endothelial cells from haplogroup L used less oxygen for ATP production and had increased levels of mtDNA damage compared with those in haplogroup H. Differences in bioenergetic capacity were also observed in that human umbilical vein endothelial cells belonging to haplogroup L had decreased maximal bioenergetic capacities compared with haplogroup H. Analysis of peripheral blood mononuclear cells from age-matched healthy controls with West Eurasian or African maternal ancestries showed that haplogroups sharing an A to G mtDNA mutation at nucleotide pair 10398 had increased mtDNA damage compared with those lacking this mutation. Further study of angiographically proven patients with coronary artery disease and age-matched healthy controls revealed that mtDNA damage was associated with vascular function and remodeling and that age of disease onset was later in individuals from haplogroups lacking the A to G mutation at nucleotide pair 10398. CONCLUSIONS: Differences in mitochondrial bioenergetics and mtDNA damage associated with maternal ancestry may contribute to endothelial dysfunction and vascular disease.


Subject(s)
Black People/genetics , DNA Damage , DNA, Mitochondrial , Energy Metabolism/genetics , Haplotypes , Human Umbilical Vein Endothelial Cells/metabolism , White People/genetics , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Female , Humans , Male , Mutation , Oxidative Stress/genetics
12.
Free Radic Biol Med ; 33(7): 974, 2002 Oct 01.
Article in English | MEDLINE | ID: mdl-12361807

ABSTRACT

Glutathione is the most abundant non-protein thiol in the cell, with roles in cell cycle regulation, detoxification of xenobiotics, and maintaining the redox tone of the cell. The glutathione content is controlled at several levels, the most important being the rate of de novo synthesis, which is mediated by two enzymes, glutamate cysteine ligase (GCL), and glutathione synthetase (GS), with GCL being rate-limiting generally. The GCL holoenzyme consists of a catalytic (GCLC) and a modulatory (GCLM) subunit, which are encoded by separate genes. In the present study, the signaling mechanisms leading to de novo synthesis of GSH in response to physiologically relevant concentrations of 4-hydroxy-2-nonenal (4HNE), an endproduct of lipid peroxidation, were investigated. We demonstrated that exposure to 4HNE resulted in increased content of both Gcl mRNAs, both GCL subunits, phosphorylated JNK1 and c-Jun proteins, as well as Gcl TRE sequence-specific AP-1 binding activity. These increases were attenuated by pretreating the cells with a novel membrane-permeable JNK pathway inhibitor, while chemical inhibitors of the p38 or ERK pathways were ineffective. These data reveal that de novo GSH biosynthesis in response to 4HNE signals through the JNK pathway and suggests a major role for AP-1 driven expression of both Gcl genes in HBE1 cells.


Subject(s)
Aldehydes/pharmacology , Glutamate-Cysteine Ligase/genetics , Mitogen-Activated Protein Kinases/metabolism , Cell Line , Enzyme Induction/drug effects , Glutamate-Cysteine Ligase/biosynthesis , Glutathione/metabolism , Growth Inhibitors/pharmacology , Humans , JNK Mitogen-Activated Protein Kinases , Kinetics , Phosphorylation , Proto-Oncogene Proteins c-jun/metabolism
13.
Free Radic Biol Med ; 53(4): 951-61, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22705369

ABSTRACT

Tissues are exposed to exogenous and endogenous nitrogen dioxide ((·)NO(2)), which is the terminal agent in protein tyrosine nitration. Besides iron chelation, the hydroxamic acid (HA) desferrioxamine (DFO) shows multiple functionalities including nitration inhibition. To investigate mechanisms whereby DFO affects 3-nitrotyrosine (3-NT) formation, we utilized gas-phase (·)NO(2) exposures, to limit introduction of other reactive species, and a lung surface model wherein red cell membranes (RCM) were immobilized under a defined aqueous film. When RCM were exposed to ()NO(2) covered by +/- DFO: (i) DFO inhibited 3-NT formation more effectively than other HA and non-HA chelators; (ii) 3-NT inhibition occurred at very low[DFO] for prolonged times; and (iii) 3-NT formation was iron independent but inhibition required DFO present. DFO poorly reacted with (·)NO(2) compared to ascorbate, assessed via (·)NO(2) reactive absorption and aqueous-phase oxidation rates, yet limited 3-NT formation at far lower concentrations. DFO also inhibited nitration under aqueous bulk-phase conditions, and inhibited 3-NT generated by active myeloperoxidase "bound" to RCM. Per the above and kinetic analyses suggesting preferential DFO versus (·)NO(2) reaction within membranes, we conclude that DFO inhibits 3-NT formation predominantly by facile repair of the tyrosyl radical intermediate, which prevents (·)NO(2) addition, and thus nitration, and potentially influences biochemical functionalities.


Subject(s)
Deferoxamine/pharmacology , Free Radical Scavengers/pharmacology , Proteins/metabolism , Tyrosine/analogs & derivatives , Cell Membrane/drug effects , Cell Membrane/enzymology , Cell Membrane/metabolism , Deferoxamine/chemistry , Erythrocytes/drug effects , Erythrocytes/enzymology , Erythrocytes/metabolism , Free Radical Scavengers/chemistry , Humans , Lung/drug effects , Lung/physiology , Nitrogen Dioxide/chemistry , Nitrogen Dioxide/pharmacology , Oxidants, Photochemical/chemistry , Oxidants, Photochemical/pharmacology , Peroxidase/metabolism , Proteins/chemistry , Siderophores/chemistry , Siderophores/pharmacology , Tyrosine/chemistry , Tyrosine/metabolism
14.
Arch Biochem Biophys ; 423(1): 116-25, 2004 Mar 01.
Article in English | MEDLINE | ID: mdl-14871475

ABSTRACT

Glutamate cysteine ligase (GCL), composed of a catalytic (GCLC) and modulatory (GCLM) subunit, catalyzes the first step of glutathione (GSH) biosynthesis. Using 4-hydroxy-2-nonenal (4HNE), 2,3-dimethoxy-1,4-naphthoquinone (DMNQ), and tertiary-butylhydroquinone (tBHQ) as models of oxidative stress which are known to work through different mechanisms, we measured changes in cellular GSH, GCL mRNA, and GCL protein. 4HNE and tBHQ treatments increased cellular GSH levels, while DMNQ exposure depleted GSH. Furthermore, changes in the two GCL mRNAs largely paralleled changes in the GCL proteins; however, the magnitudes differed, suggesting some form of translational control. The molar ratio of GCLC:GCLM ranged from 3:1 to 17:1 in control human bronchial epithelial (HBE1) cells and all treatments further increased this ratio. Data from several mouse tissues show molar ratios of GCLC:GCLM that range from 1:1 to 10:1 in support of these findings. These data demonstrate that alterations in cellular GSH are clearly correlated with GCLC to a greater extent than GCLM. Surprisingly, both control HBE1 cells and some mouse tissues have more GCLC than GCLM and GCLM increases to a much lesser extent than GCLC, suggesting that the regulatory role of GCLM is minimal under physiologically relevant conditions of oxidative stress.


Subject(s)
Glutamate-Cysteine Ligase/metabolism , Glutathione/biosynthesis , Oxidative Stress/physiology , Aldehydes/pharmacology , Antioxidants/pharmacology , Bronchi/drug effects , Bronchi/enzymology , Bronchi/metabolism , Epithelium/drug effects , Epithelium/enzymology , Epithelium/metabolism , Glutamate-Cysteine Ligase/drug effects , Glutathione/drug effects , Humans , Hydroquinones/pharmacology , Naphthoquinones/pharmacology , Oxidative Stress/drug effects
15.
Am J Physiol Lung Cell Mol Physiol ; 283(4): L726-36, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12225949

ABSTRACT

In A549 cell culture, significant variability was found in sensitivity to actinomycin D. Using limiting dilution, actinomycin D-susceptible (G4S) and -resistant (D3R) subclones were isolated. G4S cells were also susceptible to protein synthesis inhibitors, a redox cycling quinone, and an electrophile with concomitant activation of caspases 3 and 9. D3R cells were resistant to these agents without caspase activation. Antioxidant profiles revealed that D3R cells had significantly higher glutathione and glutathione reductase activity but markedly lower catalase, glutathione peroxidase, and aldehyde reductase activities than G4S cells. Thus A549 cells contain at least two distinct subpopulations with respect to predisposition to cell death and antioxidant profile. Because sensitivities to agents and the antioxidant profile were inconsistent, mechanisms independent of antioxidants, including the apparent inability to activate caspases in D3R cells, may play an important role. Regardless, the results suggest that antioxidant profiles of asymmetrical cell populations cannot predict sensitivity to oxidants and warn that the use of single subclones is advisable for mechanistic studies using A549 or other unstable cell lines.


Subject(s)
Adenocarcinoma , Antioxidants/metabolism , Lung Neoplasms , Oxidative Stress/physiology , Aldehydes/pharmacology , Anisomycin/pharmacology , Apoptosis/physiology , Caspases/metabolism , Cytotoxins/pharmacology , Dactinomycin/pharmacology , Genetic Heterogeneity , Glutathione/metabolism , Growth Inhibitors/pharmacology , Humans , Hydrogen Peroxide/metabolism , Male , Naphthoquinones/pharmacology , Oxidation-Reduction , Protein Synthesis Inhibitors/pharmacology , Superoxide Dismutase/metabolism , Tumor Cells, Cultured/cytology , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL